Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Genes (Basel) ; 15(6)2024 May 27.
Article in English | MEDLINE | ID: mdl-38927636

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia/lymphoma. The oncogene product Tax of HTLV-I is thought to play crucial roles in leukemogenesis by promoting proliferation of the virus-infected cells through activation of growth-promoting genes. These genes code for growth factors and their receptors, cytokines, cell adhesion molecules, growth signal transducers, transcription factors and cell cycle regulators. We show here that Tax activates the gene coding for coactivator-associated arginine methyltransferase 1 (CARM1), which epigenetically enhances gene expression through methylation of histones. Tax activated the Carm1 gene and increased protein expression, not only in human T-cell lines but also in normal peripheral blood lymphocytes (PHA-PBLs). Tax increased R17-methylated histone H3 on the target gene IL-2Rα, concomitant with increased expression of CARM1. Short hairpin RNA (shRNA)-mediated knockdown of CARM1 decreased Tax-mediated induction of IL-2Rα and Cyclin D2 gene expression, reduced E2F activation and inhibited cell cycle progression. Tax acted via response elements in intron 1 of the Carm1 gene, through the NF-κB pathway. These results suggest that Tax-mediated activation of the Carm1 gene contributes to leukemogenic target-gene expression and cell cycle progression, identifying the first epigenetic target gene for Tax-mediated trans-activation in cell growth promotion.


Subject(s)
Gene Products, tax , Human T-lymphotropic virus 1 , Protein-Arginine N-Methyltransferases , Humans , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Gene Products, tax/genetics , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/genetics , Cyclin D2/genetics , Cyclin D2/metabolism , Transcriptional Activation , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , NF-kappa B/metabolism , NF-kappa B/genetics , Histones/metabolism , Histones/genetics , Epigenesis, Genetic , Jurkat Cells
2.
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.

3.
Int J Nanomedicine ; 18: 5343-5363, 2023.
Article in English | MEDLINE | ID: mdl-37746048

ABSTRACT

Objective: This study aimed to deliver a polypeptide from the Bax-BH3 domain (BHP) through the synthesis of self-assembled amphiphile nanovectors (NVs) and to assess their potential for cancer therapeutic applications and biological safety in vitro and in vivo. These findings provide valuable options for cancer intervention and a novel approach for the rational design of therapeutics. Methods: We studied the antitumor activity of BHP by preparing RGDfK-PHPMA-b-Poly (MMA-alt-(Rhob-MA)) (RPPMMRA) and encapsulating it in BHP-NV. We also performed a series of characterizations and property analyses of RPPMMRA, including its size, stability, and drug-carrying capacity. The biocompatibility of RPPMMRA was evaluated in terms of cytotoxicity and hemolytic effects. The pro-apoptotic capacity of BHP was evaluated in vitro using mitochondrial membrane potential, flow cytometry, and apoptosis visualization techniques. The potential therapeutic effects of BHP on tumors were explored using reverse molecular docking. We also investigated the in vivo proapoptotic effect of BHP-NV in a nude mouse tumor model. Results: NVs were successfully prepared with hydrated particle sizes ranging from 189.6 nm to 256.6 nm, spherical overall, and were able to remain stable in different media for 72 h with drug loading up to 15.2%. The NVs were be successfully internalized within 6 h with good biocompatibility. Neither BHP nor NV showed significant toxicity when administered alone, however, BHP-NV demonstrated significant side effects in vitro and in vivo. The apoptosis rate increased significantly from 14.13% to 66.34%. Experiments in vivo showed that BHP-NV exhibited significant apoptotic and tumor-suppressive effects. Conclusion: A targeted fluorescent NV with high drug delivery efficiency and sustained release protected the active center of BHP, constituting BHP-NV for targeted delivery. RPPMMRA demonstrated excellent biocompatibility, stability, and drug loading ability, whereas and BHP-NV demonstrated potent antitumor effects in vivo and in vitro.


