Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Oncogene ; 36(22): 3104-3118, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28068324

ABSTRACT

Survival signaling is critical for the metastatic program of cancer cells. The current study investigated the role of Akt survival proteins in colorectal cancer (CRC) metastasis and explored potential mechanisms of Akt-mediated metastasis regulation. Using an orthotopic implantation model in mice, which uniquely recapitulates the entire multistep process of CRC metastasis, combined with an inducible system of short hairpin RNA-mediated Akt isoform knockdown in human CRC cells, our studies confirm a role of Akt2 in CRC cell dissemination to distant organs in vivo. Akt2 deficiency profoundly inhibited the development of liver lesions in mice, whereas Akt1 had no effect under the experimental conditions used in the study. Array analysis of human metastatic genes identified the scaffolding protein metastasis suppressor 1 (MTSS1) as a novel Akt2-regulated gene. Inducible loss of Akt2 in CRC cells robustly upregulated MTSS1 at the messenger RNA and protein level, and the accumulated protein was functionally active as shown by its ability to engage an MTSS1-Src-cortactin inhibitory axis. MTSS1 expression led to a marked reduction in levels of functional cortacin (pcortactin Y421), an actin nucleation-promoting factor that has a crucial role in cancer cell invasion and metastasis. MTSS1 was also shown to mediate suppressive effects of Akt2 deficiency on CRC cell viability, survival, migration and actin polymerization in vitro. The relevance of these findings to human CRC is supported by analysis of The Cancer Genome Atlas (TCGA) and NCBI GEO data sets, which demonstrated inverse changes in expression of Akt2 and MTSS1 during CRC progression. Taken together, the data identify MTSS1 as a new Akt2-regulated gene, and point to suppression of MTSS1 as a key step in the metastasis-promoting effects of Akt2 in CRC cells.


Subject(s)
Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Microfilament Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Animals , Colorectal Neoplasms/genetics , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Nude , Microfilament Proteins/genetics , Neoplasm Metastasis , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics
2.
Curr Med Chem ; 18(36): 5528-44, 2011.
Article in English | MEDLINE | ID: mdl-22172063

ABSTRACT

The phosphatidylinositol-3-kinase (PI3K)/AKT/mTOR signaling pathway is a central regulator in cell proliferation, growth, and angiogenesis. Inhibition of this pathway therefore is a major strategy for cancer chemotherapy. In order to induce the maximal therapeutic outcome in cancer treatment, vertical inhibition of the PI3K/AKT/mTOR pathway or horizontal inhibition of PI3K/AKT/mTOR and other kinases has been reported. In this review, we discuss the drug design and clinical development of dual inhibitors of PI3K and mTOR as well as the mTOR-selective inhibitors, classified based on the mechanism of action and the chemical structures. Structural determinants for increasing selectivity toward PI3Kα or mTOR are revealed from the structure-activity relationship of the reported inhibitors. Current clinical development in combination therapy of inhibitors involving in the PI3K/AKT/mTOR pathway is also discussed.


Subject(s)
Enzyme Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Humans , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/enzymology , Signal Transduction/drug effects , Structure-Activity Relationship , Substrate Specificity
3.
Cancer Res ; 61(16): 6239-47, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507078

ABSTRACT

The pancreatic cancer cell line, MIA PaCa-2 is not responsive to transforming growth factor beta (TGF-beta) because of a lack of expression of the TGF-beta type II receptor (RII). We show that the lack of RII expression is caused by a deficit of the transcription factor Sp1. Nuclear run-off assays and Western immunoblot showed low levels of transcription and protein levels of Sp1, respectively. Treatment of MIA PaCa-2 cells with the DNA methyl transferase inhibitor, 5-aza-2'-deoxycytidine, resulted in an increase in the rate of Sp1 transcription, in Sp1 protein expression, and in the binding of Sp1 to the RII promoter. Ectopic expression of Sp1 cDNA in MIA PaCa-2 cells led to an increase in RII promoter-chloramphenicol acetyltransferase activity and RII expression. Expression of Sp1 cDNA also caused a reduction in both growth and clonogenicity that was associated with restoration of responsiveness to TGF-beta. Conversely, cells that express RII (BxPC-3 and MIA PaCa-2 Sp1 transfectants) when treated with mithramycin, an inhibitor of Sp1 binding, showed a reduction in RII mRNA expression. The reduction of RII mRNA was attributed to a decrease in RII promoter-chloramphenicol acetyltransferase activity that was associated with a decrease in Sp1 binding to the RII promoter. These data indicate that transcriptional repression of the Sp1 gene in MIA PaCa-2 cells plays a role in the transcriptional inactivation of the RII gene and thus lack of responsiveness to TGF-beta.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Pancreatic Neoplasms/genetics , Receptors, Transforming Growth Factor beta/genetics , Sp1 Transcription Factor/genetics , Transcriptional Activation/drug effects , Antibiotics, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Cell Division/drug effects , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA Modification Methylases/antagonists & inhibitors , DNA, Complementary/genetics , DNA, Complementary/metabolism , Decitabine , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Genetic Vectors/genetics , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Plicamycin/pharmacology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Sp1 Transcription Factor/biosynthesis , Sp1 Transcription Factor/metabolism , Transcriptional Activation/physiology , Transfection , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology , Tumor Cells, Cultured/drug effects
4.
J Cell Physiol ; 188(3): 383-93, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11473365

