Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Oncol Rep ; 19(3): 721-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18288407

ABSTRACT

Arctiin is a major lignan constituent of Arctium lappa and has anti-cancer properties in animal models. It was recently reported that arctiin induces growth inhibition in human prostate cancer PC-3 cells. However, the growth inhibitory mechanism of arctiin remains unknown. Herein we report that arctiin induces growth inhibition and dephosphorylates the tumor-suppressor retinoblastoma protein in human immortalized keratinocyte HaCaT cells. We also show that the growth inhibition caused by arctiin is associated with the down-regulation of cyclin D1 protein expression. Furthermore, the arctiin-induced suppression of cyclin D1 protein expression occurs in various types of human tumor cells, including osteosarcoma, lung, colorectal, cervical and breast cancer, melanoma, transformed renal cells and prostate cancer. Depletion of the cyclin D1 protein using small interfering RNA-rendered human breast cancer MCF-7 cells insensitive to the growth inhibitory effects of arctiin, implicates cyclin D1 as an important target of arctiin. Taken together, these results suggest that arctiin down-regulates cyclin D1 protein expression and that this at least partially contributes to the anti-proliferative effect of arctiin.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cyclin D1/metabolism , Down-Regulation , Furans/pharmacology , Glucosides/pharmacology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Neoplasms/metabolism , Retinoblastoma Protein/metabolism
2.
Cancer Sci ; 98(11): 1809-16, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17784872

ABSTRACT

The INK4 family members p16(INK4a) and p15(INK4b) negatively regulate cell cycle progression by inhibition of cyclin-dependent kinase (CDK) 4/6. Loss of p16(INK4a) functional activity is frequently observed in tumor cells, and is thought to be one of the primary causes of carcinogenesis. In contrast, despite the biochemical similarity to p16(INK4a), the frequency of defects in p15(INK4b) was found to be lower than in p16(INK4a), suggesting that p15(INK4b)-inductive agents may be useful for tumor suppression. Here we report the discovery of a novel pyrido-pyrimidine derivative, JTP-70902, which exhibits p15(INK4b)-inducing activity in p16(INK4a)-inactivated human colon cancer HT-29 cells. JTP-70902 also induced another CDK-inhibitor, p27(KIP1), and downregulated the expression of c-Myc and cyclin D1, resulting in G(1) cell cycle arrest. MEK1/2 was identified by compound-immobilized affinity chromatography as the molecular target of JTP-70902, and this was further confirmed by the inhibitory activity of JTP-70902 against MEK1/2 in kinase assays. JTP-70902 suppressed the growth of most colorectal and some other cancer cell lines in vitro, and showed antitumor activity in an HT-29 xenograft model. However, JTP-70902 did not inhibit the growth of COLO320 DM cells; in these, constitutive extracellular signal-regulated kinase phosphorylation was not detected, and neither p15(INK4b) nor p27(KIP1) induction was observed. Moreover, p15(INK4b)-deficient mouse embryonic fibroblasts were found to be more resistant to the growth-inhibitory effect of JTP-70902 than wild-type mouse embryonic fibroblasts. These findings suggest that JTP-70902 restores CDK inhibitor-mediated cell cycle control by inhibiting MEK1/2 and exerts a potent antitumor effect.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , Cell Line, Tumor , Colonic Neoplasms , Colorectal Neoplasms , Humans , Mice , Mice, Nude , Transplantation, Heterologous
3.
Cancer Sci ; 98(9): 1447-53, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17640297

ABSTRACT

Sesamin is a major lignan constituent of sesame and possesses multiple functions such as antihypertensive, cholesterol-lowering, lipid-lowering and anticancer activities. Several groups have previously reported that sesamin induces growth inhibition in human cancer cells. However, the nature of this growth inhibitory mechanism remains unknown. The authors here report that sesamin induces growth arrest at the G1 phase in cell cycle progression in the human breast cancer cell line MCF-7. Furthermore, sesamin dephosphorylates tumor-suppressor retinoblastoma protein (RB). It is also shown that inhibition of MCF-7 cell proliferation by sesamin is correlated with down-regulated cyclin D1 protein expression, a proto-oncogene that is overexpressed in many human cancer cells. It was found that sesamin-induced down-regulation of cyclin D1 was inhibited by proteasome inhibitors, suggesting that sesamin suppresses cyclin D1 protein expression by promoting proteasome degradation of cyclin D1 protein. Sesamin down-regulates cyclin D1 protein expression in various kinds of human tumor cells, including lung cancer, transformed renal cells, immortalized keratinocyte, melanoma and osteosarcoma. Furthermore, depletion of cyclin D1 protein using small interfering RNA rendered MCF-7 cells insensitive to the growth inhibitory effects of sesamin, implicating that cyclin D1 is at least partially related to the antiproliferative effects of sesamin. Taken together, these results suggest that the ability of sesamin to down-regulate cyclin D1 protein expression through the activation of proteasome degradation could be one of the mechanisms of the antiproliferative activity of this agent.