Subject(s)
Drug Delivery Systems , Peptides , Animals , Mice , Molecular Docking Simulation , Apoptosis , Coloring Agents , Mice, Nude
4.
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
5.
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
6.
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
7.
J Biomed Opt ; 23(9): 1-7, 2018 09.
Article in English | MEDLINE | ID: mdl-30182581

ABSTRACT

Virus infection of a human cell was determined only 3 h after invagination. We used viral vector Ad-CMV-control (AdC), which lacks the E1 gene coding for early polypeptide 1 (E1). AdC can replicate in human embryonic kidney 293 (HEK293) cells into which the E1 gene has been transfected. According to partial least-square regression discriminant analysis, it was assumed that two kinds of reaction take place in the cell during viral invasion. The first response of the cell was determined 3 h after the virus invasion, and the second one was determined ∼9 h later. The first one seems to be due to compositional changes in DNA. Analysis of large-scale datasets strongly indicated that the second reaction can be attributed to a reduction in protein concentration or uptake of phenylalanine into the nucleus.


Subject(s)
Single-Cell Analysis/methods , Spectrum Analysis, Raman/methods , Virology/methods , Virus Diseases/virology , Adenovirus E1 Proteins/genetics , HEK293 Cells , Humans , Models, Biological
8.
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
9.
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
10.
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
11.
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
12.
Genes Cells ; 20(9): 739-57, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26201719

ABSTRACT

The transcription factor E2F is the principal target of the tumor suppressor pRB. E2F plays crucial roles not only in cell proliferation by activating growth-related genes but also in tumor suppression by activating pro-apoptotic and growth-suppressive genes. We previously reported that, in human normal fibroblasts, the tumor suppressor genes ARF, p27(Kip1) and TAp73 are activated by deregulated E2F activity induced by forced inactivation of pRB, but not by physiological E2F activity induced by growth stimulation. In contrast, growth-related E2F targets are activated by both E2F activities, underscoring the roles of deregulated E2F in tumor suppression in the context of dysfunctional pRB. In this study, to further understand the roles of deregulated E2F, we explored new targets that are specifically activated by deregulated E2F using DNA microarray. The analysis identified nine novel targets (BIM, RASSF1, PPP1R13B, JMY, MOAP1, RBM38, ABTB1, RBBP4 and RBBP7), many of which are involved in the p53 and RB tumor suppressor pathways. Among these genes, the BIM gene was shown to be activated via atypical E2F-responsive promoter elements and to contribute to E2F1-mediated apoptosis. Our results underscore crucial roles of deregulated E2F in growth suppression to counteract loss of pRB function.


Subject(s)
Apoptosis Regulatory Proteins/genetics , E2F Transcription Factors/metabolism , E2F1 Transcription Factor/metabolism , Fibroblasts/metabolism , Genes, Tumor Suppressor , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Apoptosis , Bcl-2-Like Protein 11 , Cell Line , Promoter Regions, Genetic
13.
J Biomed Opt ; 19(6): 067003, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24898605

ABSTRACT

The present study demonstrates that Raman spectroscopy is a powerful tool for the detection of virus-infected cells. Adenovirus infection of human embryonic kidney 293 cells was successfully detected at 12, 24, and 48 h after initiating the infection. The score plot of principal component analysis discriminated the spectra of the infected cells from those of the control cells. The viral infection was confirmed by the conventional immunostaining method performed 24 h after the infection. The newly developed method provides a fast and label-free means for the detection of virus-infected cells.


