Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cells ; 12(20)2023 10 11.
Article in English | MEDLINE | ID: mdl-37887282

ABSTRACT

Oncogenic K-RAS mutations occur in approximately 25% of human lung cancers and are most frequently found in codon 12 (G12C, G12V, and G12D). Mutated K-RAS inhibitors have shown beneficial results in many patients; however, the inhibitors specifically target K-RASG12C and acquired resistance is a common occurrence. Therefore, new treatments targeting all kinds of oncogenic K-RAS mutations with a durable response are needed. RUNX3 acts as a pioneer factor of the restriction (R)-point, which is critical for the life and death of cells. RUNX3 is inactivated in most K-RAS-activated mouse and human lung cancers. Deletion of mouse lung Runx3 induces adenomas (ADs) and facilitates the development of K-Ras-activated adenocarcinomas (ADCs). In this study, conditional restoration of Runx3 in an established K-Ras-activated mouse lung cancer model regressed both ADs and ADCs and suppressed cancer recurrence, markedly increasing mouse survival. Runx3 restoration suppressed K-Ras-activated lung cancer mainly through Arf-p53 pathway-mediated apoptosis and partly through p53-independent inhibition of proliferation. This study provides in vivo evidence supporting RUNX3 as a therapeutic tool for the treatment of K-RAS-activated lung cancers with a durable response.


Subject(s)
Adenocarcinoma , Lung Neoplasms , Animals , Humans , Mice , Adenocarcinoma/pathology , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Genes, ras , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Recurrence, Local/genetics , Tumor Suppressor Protein p53/genetics
2.
Mol Cells ; 46(10): 592-610, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37706312

ABSTRACT

The Hippo kinase cascade functions as a central hub that relays input from the "outside world" of the cell and translates it into specific cellular responses by regulating the activity of Yes-associated protein 1 (YAP1). How Hippo translates input from the extracellular signals into specific intracellular responses remains unclear. Here, we show that transforming growth factor ß (TGFß)-activated TAK1 activates LATS1/2, which then phosphorylates YAP1. Phosphorylated YAP1 (p-YAP1) associates with RUNX3, but not with TEAD4, to form a TGFß-stimulated restriction (R)-point-associated complex which activates target chromatin loci in the nucleus. Soon after, p-YAP1 is exported to the cytoplasm. Attenuation of TGFß signaling results in re-localization of unphosphorylated YAP1 to the nucleus, where it forms a YAP1/TEAD4/SMAD3/AP1/p300 complex. The TGFß-stimulated spatiotemporal dynamics of YAP1 are abrogated in many cancer cells. These results identify a new pathway that integrates TGFß signals and the Hippo pathway (TGFß→TAK1→LATS1/2→YAP1 cascade) with a novel dynamic nuclear role for p-YAP1.


Subject(s)
Adaptor Proteins, Signal Transducing , Transforming Growth Factor beta , YAP-Signaling Proteins , Adaptor Proteins, Signal Transducing/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/physiology
3.
Cells ; 12(5)2023 02 23.
Article in English | MEDLINE | ID: mdl-36899846

ABSTRACT

A cell cycle is a series of events that takes place in a cell as it grows and divides. At the G1 phase of cell cycle, cells monitor their cumulative exposure to specific signals and make the critical decision to pass through the restriction (R)-point. The R-point decision-making machinery is fundamental to normal differentiation, apoptosis, and G1-S transition. Deregulation of this machinery is markedly associated with tumorigenesis. Therefore, identification of the molecular mechanisms that govern the R-point decision is one of the fundamental issues in tumor biology. RUNX3 is one of the genes frequently inactivated in tumors by epigenetic alterations. In particular, RUNX3 is downregulated in most K-RAS-activated human and mouse lung adenocarcinomas (ADCs). Targeted inactivation of Runx3 in the mouse lung induces adenomas (ADs), and markedly shortens the latency of ADC formation induced by oncogenic K-Ras. RUNX3 participates in the transient formation of R-point-associated activator (RPA-RX3-AC) complexes, which measure the duration of RAS signals and thereby protect cells against oncogenic RAS. This review focuses on the molecular mechanism by which the R-point participates in oncogenic surveillance.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Lung Neoplasms , Animals , Humans , Mice , Cell Transformation, Neoplastic , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Lung Neoplasms/genetics
4.
Cell Death Differ ; 28(4): 1251-1269, 2021 04.
Article in English | MEDLINE | ID: mdl-33116296