Subject(s)
Cyclins/antagonists & inhibitors , Cyclins/biosynthesis , Dioxoles/pharmacology , Down-Regulation/drug effects , Lignans/pharmacology , Sesame Oil/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D , Cyclins/genetics , Dioxoles/antagonists & inhibitors , G1 Phase/drug effects , Growth Inhibitors/antagonists & inhibitors , Growth Inhibitors/pharmacology , Humans , Lignans/antagonists & inhibitors , Phosphorylation/drug effects , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/physiology , Proteasome Inhibitors , Proto-Oncogene Mas , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/metabolism
4.
Mol Cancer Ther ; 6(5): 1579-87, 2007 May.
Article in English | MEDLINE | ID: mdl-17513607

ABSTRACT

Inactivation of the retinoblastoma protein pathway is the most common abnormality in malignant tumors. We therefore tried to detect agents that induce the cyclin-dependent kinase inhibitor p15(INK4b) and found that ZD1839 (gefitinib, Iressa) could up-regulate p15(INK4b) expression. ZD1839 has been shown to inhibit cell cycle progression through inhibition of signaling pathways such as phosphatidylinositol 3'-kinase-Akt and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) cascades. However, the mechanism responsible for the differential sensitivity of the signaling pathways to ZD1839 remains unclear. We here showed that ZD1839 up-regulated p15(INK4b), resulting in retinoblastoma hypophosphorylation and G(1) arrest in human immortalized keratinocyte HaCaT cells. p15(INK4b) induction was caused by MAPK/ERK kinase inhibitor (PD98059), but not by Akt inhibitor (SH-6, Akt-III). Moreover, mouse embryo fibroblasts lacking p15(INK4b) were resistant to the growth inhibitory effects of ZD1839 compared with wild-type mouse embryo fibroblasts. Additionally, the status of ERK phosphorylation was related to the antiproliferative activity of ZD1839 in human colon cancer HT-29 and Colo320DM cell lines. Our results suggest that induction of p15(INK4b) by inhibition of the MAPK/ERK pathway is associated with the antiproliferative effects of ZD1839.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Quinazolines/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Flavonoids/pharmacology , G1 Phase , Gefitinib , Humans , Models, Biological , Phosphorylation , Signal Transduction
5.
FEBS Lett ; 581(6): 1087-92, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17316622

ABSTRACT

p15(INK4b) functions as a tumor suppressor and implicated in cellular senescence. Here, we show that the Oct-1 binding site in the human p15(INK4b) gene promoter functions as a silencer. Oct-1 specifically interacts with this binding site in vitro and in vivo and SMRT and HDAC1 are present in the p15(INK4b) proximal promoter region. Moreover, mouse embryo fibroblasts (MEFs) lacking Oct-1 have shown significantly increased levels of p15(INK4b) protein compared to their normal counterparts. Treatment with a histone deacetylase (HDAC) inhibitor has activated the expression of p15(INK4b) in wild-type MEFs but has no effect in MEFs lacking Oct-1, suggesting that Oct-1 represses p15(INK4b) gene expression in an HDAC-dependent manner. Finally, we show that the expression of Oct-1 protein significantly decreases, whereas p15(INK4b) protein significantly increases with the cellular aging process. Taken together, these results suggest that Oct-1 is an important transcriptional repressor for p15(INK4b) gene and the transcriptional repression of the p15(INK4b) gene by Oct-1 may be one of the regulatory mechanisms of cellular senescence.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Octamer Transcription Factor-1/physiology , Repressor Proteins/physiology , Transcription, Genetic , Cell Line , Cellular Senescence/genetics , Gene Expression Regulation/physiology , Humans , Transfection
6.
Int J Oncol ; 27(2): 497-503, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16010433