Subject(s)
Adenoviridae Infections/diagnosis , Adenoviridae , HEK293 Cells/virology , Spectrum Analysis, Raman/methods , Adenoviridae Infections/pathology , Humans , Principal Component Analysis , Reproducibility of Results , Signal Processing, Computer-Assisted , Time Factors
14.
Biochem Biophys Res Commun ; 450(1): 240-6, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24893334

ABSTRACT

In current cancer treatment protocols, such as radiation and chemotherapy, side effects on normal cells are major obstacles to radical therapy. To avoid these side effects, a cancer cell-specific approach is needed. One way to specifically target cancer cells is to utilize a cancer specific promoter to express a cytotoxic gene (suicide gene therapy) or a viral gene required for viral replication (oncolytic virotherapy). For this purpose, the selected promoter should have minimal activity in normal cells to avoid side effects, and high activity in a wide variety of cancers to obtain optimal therapeutic efficacy. In contrast to the AFP, CEA and PSA promoters, which have high activity only in a limited spectrum of tumors, the E2F1 promoter exhibits high activity in wide variety of cancers. This is based on the mechanism of carcinogenesis. Defects in the RB pathway and activation of the transcription factor E2F, the main target of the RB pathway, are observed in almost all cancers. Consequently, the E2F1 promoter, which is mainly regulated by E2F, has high activity in wide variety of cancers. However, E2F is also activated by growth stimulation in normal growing cells, suggesting that the E2F1 promoter may also be highly active in normal growing cells. In contrast, we found that the tumor suppressor ARF promoter is activated by deregulated E2F activity, induced by forced inactivation of pRB, but does not respond to physiological E2F activity induced by growth stimulation. We also found that the deregulated E2F activity, which activates the ARF promoter, is detected only in cancer cell lines. These observations suggest that ARF promoter is activated by E2F only in cancer cells and therefore may be more cancer cell-specific than E2F1 promoter to drive gene expression. We show here that the ARF promoter has lower activity in normal growing fibroblasts and shows higher cancer cell-specificity compared to the E2F1 promoter. We also demonstrate that adenovirus expressing HSV-TK under the control of the ARF promoter shows lower cytotoxicity than that of the E2F1 promoter, in normal growing fibroblasts but has equivalent cytotoxicity in cancer cell lines. These results suggest that the ARF promoter, which is specifically activated by deregulated E2F activity, is an excellent candidate to drive therapeutic cytotoxic gene expression, specifically in cancer cells.


Subject(s)
E2F1 Transcription Factor/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Promoter Regions, Genetic/genetics , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Proteins/genetics , Apoptosis/genetics , Cell Line, Tumor , Genes, Transgenic, Suicide/genetics , Genes, Tumor Suppressor , Genetic Therapy/methods , Humans
15.
Genes Cells ; 17(8): 660-72, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22702391

ABSTRACT

Discrimination of oncogenic growth signals from normal growth signals is crucial for tumor suppression. The transcription factor E2F, the main target of pRB, plays central role in cell proliferation by activating growth-promoting genes. E2F also plays an important role in tumor suppression by activating growth-suppressive genes such as pro-apoptotic genes. The regulatory mechanism of the latter genes is not known in detail, especially in response to normal and oncogenic growth signals. E2F is physiologically activated by growth stimulation through phosphorylation of pRB. In contrast, upon dysfunction of pRB, a major oncogenic change, E2F is activated out of control by pRB, generating deregulated E2F activity. We show here that the tumor suppressor TAp73 gene, which can induce apoptosis independently of p53, responds to deregulated E2F activity, but not to physiological E2F activity induced by growth stimulation in human normal fibroblasts. We identified E2F-responsive elements (ERE73s) in TAp73 promoter that can specifically sense deregulated E2F activity. Moreover, RB1-deficient cancer cell lines harbored deregulated E2F activity that activated ERE73s and the TAp73 gene, which were suppressed by re-introduction of pRB. These results underscore the important role of deregulated E2F in activation of the TAp73 gene, a component of major intrinsic tumor suppressor pathways.