ABSTRACT

In this study, we address whether TGFbeta signaling mediates vitamin D3 analog-induced growth inhibition in nonmalignant and malignant breast cells. Normal mammary epithelial cells (184), immortalized nonmalignant mammary epithelial cells (184A1 and MCF10A), and breast cancer cells (early passage MCF7: MCF7E) were sensitive to the inhibitory effects of vitamin D3 analogs (EB1089 and MC1288) while late passage MCF7 breast cancer (MCF7L) cells were relatively resistant. A similar pattern of sensitivity to TGFbeta was observed with these cells. Thus, the sensitivity to the vitamin D3 analogs correlated with the sensitivity to TGFbeta. MCF7L TGFbetaRII-transfected cells, which have autocrine TGFbeta activity, were more sensitive to EB1089 than MCF7L cells. TGFbeta neutralizing antibody was found to block the inhibitory effects of these analogs. These results are consistent with the idea that autocrine TGFbeta signaling mediates the anti-proliferative effects of the vitamin D3 analogs in these cells. The expression of TGFbeta isoforms and/or TGFbeta receptors was induced by the analogs in the vitamin D3 and TGFbeta sensitive cells. Vitamin D3 analogs did not induce TGFbeta or TGFbeta receptor expression in the resistant MCF7L cells. Therefore, EB1089 induces autocrine TGFbeta activity through increasing expression of TGFbeta isoforms and/or TGFbeta receptors. In addition, EB1089 induced nuclear VDR protein levels in the sensitive 184A1 cells but not in the resistant MCF7L cells. 184A1 cells were more sensitive to EB1089-induced VDR-dependent transactivation than MCF7L cells as measured by a luciferase reporter construct containing the VDRE, indicating a defect of VDR signaling in MCF7L cells. Smad3, a TGFbeta signaling mediator, coactivated VDR-dependent transactivation in 184A1 cells but not in MCF7L cells. These results indicate that Smad3 coactivates VDR to further enhance TGFbeta signaling and vitamin D3 signaling in the sensitive 184A1 cells. The results also indicate that Smad3 is not of itself sufficient to coactivate VDR in TGFbeta/vitamin D3 resistant MCF7L cells and other factors are required. We found that the PI 3-kinase pathway inhibitor LY29004 inhibited the synergy of TGFbeta and EB1089 on VDR-dependent transactivation activity. This indicates that the crosstalk between TGFbeta and vitamin D signaling is also PI 3-kinase pathway dependent.


Subject(s)
Activin Receptors, Type I , Autocrine Communication/drug effects , Breast Neoplasms/metabolism , Breast/drug effects , Cholecalciferol/pharmacology , Transforming Growth Factor beta/metabolism , Antibodies, Blocking/pharmacology , Antineoplastic Agents/pharmacology , Breast/cytology , Breast/metabolism , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cell Division/drug effects , Cell Line , Cholecalciferol/analogs & derivatives , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Female , Gene Expression/drug effects , Genes, Reporter , Humans , Protein Serine-Threonine Kinases/biosynthesis , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Calcitriol/metabolism , Receptors, Transforming Growth Factor beta/biosynthesis , Signal Transduction/drug effects , Smad3 Protein , Thymidine/metabolism , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transfection , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology
5.
J Biol Chem ; 276(35): 32854-9, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11443124

ABSTRACT

We have previously reported that Sp3 acts as a transcriptional repressor of transforming growth factor-beta receptors type I (RI) and type II (RII). We now present data suggesting that treatment of MCF-7L breast and GEO colon cancer cells with 5-aza cytidine (5-azaC) leads to down-regulation of Sp3 and the concomitant induction of RI and RII. Western blot and gel shift analyses on 5-azaC-treated MCF-7L and GEO nuclear extracts indicated reduced Sp3 protein levels and decreased binding of Sp3 protein to radiolabeled consensus Sp1 oligonucleotide. Southwestern analysis detected decreased binding of Sp3 to RI and RII promoters in 5-azaC-treated MCF-7L and GEO cells, suggesting a correlation between decreased Sp3 binding and enhanced RI and RII expression in these cells. Reverse transcription-polymerase chain reaction and nuclear run-on data from 5-azaC-treated MCF-7L and GEO cells indicated down-regulation of Sp3 mRNA as a result of decreased transcription of Sp3. We reported earlier that 5-azaC treatment induces RI and RII expression through increased Sp1 protein levels/activities in these cells. These studies demonstrate that the effect of 5-azaC involves a combination of effects on Sp1 and Sp3.