ABSTRACT

15-deoxy-Delta12, 14-prostaglandin J2 (15-d-PGJ2) inhibits cellular proliferation primarily in the G1 phase of the cell cycle. However, the molecular mechanism responsible for this effect has not been sufficiently elucidated. Here, we show that the treatment of human immortalized keratinocyte HaCaT cells with 15-d-PGJ2 arrests the cell cycle at the G1 phase. We also show that inhibition of the proliferation of HaCaT cells by 15-d-PGJ2 correlates with induction of the expression of p15INK4b protein, a cyclin-dependent kinase inhibitor. 15-d-PGJ2 also induces p15INK4b mRNA and its promoter activity, suggesting that 15-d-PGJ2 transcriptionally activates p15INK4b gene expression. Deletion and mutation analyses indicated that one of the elements responsible for 15-d-PGJ2-mediated induction is located between nucleotides -385 and -373 upstream of the initiation codon. An electrophoretic mobility shift assay revealed the specific binding of an unknown protein to this element, and that the binding is augmented by the treatment of 15-d-PGJ2. Although 15-d-PGJ2 is a natural ligand of the peroxisome proliferator-activated receptor gamma (PPARgamma), treatment with ciglitazone, a synthetic PPARgamma ligand, had no effect on p15INK4b gene expression. Collectively, these results indicate that 15-d-PGJ2 activates p15INK4b gene expression through a novel 15-d-PGJ2-responsive element in the p15INK4b promoter in a PPARgamma-independent manner.


Subject(s)
Prostaglandin D2/analogs & derivatives , Blotting, Northern , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , G1 Phase , Gene Expression/drug effects , Humans , Luciferases/genetics , Luciferases/metabolism , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , Prostaglandin D2/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection
7.
Environ Health Prev Med ; 10(2): 72-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-21432144

ABSTRACT

Inactivation of the p16(INK4a) gene is one of the most frequent defects that contribute to oncogenesis in human cancer, since it is a tumor-suppressor gene. Therefore, functional restoration of p16(INK4a) is one of the most effective methods for cancer prevention. We proposed the concept of 'gene-regulating chemoprevention' and 'molecular-targeting prevention' of cancer, which assumes that transcriptional regulation by drugs on tumor-suppressor genes or functionally similar genes to the tumor-suppressor genes contributes to the prevention of human malignancies. The p16(INK4a) homologs p15(INK4b), p18(INK4c) and p19(INK4d) have been recently identified, and these four members constitute the INK4 family of proteins. All directly bind to cyclin D-cyclin dependent kinase (CDK) 4/6 and are therefore specific inhibitors of these complexes. We recently showed that histone deacetylase (HDAC) inhibitors, promising chemopreventive and chemotherapeutical agents, induce p15(INK4b) and p19(INK4d) gene expression and cause growth arrest, suggesting that both genes are important molecular targets for HDAC inhibitors. Furthermore, we found that 12-O-tetradecanoylphorbol-13-acetate (TPA), which is widely used as a tumor promoter and protein kinase C activator, promotes human cancer cell growth through the down-regulation of p18(INK4c) gene expression. This suggests that a mouse two-stage carcinogenesis model using TPA might partially represent the most common human carcinogenesis pathway related to RB. Our results suggest that the INK4 family consists of attractive and promising molecular targets for the 'gene-regulating chemoprevention' and 'molecular-targeting prevention' of cancer.

8.
FEBS Lett ; 576(1-2): 137-40, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15474025

ABSTRACT

Indole-3-carbinol (I3C) is a naturally occurring compound found in vegetables such as broccoli and cauliflower, and has been shown to arrest human tumor cells in the G1 phase of the cell cycle. However, the molecular mechanism responsible for this effect has not been sufficiently elucidated. We report here that I3C activates the cyclin-dependent kinase (CDK) inhibitor p15INK4b gene through its promoter, accompanied by cell growth inhibition in HaCaT cells. Treatment with I3C almost did not affect the expressions of the other CDK inhibitors such as p19INK4d, p21WAF1 and p27Kip1. These results suggest that p15INK4b is an important molecular target of I3C among CDK inhibitors.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Indoles/pharmacology , Tumor Suppressor Proteins/metabolism , Cell Cycle Proteins/drug effects , Cell Line, Transformed , Cyclin-Dependent Kinase Inhibitor p15 , Dose-Response Relationship, Drug , Down-Regulation , G1 Phase/drug effects , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Keratinocytes/metabolism , Kinetics , Promoter Regions, Genetic , RNA, Messenger/drug effects , Tumor Suppressor Proteins/drug effects
9.
FEBS Lett ; 574(1-3): 171-5, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15358560

ABSTRACT

We have reported that histone deacetylase (HDAC) inhibitors activate a member of the INK4 family, the p19INK4d gene, causing G1 phase arrest. We report here that HDAC inhibitor, Trichostatin A, activates another member of the INK4 family, the p18INK4c gene, through its promoter in Jurkat cells. Interestingly, the activation patterns of the p18INK4c gene were different from those of p19INK4d. Furthermore, mouse embryo fibroblasts lacking p18Ink4c or p18Ink4c/p19Ink4d were resistant to the growth inhibitory effects of TSA as compared to their wild-type counterpart. Our findings suggest that p18INK4c is involved in TSA-mediated cell growth inhibition and cooperates with p19INK4d.