Subject(s)
DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/metabolism , Fibroblasts/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Apoptosis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Culture Media/metabolism , DNA-Binding Proteins/genetics , E2F1 Transcription Factor/genetics , Etoposide/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Flow Cytometry , Gene Expression Regulation , Humans , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Point Mutation , Promoter Regions, Genetic , Protein Binding , Serum/metabolism , Transcriptional Activation , Transfection , Tumor Protein p73 , Tumor Suppressor Proteins/genetics
16.
Biochem Biophys Res Commun ; 417(3): 931-7, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22166212

ABSTRACT

The p53 tumor suppressor protein plays key roles in protecting cells from tumorigenesis. Phosphorylation of p53 at Ser46 (p53Ser46) is considered to be a crucial modification regulating p53-mediated apoptosis. Because the activity of p53 is impaired in most human cancers, restoration of wild-type p53 (wt-p53) function by its gene transfer or by p53-reactivating small molecules has been extensively investigated. The p53-reactivating compounds Nutlin-3 and RITA activate p53 in the absence of genotoxic stress by antagonizing the action of its negative regulator Mdm2. Although controversial, Nutlin-3 was shown to induce p53-mediated apoptosis in a manner independent of p53 phosphorylation. Recently, RITA was shown to induce apoptosis by promoting p53Ser46 phosphorylation. Here we examined whether Nutlin-3 or RITA can overcome resistance to p53-mediated apoptosis in p53-resistant tumor cell lines lacking the ability to phosphorylate p53Ser46. We show that Nutlin-3 did not rescue the apoptotic defect of a Ser46 phosphorylation-defective p53 mutant in p53-sensitive tumor cells, and that RITA neither restored p53Ser46 phosphorylation nor induced apoptosis in p53Ser46 phosphorylation-deficient cells retaining wt-p53. Furthermore, treatment with Nutlin-3 or RITA together with adenoviral p53 gene transfer also failed to induce apoptosis in p53Ser46 phosphorylation-deficient cells either expressing or lacking wt-p53. These results indicate that neither Nutlin-3 nor RITA in able to induce p53-mediated apoptosis in the absence of p53Ser46 phosphorylation. Thus, the dysregulation of this phosphorylation in tumor cells may be a critical factor that limits the efficacy of these p53-based cancer therapies.


Subject(s)
Apoptosis/drug effects , Furans/pharmacology , Imidazoles/pharmacology , Piperazines/pharmacology , Serine/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Caspase 3 , Cell Line, Tumor , Genes, p53 , Humans , Phosphorylation/drug effects , Phosphorylation/genetics , Poly(ADP-ribose) Polymerases/metabolism , Serine/genetics , Transduction, Genetic , Tumor Suppressor Protein p53/genetics
17.
Proc Natl Acad Sci U S A ; 107(50): 21529-34, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21098278

ABSTRACT

Retrovirus-mediated transduction of Hoxb4 enhances hematopoietic stem cell (HSC) activity and enforced expression of Hoxb4 induces in vitro development of HSCs from differentiating mouse embryonic stem cells, but the underlying molecular mechanism remains unclear. We previously showed that the HSC activity was abrogated by accumulated Geminin, an inhibitor for the DNA replication licensing factor Cdt1 in mice deficient in Rae28 (also known as Phc1), which encodes a member of Polycomb-group complex 1. In this study we found that Hoxb4 transduction reduced accumulated Geminin in Rae28-deficient mice, despite increasing the mRNA, and restored the impaired HSC activity. Supertransduction of Geminin suppressed the HSC activity induced by Hoxb4 transduction, whereas knockdown of Geminin promoted the clonogenic and replating activities, indicating the importance of Geminin regulation in the molecular mechanism underlying Hoxb4 transduction-mediated enhancement of the HSC activity. This facilitated our investigation of how transduced Hoxb4 reduced Geminin. We showed in vitro and in vivo that Hoxb4 and the Roc1 (also known as Rbx1)-Ddb1-Cul4a ubiquitin ligase core component formed a complex designated as RDCOXB4, which acted as an E3 ubiquitin ligase for Geminin and down-regulated Geminin through the ubiquitin-proteasome system. Down-regulated Geminin and the resultant E2F activation may provide cells with proliferation potential by increasing a DNA prereplicative complex loaded onto chromatin. Here we suggest that transduced Hoxb4 down-regulates Geminin protein probably by constituting the E3 ubiquitin ligase for Geminin to provide hematopoietic stem and progenitor cells with proliferation potential.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Cell Proliferation , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transduction, Genetic , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cullin Proteins/genetics , Cullin Proteins/metabolism , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Geminin , HEK293 Cells , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Minichromosome Maintenance Complex Component 2 , Multiprotein Complexes/metabolism , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Transcription Factors/genetics
18.
Genes Cells ; 14(1): 89-99, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19077036