Subject(s)
Activin Receptors, Type I , Azacitidine/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation/physiology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Breast Neoplasms , Cell Nucleus/metabolism , Colonic Neoplasms , Female , Gene Expression Regulation/drug effects , Humans , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sp3 Transcription Factor , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured
6.
J Biol Chem ; 276(5): 3348-52, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11027677

ABSTRACT

MCF-7E breast cancer cells express transforming growth factor-beta (TGF-beta) receptors RI and RII in comparison to MCF-7L cells. We present data showing that Sp3 acts as a transcriptional repressor of RI and RII in MCF-7L cells and GEO colon cancer cells. MCF-7L and GEO cells express high levels of Sp3 protein. Gel shift analysis indicated enhanced binding of Sp3 from MCF-7L cells to a consensus Sp1 oligonucleotide. Southwestern data indicated increased binding of Sp3 to RI and RII promoters in MCF-7L cells, suggesting a correlation between Sp3 binding and reduced expression of TGF-beta receptors in MCF-7L cells. Cotransfection of CMV-Sp3 cDNA with RI and RII promoter-luciferase reporter constructs decreased RI and RII promoter activities by 70% in MCF-7E and GEO cells. Southwestern analysis detected the binding of transiently expressed Sp3 to RI and RII promoters in MCF-7E cells. Significantly, ectopic Sp3 expression led to repression of RI and RII transcripts in MCF-7E cells. This report demonstrates that inappropriate overexpression of Sp3 is a mechanism that contributes to repression of TGF-beta receptors.


Subject(s)
DNA-Binding Proteins/physiology , Receptors, Transforming Growth Factor beta/genetics , Transcription Factors/physiology , Transcription, Genetic/physiology , DNA/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Promoter Regions, Genetic/physiology , Sp3 Transcription Factor , Tumor Cells, Cultured
7.
Oncogene ; 19(40): 4574-81, 2000 Sep 21.
Article in English | MEDLINE | ID: mdl-11030146

ABSTRACT

Akt, when activated by IGF/insulin, can phosphorylate forkhead transcription factors. We undertook this study to determine whether epidermal growth factor (EGF) treatment could produce a signaling cascade resulting in phosphorylation of the forkhead transcription factor FKHR in a breast cancer cell line, MDA-MB-231. After establishing ErbB1, cbl, PI3 kinase and Akt were activated in EGF treated MDA-MB-231, we determined by immunoblot with FKHR antiserum that the electrophoretic mobility of FKHR was retarded after EGF treatment. This mobility retardation was reversible by treatment with alkaline phosphatase, and immunoblot with phospho-Ser256 FKHR antibody further confirmed phosphorylation on an Akt consensus site after EGF treatment. EGF stimulated FKHR phosphorylation was blocked by the PI3 kinase inhibitor LY294002, and the ErbB1 inhibitor AG1478. FKHR immunoblotting after purification of nuclear and cytoplasmic proteins showed that EGF induced a simultaneous increase of FKHR in the cytoplasm and decrease in the nucleus. This finding was confirmed by immunofluorescence staining. Treatment of cells with pharmacological inhibitors of PI3 kinase or ErbB1 blocked this effect. Thus, these results demonstrate the phosphorylation and nuclear exclusion of FKHR after EGF treatment by a PI3 kinase dependent mechanism, and represent the first report of growth factor regulation of endogenous FKHR localization.


Subject(s)
Breast Neoplasms/pathology , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/physiology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/physiology , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Ubiquitin-Protein Ligases , Alkaline Phosphatase/pharmacology , Biological Transport/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chromones/pharmacology , Cytoplasm/metabolism , ErbB Receptors/drug effects , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors , Humans , Macromolecular Substances , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-cbl , Quinazolines , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tyrphostins/pharmacology
8.
Oncogene ; 19(40): 4660-7, 2000 Sep 21.
Article in English | MEDLINE | ID: mdl-11030155

ABSTRACT

In this report, we describe the mechanism of TGF-beta receptor type I (RI) repression in the GEO human colon carcinoma cells. Treatment of GEO cells with the DNA methyltransferase inhibitor, 5 azacytidine induced RI expression and restored TGF-beta response. A stably transfected RI promoter-reporter construct (RI-Luc) expressed higher activity in the 5 aza C treated GEO cells, suggesting the activation of a transactivator for RI transcription. Gel shift analysis indicated enhanced binding of proteins from the 5 aza C treated nuclear extracts to radiolabeled Sp1 oligonucleotides specifically contained in the RI promoter. Protein stability studies after cyclohexamide treatment suggested an increase in the Sp1 protein stability from the 5 aza C treated GEO cells. Further, transfection of Sp1 cDNA into untreated GEO control cells increased RI promoter activity and thus induced RI expression. 5 aza C mediated Sp1 expression in Sp1 deficient GEO colon and MCF-7 breast cancer cells also enhanced the activity of several other Sp1 dependent promoters such as TGF-beta receptor type II (RII), Cyclin A and p21/waf1/cip1. These results indicate that restoration of Sp1 in several different types of Sp1 deficient cells leads to enhanced activation of a wide range of Sp1 dependent promoters.