Subject(s)
Cell Cycle Proteins/genetics , Gene Expression Regulation/drug effects , Hydroxamic Acids/pharmacology , Tumor Suppressor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p18 , Cyclin-Dependent Kinase Inhibitor p19 , Humans , Jurkat Cells , Promoter Regions, Genetic , RNA, Messenger/genetics
10.
Oncogene ; 23(31): 5409-14, 2004 Jul 08.
Article in English | MEDLINE | ID: mdl-15107819

ABSTRACT

p18(INK4c), a member of INK4 family of cyclin-dependent kinase inhibitors, negatively regulates the cyclin D-cyclin-dependent kinase 4/6 complexes which promote G1/S transition by phosphorylating the retinoblastoma tumor-suppressor gene product. Several recent studies using p18(INK4c)-null mice revealed that the p18(INK4c) plays an important role in cell proliferation and tumor development. We report here that 12-O-tetradecanoylphorbol-13-acetate (TPA), widely used as a protein kinase C (PKC) activator, suppresses the expression of p18(INK4c) through its promoter, accompanied by the induction of human cancer cell growth. Reduction of p18(INK4c) using small interfering RNA (siRNA) also enhanced cell growth, suggesting that p18(INK4c) is a critical target of TPA. Ro 31-8425, a potent and highly specific PKC inhibitor abrogated the suppressive effect of TPA on p18(INK4c) gene expression. However, the expression of dominant-negative c-Jun (TAM-67) did not inhibit the action of TPA on p18(INK4c). These findings suggest that activation of PKC promotes human cancer cell growth through downregulation of p18(INK4c) in an AP-1 activation-independent manner. These results suggest that the accelerated cellular proliferation of some human tumors caused by enhanced PKC activity at least partially involves the suppression of p18(INK4c), which is a ubiquitously expressed cyclin-dependent kinase inhibitor.


Subject(s)
Cell Cycle Proteins/metabolism , Down-Regulation , Protein Kinase C/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Blotting, Western , Cell Division , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p18 , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , G1 Phase , Genes, Dominant , Humans , Mice , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , S Phase , Tetradecanoylphorbol Acetate , Time Factors , Transfection
11.
Oncogene ; 23(31): 5340-9, 2004 Jul 08.
Article in English | MEDLINE | ID: mdl-15107822

ABSTRACT

Histone deacetylase (HDAC) inhibitors are known to arrest human tumor cells at the G1 phase of the cell cycle and activate the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1). However, several studies have suggested the existence of a p21(WAF1/Cip1)-independent molecular pathway. We report here that HDAC inhibitors activate a member of the INK4 family, the INK4d gene, causing G1 phase arrest, in the human T cell leukemia cell line, Jurkat. One of the major Trichostatin A (TSA)-responsive elements is a specific Sp1 binding site in the INK4d promoter. Electrophoretic mobility-shift assay revealed that Sp1 and Sp3 can specifically interact with this Sp1 binding site. Furthermore, using chromatin immunoprecipitation assay, we demonstrated that HDAC2 was present in the INK4d proximal promoter region in the absence, but not the presence, of TSA. Taken together, these results suggest that treatment with TSA transcriptionally activates INK4d by releasing HDAC2 from the histone-DNA complex at the INK4d promoter. Using a p21(WAF1/Cip1)-deleted human colorectal carcinoma cell line, HCT116 p21 (-/-), we show that upregulation of p19(INK4d) by TSA is associated with inhibition of cell proliferation. Moreover, mouse embryo fibroblasts lacking Ink4d were resistant to the growth inhibitory effects of TSA as compared to their wild-type counterpart. Our findings suggest that p19(INK4d) in addition to p21(WAF1/Cip1) is an important molecular target of HDAC inhibitors inducing growth arrest.