ABSTRACT

The transcription factor E2F, the main target of the RB tumor suppressor pathway, plays crucial roles not only in cell proliferation but also in tumor suppression. The cyclin-dependent kinase inhibitor p27(Kip1) gene, an upstream negative regulator of E2F, is induced by ectopically expressed E2F1 but not by normal growth stimulation that physiologically activates endogenous E2F. This suggests that the gene can discriminate between deregulated and physiological E2F activity. To address this issue, we examined regulation of the p27(Kip1) gene by E2F. Here we show that p27(Kip1) promoter specifically senses deregulated E2F activity through elements similar to typical E2F sites. This E2F-like elements were activated by deregulated E2F activity induced by forced inactivation of pRb but not by physiological E2F activity induced by serum stimulation, contrary to typical E2F sites activated by both E2F activity. The endogenous p27(Kip1) gene responded to deregulated and physiological E2F activity in the same manner to the E2F-like elements. Moreover, the E2F-like elements bound ectopically expressed E2F1 but not physiologically activated E2F1 or E2F4 in vivo. These results suggest that the p27(Kip1) gene specifically senses deregulated E2F activity through the E2F-like elements to suppress inappropriate cell cycle progression in response to loss of pRb function.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , E2F1 Transcription Factor/genetics , Gene Expression Regulation , Promoter Regions, Genetic/genetics , Cell Line, Tumor , E2F4 Transcription Factor/metabolism , Humans , Protein Binding , Response Elements/genetics , Retinoblastoma Protein/metabolism , Serum/metabolism
20.
Cancer Sci ; 99(6): 1155-63, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18422743

ABSTRACT

The viral product Tax encoded by human T-cell leukemia virus type I (HTLV-I) is thought to play a central role in leukemogenesis. Clonal expansion of HTLV-I-infected cells requires the extension of cell division with telomere maintenance, which is regulated by the ribonucleoprotein enzyme telomerase. However, the roles of Tax in the expression of telomerase activity in T-cells remains controversial. Our previous study indicated that expression of the human telomerase reverse transcriptase subunit (hTERT) gene, which determines telomerase activity, is tightly regulated in human T-cells. In the present study, we investigated Tax-mediated regulation of hTERT gene expression by Tax in human T-cells. HTLV-I Tax induced expression of the hTERT gene in human peripheral blood leukocytes. Reporter assays revealed that Tax activated the hTERT promoter in quiescent Kit 225 cells, while the promoter activity was repressed by Tax in proliferating Jurkat cells. Both up-regulation and down-regulation by Tax were mediated through the 43-bp sequences in the promoter, which carried at least two elements that independently functioned as repressors. The two elements bound distinct factors. G1 to S phase transition induced by introduction of either cyclin D2 with cdk4 or p130-specific shRNA also activated the hTERT promoter, implying that activation of the hTERT promoter in quiescent Kit 225 cells is associated with cell cycle progression. Our findings suggest that the cell cycle state critically influences Tax-mediated regulation of hTERT expression.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Products, tax/physiology , Human T-lymphotropic virus 1/genetics , T-Lymphocytes/metabolism , Telomerase/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cells, Cultured , DNA Primers/chemistry , Electrophoretic Mobility Shift Assay , G1 Phase/physiology , Humans , Jurkat Cells , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , S Phase/physiology , Telomerase/metabolism , Transcription, Genetic , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...