Subject(s)
Activin Receptors, Type I , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Sp1 Transcription Factor/deficiency , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin A/biosynthesis , Cyclin A/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Protein Serine-Threonine Kinases/biosynthesis , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Recombinant Fusion Proteins/physiology , Sp1 Transcription Factor/physiology , Transcriptional Activation , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
9.
Oncogene ; 19(15): 1901-14, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10773880

ABSTRACT

Transforming growth factor alpha (TGFalpha) is widely expressed in malignant as well as normal cells and is involved in regulating cell growth and differentiation. Although processing of TGFalpha has been extensively studied in normal cells, there is little information regarding TGFalpha cleavage in malignant cells. Therefore, we compared the processing of TGFalpha in two human colon carcinoma cell lines. We found that there was a defective cleavage pattern for the TGFalpha precursor resulting in retention of partially processed TGFalpha on the cell surface of both the HCT116a2alphaS3 and CBS4alphaS2 cell lines. This raised the possibility that signaling from the resulting defective cleavage species could differ from that of soluble TGFalpha. The membrane-associated TGFalpha induced higher phosphorylation of EGFR on the cell surface of adjacent cells than equivalent levels of mature TGFalpha. The interaction of membrane bound TGFalpha precursor with the EGFR caused a slower internalization of activated EGFR relative to the internalization of the soluble TGFalpha/EGFR complexes. In addition, the tethered TGFalpha was resistant to the ability of protein-tyrosine phosphatases (PTPs) to reduce EGFR tyrosine phosphorylation, also contributing to higher activation of EGFR. The enhanced activation of EGFR by the tethered form of TGFalpha was reflected by higher activation of Grb2, SHC and Erk downstream mediators of EGF receptor signaling. The higher activation of EGFR by membrane tethered TGFalpha indicates that defective TGFalpha processing provides a mechanism whereby malignant cells can obtain a growth advantage over normal cells.


Subject(s)
Colonic Neoplasms/metabolism , ErbB Receptors/metabolism , Transforming Growth Factor alpha/metabolism , Cell Membrane/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Neutralization Tests , Phosphorylation , Protein Precursors/metabolism , Protein Processing, Post-Translational , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Transfection , Transforming Growth Factor alpha/immunology , Tumor Cells, Cultured
10.
J Biol Chem ; 275(16): 12231-6, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10766860

ABSTRACT

Progression of MCF-7 cells from early passage (MCF-7E, <200 passage) to late passage (MCF-7L, >500 passage) correlates with a loss of sensitivity to exogenous TGFbeta1. We have previously shown that loss of TGFbeta sensitivity is due to decreased expression of the transforming growth factor receptor type II (TbetaRII) and is associated with increased tumorigenicity in nude mice. Reduced TbetaRII expression in MCF-7L cells is caused by decreased TbetaRII promoter activity in this cell line. Our previous studies using 5' deletion constructs of this promoter revealed that MCF-7L cells were unable to support transcription of the minimal promoter (-47 to +2) to the same levels as the MCF-7E cells. This region of the promoter contains an Sp1 element at position -25 from the major transcription start site. In this study, we investigated the role of Sp1 in TbetaRII transcription. Mutation of the Sp1 site resulted in decreased transcription of TbetaRII in MCF-7E and MCF-7L cells, indicating that this site played a role in transcription of this promoter. Gel shift assays using the proximal Sp1 site from the TbetaRII promoter showed enhanced DNA:protein complex formation with nuclear proteins isolated from MCF-7E cells compared with MCF-7L cells. Supershift analysis identified this binding activity as Sp1. Western blot analysis of Sp1 levels demonstrated that MCF-7E cells contain increased Sp1 protein compared with MCF-7L cells, paralleling the increased binding activity. Differential Sp1 activity was also demonstrated by higher levels of transcription of an Sp1-dependent insulin-like growth factor II promoter construct in MCF-7E cells compared with MCF-7L cells. Co-transfection of an Sp1 expression vector with a TbetaRII promoter construct in MCF-7L cells induced the expression from the promoter-CAT constructs and resulted in an increase of endogenous TbetaRII protein levels. These results demonstrate that the transcriptional repression of TbetaRII in MCF-7L cells is caused, in part, by lower Sp1 levels.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Sp1 Transcription Factor/physiology , Transforming Growth Factor beta/biosynthesis , Adenocarcinoma/genetics , Animals , Breast Neoplasms/genetics , Electrophoresis, Polyacrylamide Gel , Female , Humans , Mice , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Transfection , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
11.
Oncogene ; 18(43): 5923-35, 1999 Oct 21.
Article in English | MEDLINE | ID: mdl-10557079