ABSTRACT

Inactivation of tumor suppressor Runt-related transcription factor 3 (RUNX3) plays an important role during early tumorigenesis. However, posttranslational modifications (PTM)-based mechanism for the inactivation of RUNX3 under hypoxia is still not fully understood. Here, we demonstrate a mechanism that G9a, lysine-specific methyltransferase (KMT), modulates RUNX3 through PTM under hypoxia. Hypoxia significantly increased G9a protein level and G9a interacted with RUNX3 Runt domain, which led to increased methylation of RUNX3 at K129 and K171. This methylation inactivated transactivation activity of RUNX3 by reducing interactions with CBFß and p300 cofactors, as well as reducing acetylation of RUNX3 by p300, which is involved in nucleocytoplasmic transport by importin-α1. G9a-mediated methylation of RUNX3 under hypoxia promotes cancer cell proliferation by increasing cell cycle or cell division, while suppresses immune response and apoptosis, thereby promoting tumor growth during early tumorigenesis. Our results demonstrate the molecular mechanism of RUNX3 inactivation by G9a-mediated methylation for cell proliferation and antiapoptosis under hypoxia, which can be a therapeutic or preventive target to control tumor growth during early tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Cell Hypoxia/genetics , Core Binding Factor Alpha 3 Subunit/genetics , DNA Methylation/genetics , Acetylation , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
5.
Mol Cells ; 43(10): 889-897, 2020 Oct 31.
Article in English | MEDLINE | ID: mdl-33115981

ABSTRACT

K-RAS is frequently mutated in human lung adenocarcinomas (ADCs), and the p53 pathway plays a central role in cellular defense against oncogenic K-RAS mutation. However, in mouse lung cancer models, oncogenic K-RAS mutation alone can induce ADCs without p53 mutation, and loss of p53 does not have a significant impact on early K-RAS-induced lung tumorigenesis. These results raise the question of how K-RAS-activated cells evade oncogene surveillance mechanisms and develop into lung ADCs. RUNX3 plays a key role at the restriction (R)-point, which governs multiple tumor suppressor pathways including the p14ARF-p53 pathway. In this study, we found that K-RAS activation in a very limited number of cells, alone or in combination with p53 inactivation, failed to induce any pathologic lesions for up to 1 year. By contrast, when Runx3 was inactivated and K-RAS was activated by the same targeting method, lung ADCs and other tumors were rapidly induced. In a urethane-induced mouse lung tumor model that recapitulates the features of K-RAS-driven human lung tumors, Runx3 was inactivated in both adenomas (ADs) and ADCs, whereas K-RAS was activated only in ADCs. Together, these results demonstrate that the R-point-associated oncogene surveillance mechanism is abrogated by Runx3 inactivation in AD cells and these cells cannot defend against K-RAS activation, resulting in the transition from AD to ADC. Therefore, K-RAS-activated lung epithelial cells do not evade oncogene surveillance mechanisms; instead, they are selected if they occur in AD cells in which Runx3 has been inactivated.


Subject(s)
Adenocarcinoma of Lung/pathology , Core Binding Factor Alpha 3 Subunit/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Urethane/adverse effects , Adenocarcinoma of Lung/chemically induced , Adenocarcinoma of Lung/genetics , Animals , Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Nat Commun ; 10(1): 1897, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015486

ABSTRACT

The cellular decision regarding whether to undergo proliferation or death is made at the restriction (R)-point, which is disrupted in nearly all tumors. The identity of the molecular mechanisms that govern the R-point decision is one of the fundamental issues in cell biology. We found that early after mitogenic stimulation, RUNX3 binds to its target loci, where it opens chromatin structure by sequential recruitment of Trithorax group proteins and cell-cycle regulators to drive cells to the R-point. Soon after, RUNX3 closes these loci by recruiting Polycomb repressor complexes, causing the cell to pass through the R-point toward S phase. If the RAS signal is constitutively activated, RUNX3 inhibits cell cycle progression by maintaining R-point-associated genes in an open structure. Our results identify RUNX3 as a pioneer factor for the R-point and reveal the molecular mechanisms by which appropriate chromatin modifiers are selectively recruited to target loci for appropriate R-point decisions.


Subject(s)
Cell Cycle Checkpoints/genetics , Chromatin/chemistry , Core Binding Factor Alpha 3 Subunit/genetics , Epithelial Cells/metabolism , Gene Expression Regulation , Animals , Butadienes/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/drug effects , Chromatin/metabolism , Chromatin Assembly and Disassembly/drug effects , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Imidazoles/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Nitriles/pharmacology , Piperazines/pharmacology , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/genetics , ras Proteins/metabolism
7.
Cancer Cell ; 24(5): 603-16, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24229708

ABSTRACT

Targeted inactivation of Runx3 in mouse lung induced mucinous and nonmucinous adenomas and markedly shortened latency of adenocarcinoma formation induced by oncogenic K-Ras. RUNX3 was frequently inactivated in K-RAS mutated human lung adenocarcinomas. A functional genetic screen of a fly mutant library and molecular analysis in cultured cell lines revealed that Runx3 forms a complex with BRD2 in a K-Ras-dependent manner in the early phase of the cell cycle; this complex induces expression of p14(ARF)/p19(Arf) and p21(WAF/CIP). When K-Ras was constitutively activated, the Runx3-BRD2 complex was stably maintained and expression of both p14(ARF) and p21(WAF/CIP) was prolonged. These results provide a missing link between oncogenic K-Ras and the p14(ARF)-p53 pathway, and may explain how cells defend against oncogenic K-Ras.