Subject(s)
Cell Cycle Proteins/metabolism , Histone Deacetylase Inhibitors , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Animals , Binding Sites , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Division , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p19 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , G1 Phase , Gene Deletion , Humans , Hydroxamic Acids/pharmacology , Jurkat Cells , Luciferases/metabolism , Mice , Mutation , Plasmids/metabolism , Precipitin Tests , Protein Binding , RNA, Messenger/metabolism , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor , T-Lymphocytes/metabolism , Time Factors , Transcription Factors/metabolism , Transfection , Up-Regulation
12.
FEBS Lett ; 554(3): 347-50, 2003 Nov 20.
Article in English | MEDLINE | ID: mdl-14623092

ABSTRACT

Histone deacetylase (HDAC) inhibitors arrest human tumor cells at the G1 phase of the cell cycle and activate the cyclin-dependent kinase inhibitor, p21(WAF1/Cip1). However, several studies have suggested the existence of a p21(WAF1/Cip1)-independent molecular pathway. We report here that HDAC inhibitors, trichostatin A (TSA) and sodium butyrate, activate the p15(INK4b) gene, a member of the INK4 gene family, through its promoter in HaCaT cells. Furthermore, we show that up-regulation of p15(INK4b) by TSA is associated with cell growth inhibition of HCT116 p21 (-/-) cells. Our findings suggest that p15(INK4b) is one of the important molecular targets of HDAC inhibitors.


Subject(s)
Butyrates/pharmacology , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Tumor Suppressor Proteins , Blotting, Northern , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA/analysis , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Genetic Vectors/genetics , HCT116 Cells , Humans , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Up-Regulation
13.
Environ Health Prev Med ; 8(5): 157-60, 2003 Nov.
Article in English | MEDLINE | ID: mdl-21432092

ABSTRACT

One of the best approaches against cancer is prevention. Inactivation of the p53 or p16(INK4a) genes has been extensively reported in most human cancer cells. Both p53 and p16(INK4a) function as tumor suppressors. Therefore, functional restoration of these molecules is considered to be one of the most useful methods for cancer prevention and therapy. We have proposed a concept termed 'gene-regulating chemoprevention and chemotherapy' regarding the above pathway. This concept assumes that transcriptional regulation by drugs on tumor-suppressor genes, downstream target genes or functionally similar genes (for example, family genes) of the tumor-suppressor genes would contribute to the prevention of human malignancies. Histone deacetylase (HDAC) inhibitors have been shown to be potent inducers of growth arrest, differentiation and apoptotic cell death. Previously, we demonstrated that HDAC inhibitors, such as sodium butyrate and trichostatin A (TSA), transcriptionally induce the cyclin-dependent kinase inhibitor p21(WAF1/Cip1), a downstream target gene of p53, in a p53-independent manner. Furthermore, we have recently shown that HDAC inhibitors activate Gadd45, another downstream target gene of p53, and p19(INK4d), a gene functionally similar to p16(INK4a). Our results, taken together with previous findings, suggest that HDAC inhibitors may be one of the most attractive and promising agents for 'gene-regulating chemoprevention' and 'molecular-targeting prevention' of cancer.

14.
FEBS Lett ; 517(1-3): 272-6, 2002 Apr 24.
Article in English | MEDLINE | ID: mdl-12062451

ABSTRACT

p19(INK4d), a member of the INK4 family of cyclin-dependent kinase (CDK) inhibitors, negatively regulates the cyclin D-CDK4/6 complexes, which promote G1/S transition by phosphorylating the retinoblastoma tumor-suppressor gene product. To investigate the mechanism of transcriptional regulation of the p19(INK4d) gene, we characterized the 5'-flanking region of the human p19(INK4d) gene. The cap-site hunting method revealed that the transcription starts at -16 nucleotide (nt) upstream of the initiation codon. The 5'-flanking region of the human p19(INK4d) gene was ligated to a luciferase reporter gene and possessed functional promoter activity. Luciferase assay with a series of truncated 5'-flanking regions indicated that the region from -81 to -2 nt could drive the transcription of the p19(INK4d) gene. Several Sp1 and activating protein 2 binding sites are located within the region from -81 to -2 nt. Mutation of the second Sp1 binding site from -33 to -25 nt decreased the promoter activity. Collectively, it was demonstrated that the human p19(INK4d) gene is under the control of TATA-less promoter and the Sp1 binding site is involved in the transcription.


Subject(s)
5' Flanking Region/genetics , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/genetics , Transcription, Genetic , Base Sequence , Binding Sites , Cloning, Molecular , Cyclin-Dependent Kinase Inhibitor p19 , DNA-Binding Proteins/genetics , Genes, Reporter , Humans , Molecular Sequence Data , Mutagenesis , Sequence Analysis , Sequence Deletion , Transcription Factor AP-2 , Transcription Factors/genetics , Transcription Initiation Site , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...