ABSTRACT

Autocrine TGFalpha is an important growth effector in the transformed phenotype. Growth stimulation of some colon cancer cells as well as other types of cancer cells is effected by activation of the epidermal growth factor receptor. Importantly, this receptor activation leads to further stimulation of TGFalpha transcription and increased peptide synthesis. However, the molecular mechanism by which TGFalpha transcription is activated is poorly understood. In this paper, we describe the localization of a cis-sequence within the TGFalpha promoter which mediates this stimulation. This region contains parallel cis-acting elements which interact to regulate both basal and EGF-induced TGFalpha expression. The well differentiated colon carcinoma cell line designated FET was employed in these studies. It produces autocrine TGFalpha but requires exogenous EGF in the medium for optimal growth. Addition of EGF to FET cells maintained in the absence of EGF resulted in a 2 - 3-fold increase of both TGF promoter activity and endogenous TGFalpha mRNA at 4 h. This addition of EGF also stimulated protein synthesis. The use of deletion constructs of the TGFalpha promoter in chimeras with chloramphenicol acetyl transferase localized EGF-responsiveness to between -247 and -201 within the TGFalpha promoter. A 25 bp sequence within this region conferred EGF-responsiveness to heterologous promoter constructs. Further use of deletion/mutation chimeric constructs revealed the presence of at least two interacting cis-elements, one binding a repressor activity and the other, an activator. Gel shift studies indicate the presence of distinct complexes representing activator and repressor binding, which are positively modulated by EGF. The type and amount of complexes formed by these proteins interact to regulate both the basal activity and EGF-responsiveness of the TGFalpha promoter. The interaction of an activator protein with an EGF-responsive repressor may serve to regulate the level of this progression-associated, transforming protein within tight limits.


Subject(s)
Epidermal Growth Factor/metabolism , Promoter Regions, Genetic , Response Elements , Transforming Growth Factor alpha/genetics , Cell Line , Epidermal Growth Factor/pharmacology , Gene Expression Regulation/drug effects , Humans , Kinetics , Oligonucleotides , Recombinant Fusion Proteins/genetics , Sequence Deletion , Thymidine Kinase/genetics , Transcription, Genetic/drug effects
12.
Cancer Res ; 59(18): 4725-31, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10493532

ABSTRACT

Transforming growth factor betas (TGF-betas) are a growth factor family with negative autocrine growth functions for most epithelial cells including colon carcinoma cell lines. Both type I (RI) and type II (RII) transmembrane TGF-beta receptors have been shown to be indispensable for TGF-beta-mediated cell growth regulation. Previous studies using different model systems have shown that both overexpression of TGF-beta1 and transfection of antisense TGF-beta1 to reduce TGF-beta1 expression could lead to increased tumorigenicity. These results are seemingly contradictory and suggest that effects of TGF-beta modulation on malignant properties of cancer cells may be contextual. This study addresses this issue using human colon carcinoma cells (CBS and FET) to determine the effects of modulation of the various components of the TGF-beta system on in vitro and in vivo growth properties in two independent isogenic models of colon carcinoma. Cells were stably transfected with a tetracycline-repressible RII expression vector (CBS4-RII), a tetracycline-repressible expression vector containing a truncated RII cDNA lacking the serine/threonine kinase domain (CBS4-deltaRII and FET6-deltaRII), or with a vector containing the TGF-beta1 cDNA (CBS4-beta1S and FET-beta1S). Expression of the truncated RII reduced TGF-beta sensitivity, whereas overexpression of RII increased TGF-beta sensitivity. TGF-beta overexpression did not affect TGF-beta response. In vivo tumorigenicity assays revealed that CBS4-RII cells had lower tumorigenicity than control cells, whereas CBS4-deltaRII and CBS4-beta1S had higher tumorigenicity than controls. The CBS4 cells are poorly tumorigenic in athymic mice, and the wild-type FET6 cells are nontumorigenic. FET6-deltaRII cells formed rapidly growing tumors, and FET-beta1S cells also formed tumors. These data illustrate the paradoxical tumor-promoting and -suppressing effects of TGF-beta signaling activity in two isogenic model systems from human colon carcinomas, thus demonstrating that the effects of modulation of TGF-beta expression or TGF-beta signaling capability affects malignancy in a contextual manner.


Subject(s)
Colonic Neoplasms/pathology , Transforming Growth Factor beta/physiology , Animals , Cell Division/drug effects , Culture Media, Conditioned , Genes, Reporter , Humans , Luciferases/genetics , Mice , Mice, Nude , Receptors, Transforming Growth Factor beta/physiology , Recombinant Proteins/metabolism , Time Factors , Transfection , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/toxicity , Transplantation, Heterologous , Tumor Cells, Cultured , beta-Galactosidase/genetics
13.
J Biol Chem ; 274(18): 12840-7, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10212271

ABSTRACT

Ectopic expression of the alpha5 integrin subunit in cancer cells with little or no endogenous expression of this integrin often results in reduced proliferation as well as reduced malignancy. We now show that inhibition resulting from ectopic expression of alpha5 integrin is due to induction of autocrine negative transforming growth factor-beta (TGF-beta) activity. MCF-7 breast cancer cells do not express either alpha5 integrin or type II TGF-beta receptor and hence are unable to generate TGF-beta signal transduction. Ectopic expression of alpha5integrin expression enhanced cell adhesion to fibronectin, reduced proliferation, and increased the expression of type II TGF-beta receptor mRNA and cell surface protein. Receptor expression was increased to a higher level in alpha5 transfectants by growth on fibronectin-coated plates. Induction of type II TGF-beta receptor expression also resulted in the generation of autocrine negative TGF-beta activity because colony formation was increased after TGF-beta neutralizing antibody treatment. Transient transfection with a TGF-beta promoter response element in tandem with a luciferase cDNA into cells stably transfected with alpha5 integrin resulted in basal promoter activities 5-10-fold higher than those of control cells. Moreover, when alpha5 transfectants were treated with a neutralizing antibody to either TGF-beta or integrin alpha5, this increased basal promoter activity was blocked. Autocrine TGF-beta activity also induced 3-fold higher endogenous fibronectin expression in alpha5 transfectants relative to that of control cells. Re-expression of type II receptor by alpha5 transfection also restored the ability of the cells to respond to exogenous TGF-beta and led to reduced tumor growth in athymic nude mice. Taken together, these results show for the first time that TGF-beta type II receptor expression can be controlled by alpha5beta1 ligation and integrin signal transduction. Moreover, TGF-beta and integrin signal transduction appear to cooperate in their tumor-suppressive functions.


Subject(s)
Receptors, Fibronectin/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Cell Division , Fibronectins/metabolism , Humans , Mice , Mice, Nude , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Transfection/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
14.
Cancer Res ; 59(2): 320-4, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9927040

ABSTRACT

We previously demonstrated that mutational inactivation of transforming growth factor beta type II receptors (RIIs) is very common among the 13% of human colon cancers with microsatellite instability. These mutations principally cluster in the BAT-RII polyadenine sequence repeat. Among microsatellite stable (MSS) colon cancers, we now find that non-BAT-RII point mutations inactivate RII in another 15% of cases, thus doubling the known number of colon cancers in which RII mutations are pathogenetic. Functional analysis confirms that these mutations inactivate RII signaling. Moreover, another 55% of MSS colon cancers demonstrate a transforming growth factor beta signaling blockade distal to RII. The transforming growth factor beta pathway and RII in particular are major targets for inactivation in MSS colon cancers as well as in colon cancers with microsatellite instability.


Subject(s)
Colonic Neoplasms/genetics , Microsatellite Repeats , Mutation , Receptors, Transforming Growth Factor beta/genetics , Adult , Female , Humans , Male , Middle Aged , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured
15.
J Cell Physiol ; 177(3): 387-95, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9808147

ABSTRACT

Previously, we reported that unaggressive, growth factor-dependent FET human colon carcinoma cells downregulated their transforming growth factor alpha (TGFalpha) expression in a quiescent state (G0/G1) induced by growth factor and nutrient deprivation (Mulder, 1991, Cancer Res., 51:2256-2262). In contrast, highly aggressive, growth factor-independent HCT116 human colon carcinoma cells aberrantly upregulated this autocrine activity in the quiescent state (Mulder, 1991, Cancer Res., 51:2256-2262; Howell et al., 1998, Mol. Cell. Biol., 18:303-313). In this report, the role of autocrine TGFalpha and the mechanism of its regulation of expression during reentry into the cell cycle from a noncycling growth state were determined in FET cells. Optimal induction of DNA synthesis from a quiescent state in FET cells is dependent upon autocrine TGFalpha as well as exogenous transferrin and insulin. Reentry into the cell cycle resulting from treatment with exogenous transferrin and insulin resulted in approximately 3-fold induction of TGFalpha expression within 1 hr. TGFalpha induction was controlled at the transcription level, and the cis-controlling element was localized to the region between bp -370 - -201 relative to the translation start codon within the TGFalpha promoter. Thus neutralization of autocrine TGFalpha protein revealed that the induced TGFalpha autocrine activity was necessary for DNA synthesis and acted only in the early G1 phase of the cell cycle. Blockade of autocrine TGFalpha expression early in the cell cycle resulted in the reduction of DNA synthesis, whereas treatment with neutralization antibody at later times had no effect. This suggested that autocrine TGFalpha functions to initiate cell growth from noncycling states. This was further confirmed by the dependence of FET cells upon autocrine TGFalpha for colony formation in experiments where the plating density was sufficiently low to generate a lag phase in tissue culture. In contrast, TGFalpha autocrine activity was not required for exponential phase cells, as evidenced by the failure of TGFalpha neutralizing antibody to inhibit proliferation in this growth state. Taken together, these results suggest that autocrine TGFalpha acts primarily in the process of growth initiation by moving cells from a noncycling state back into the cell cycle, rather than supporting cell growth already initiated.


Subject(s)
Autocrine Communication/physiology , Carcinoma/pathology , Colonic Neoplasms/pathology , Transforming Growth Factor alpha/physiology , Cell Cycle/physiology , Cell Division/physiology , DNA/biosynthesis , G1 Phase/physiology , Humans , Transcription, Genetic/physiology , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism , Tumor Cells, Cultured
16.
J Biol Chem ; 273(47): 31471-9, 1998 Nov 20.
Article in English | MEDLINE | ID: mdl-9813060

ABSTRACT

CBS human colon carcinoma cells are poorly tumorigenic in athymic nude mice, whereas FET colon carcinoma cells are non-tumorigenic. Both cell lines have well differentiated properties in tissue culture. Transforming growth factor alpha (TGF-alpha) was ectopically expressed by stable transfection of a TGF-alpha cDNA under repressible tetracycline control. The TGF-alpha-transfected cells showed enhanced clonal initiation and shortened lag phase growth in tissue culture without an alteration in doubling time in exponential phase relative to untransfected cells. Furthermore, the TGF-alpha transfectants showed increased independence from exogenous growth factors in clonal growth assays and induction of DNA synthesis after release from quiescence. Growth factor independence was associated with sustained epidermal growth factor receptor activation in quiescent TGF-alpha-transfected cells and the requirement of exogenous insulin for stimulation of quiescent cells to re-enter the cell cycle. Higher cloning, reduced lag time in tissue, and the acquisition of growth factor independence for DNA synthesis without a change in doubling time of TGF-alpha-transfected cells indicate that autocrine TGF-alpha functions by facilitating re-entry into the cell cycle from sub-optimal growth states rather than promoting or controlling the proliferation of actively cycling cells. The modulation of growth regulation by autocrine TGF-alpha was associated with increased malignant properties as TGF-alpha transfectants showed increased tumorigenicity in athymic nude mice. The administration of tetracycline reversed the effects of TGF-alpha expression in these cells both in vivo and in vitro, indicating that the alterations of the biological properties were due to the expression of TGF-alpha. Since these cells are continuously grown in a completely chemically defined medium without serum supplementation, it was possible to assign the mechanism underlying the generation of growth factor independence to the replacement of a requirement for exogenous insulin in parental cells by autocrine TGF-alpha.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Autocrine Communication , Carcinoma/metabolism , Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Transforming Growth Factor alpha/metabolism , Animals , Cell Adhesion , Cell Cycle , ErbB Receptors/metabolism , Humans , Mice , Mice, Nude , Mitogens/genetics , Mitogens/metabolism , Neoplasms, Experimental , Proteins/metabolism , Recombinant Proteins/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transfection , Transforming Growth Factor alpha/genetics , Tumor Cells, Cultured
17.
J Cell Physiol ; 176(2): 424-34, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9648930

ABSTRACT

To analyze transforming growth factor-beta (TGF-beta) response during MCF-7 cell progression, early passage (MCF-7E, < 200 passage) and late passage (MCF-7L, > 500 passage) cells were compared. MCF-7E cells showed an IC50 of approximately 10 ng/ml of TGF-beta1, whereas MCF-7L cells were insensitive. MCF-7E cells contained approximately threefold higher levels of TGF-beta receptor type II (TbetaRII) mRNA than MCF-7L, but their TbetaRI levels were similar. MCF-7E parental cells showed higher TbetaRII promoter activity than MCF-7L cells, which could be attributed to changes in Sp1 nuclear protein levels. Receptor cross-linking studies indicated that the cell surface receptor levels parallel mRNA levels in both cell lines. Limiting dilution clones of MCF-7E cells were established to determine the heterogeneity of TbetaRII expression in this cell line, and they showed varying degrees of TbetaRII expression. Fibronectin was induced at higher levels in cells expressing higher TbetaRII levels. All three TGF-beta isoforms were detected in limiting dilution clones and parental cells, but TGF-beta1 was more abundant relative to TGF-beta2 or 3, and no correlation between TGF-beta isoform profile with TGF-beta sensitivity was found. MCF-7L cells were tumorigenic and formed xenografts rapidly and progressively, whereas MCF-7E parental and limiting dilution clonal cells showed transient tumor formation followed by regression. These results indicate that decreased TbetaRII transcription in breast cancer cells leads to a loss of TbetaRII expression, resulting in cellular resistance to TGF-beta which contributes to escape from negative growth regulation and tumor progression.


Subject(s)
Adenocarcinoma/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/physiology , Receptors, Transforming Growth Factor beta/genetics , Adenocarcinoma/chemistry , Animals , Breast Neoplasms/chemistry , Carcinogenicity Tests , Female , Fibronectins/genetics , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/physiology
18.
J Biol Chem ; 273(26): 16527-34, 1998 Jun 26.
Article in English | MEDLINE | ID: mdl-9632722

ABSTRACT

Previous studies suggest that estrogen receptor-positive (ER+) breast cancer cells acquire resistance to transforming growth factor-beta (TGF-beta) because of reduced expression levels of TGF-beta receptor type II (RII). We now report that treatment of ER+ breast cancer cells with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-aza-2'-dC) leads to accumulation of RII transcript and protein in three different cell lines. RII induction restored TGF-beta response in MCF-7L breast cancer cells as indicated by the enhanced activity of a TGF-beta responsive promoter-reporter construct (p3TP-Lux). A transiently transfected RII promoter-reporter element (RII-chloramphenicol acetyltransferase) showed an increase in activity in the 5-aza-2'-dC-treated MCF-7L cells compared with untreated cells, suggesting the activation of a transactivator of RII transcription. Using electrophoretic mobility shift assays, the enhanced binding of proteins from 5-aza-2'-dC-treated MCF-7L nuclear extracts to radiolabeled Sp1 oligonucleotides was demonstrated. An RII promoter-chloramphenicol acetyltransferase construct containing a mutation in the Sp1 site was not expressed in the 5-aza-2'-dC-treated MCF-7L cells, further demonstrating that induction of Sp1 activity by 5-aza-2'-dC in the MCF-7L cells was critical to RII expression. Northern analysis indicated that 5-aza-2'-dC treatment did not affect the Sp1 transcript levels. Western blot analysis revealed an increase of Sp1 protein in the 5-aza-2'-dC-treated MCF-7L cells, but there was no change in the c-Jun levels. Studies after cyclohexamide treatment suggested an increase in the Sp1 protein stability from the 5-aza-2'-dC-treated MCF-7L extracts compared with untreated control extracts. These results indicate that the transcriptional repression of RII in the ER+ breast cancer cells is caused by suboptimal activity of Sp1, whereas treatment with 5-aza-2'-dC stabilizes the protein thus increasing steady-state Sp1 levels and thereby leads to enhanced RII transcription and subsequent restoration of TGF-beta sensitivity.


Subject(s)
Activin Receptors, Type I , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Breast Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Estrogen/analysis , Receptors, Transforming Growth Factor beta/biosynthesis , Sp1 Transcription Factor/metabolism , Transforming Growth Factor beta/metabolism , Azacitidine/pharmacology , Base Sequence , Breast Neoplasms/chemistry , Decitabine , Female , Humans , Insulin-Like Growth Factor II/genetics , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/biosynthesis , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Repressor Proteins/metabolism , Sp1 Transcription Factor/genetics , Tumor Cells, Cultured
19.
J Biol Chem ; 273(15): 9214-23, 1998 Apr 10.
Article in English | MEDLINE | ID: mdl-9535913

ABSTRACT

Autocrine transforming growth factor alpha (TGFalpha) is an important positive growth effector in malignant cells and plays a significant role in generating the growth factor-independent phenotype associated with malignant progression. However, the molecular mechanisms by which TGFalpha confers a growth advantage in progression is poorly understood. The highly tumorigenic cell line HCT116 up-regulates TGFalpha mRNA expression during growth arrest, whereas the poorly tumorigenic growth factor-dependent FET cell line down-regulates TGFalpha mRNA expression as it becomes quiescent. We have identified a 25-bp sequence at -201 to -225 within the TGFalpha promoter which mediates the differential regulation of TGFalpha expression during quiescence establishment in these two cell lines. This same sequence confers TGFalpha promoter responsiveness to exogenous growth factor or autocrine TGFalpha. The abberant upregulation of TGFalpha mRNA in quiescent HCT116 cells may allow them to return to the dividing state under more stringent conditions (nutrient replenishment alone) then quiescent FET cells (requires nutrients and growth factors). Antisense TGFalpha approaches showed that the dysregulated TGFalpha expression in quiescent HCT116 cells is a function of the strong TGFalpha autocrine loop (not inhibited by blocking antibodies) in these cells.


Subject(s)
Cell Cycle/physiology , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic , Transcription, Genetic , Transforming Growth Factor alpha/biosynthesis , Transforming Growth Factor alpha/genetics , Base Sequence , Cell Division , Cell Line , Chloramphenicol O-Acetyltransferase/biosynthesis , Clone Cells , Cloning, Molecular , Colonic Neoplasms , Epidermal Growth Factor/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genomic Library , Humans , Insulin/pharmacology , Kinetics , Leukocytes/metabolism , Molecular Sequence Data , Phenotype , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured
20.
J Biol Chem ; 273(13): 7749-56, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9516484

ABSTRACT

In view of the tumor suppressor role of the transforming growth factor-beta (TGFbeta) type II receptor (RII), the identification and characterization of agents that can induce the expression of this receptor are of potential importance to the development of chemoprevention approaches as well as treatment of cancer. To date, the identification of exogenous agents that control RII expression has been rare. We demonstrated that proliferation of MCF-7 early passage cells (MCF-7 E), which express RII and are sensitive to TGFbeta growth inhibition activity, was significantly inhibited by vitamin D3 and its analogue EB1089. In contrast, proliferation of MCF-7 late passage cells (MCF-7 L), which have lost cell surface RII and are resistant to TGFbeta, was not affected by these two compounds. TGFbeta-neutralizing antibody was able to block the inhibitory effect on MCF-7 E cells by these compounds, indicating that treatment induced autocrine-negative TGFbeta activity. An RNase protection assay showed approximately a 3-fold induction of the RII mRNA, while a receptor cross-linking assay revealed a 3-4-fold induction of the RII protein. In contrast, there was no change in either RII mRNA or protein in the MCF-7 L cells.


Subject(s)
Breast Neoplasms/metabolism , Cholecalciferol/analogs & derivatives , Gene Expression Regulation, Neoplastic/drug effects , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/biosynthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cholecalciferol/pharmacology , DNA Replication/drug effects , Female , Humans , Kinetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...