Subject(s)
Adenocarcinoma/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Lung Neoplasms/metabolism , ADP-Ribosylation Factors/metabolism , Acetylation , Adenocarcinoma of Lung , Alveolar Epithelial Cells/physiology , Animals , Carcinogenesis/metabolism , Cell Differentiation , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression , Gene Knockout Techniques , HEK293 Cells , Histone Deacetylases/metabolism , Humans , Mice , Mice, Transgenic , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Repressor Proteins/metabolism , Respiratory Mucosa/pathology , Transcription Factors , ras Proteins/genetics , ras Proteins/metabolism
8.
J Biol Chem ; 285(13): 10122-10129, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20100835

ABSTRACT

RUNX3 is a transcription factor that functions as a tumor suppressor. In some cancers, RUNX3 expression is down-regulated, usually due to promoter hypermethylation. Recently, it was found that RUNX3 can also be inactivated by the mislocalization of the protein in the cytoplasm. The molecular mechanisms controlling this mislocalization are poorly understood. In this study, we found that the overexpression of Src results in the tyrosine phosphorylation and cytoplasmic localization of RUNX3. We also found that the tyrosine residues of endogenous RUNX3 are phosphorylated and that the protein is localized in the cytoplasm in Src-activated cancer cell lines. We further showed that the knockdown of Src by small interfering RNA, or the inhibition of Src kinase activity by a chemical inhibitor, causes the re-localization of RUNX3 to the nucleus. Collectively, our results demonstrate that the tyrosine phosphorylation of RUNX3 by activated Src is associated with the cytoplasmic localization of RUNX3 in gastric and breast cancers.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation, Neoplastic , Tyrosine/chemistry , src-Family Kinases/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , HeLa Cells , Humans , Phosphorylation , Protein Transport , RNA, Small Interfering/metabolism , Stomach Neoplasms/metabolism , Tyrosine/genetics , Tyrosine/metabolism
9.
Cancer Res ; 69(20): 8111-9, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19808967

ABSTRACT

The p14(ARF)-MDM2-p53 pathway constitutes an effective mechanism for protecting cells from oncogenic stimuli such as activated Ras and Myc. Importantly, Ras activation induces p14(ARF) and often occurs earlier than p53 inactivation during cancer development. Here, we show that RUNX3, a tumor suppressor in various tumors including stomach, bladder, colon, and lung, is stabilized by Ras activation through the p14(ARF)-MDM2 signaling pathway. RUNX3 directly binds MDM2 through its Runt-related DNA-binding domain. MDM2 blocks RUNX3 transcriptional activity by interacting with RUNX3 through an acidic domain adjacent to the p53-binding domain of MDM2 and ubiquitinates RUNX3 on key lysine residues to mediate nuclear export and proteasomal degradation. Our data indicate that the lineage-specific tumor suppressor RUNX3 and the ubiquitous p53 protein are both principal responders of the p14(ARF)-MDM2 cell surveillance pathway that prevents pathologic consequences of abnormal oncogene activation.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Ubiquitins/metabolism , Apoptosis , Blotting, Western , Cell Transformation, Neoplastic , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Mutation/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Small Interfering/pharmacology , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitination , ras Proteins/physiology
10.
J Cell Biochem ; 107(3): 557-65, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19350572

ABSTRACT

Runt-related (RUNX) transcription factors play pivotal roles in neoplastic development and have tissue-specific developmental roles in hematopoiesis (RUNX1), osteogenesis (RUNX2), as well as neurogenesis and thymopoiesis (RUNX3). RUNX3 is a tumor suppressor in gastric carcinoma, and its expression is frequently inactivated by DNA methylation or its protein mislocalized in many cancer types, including gastric and breast cancer. Jun-activation domain-binding protein 1 (Jab1/CSN5), a component of the COP9 signalosome (CSN), is critical for nuclear export and the degradation of several tumor suppressor proteins, including p53, p27(Kip1), and Smad4. Here, we find that Jab1 facilitates nuclear export of RUNX3 that is controlled by CSN-associated kinases. RUNX3 sequestered in the cytoplasm is rapidly degraded through a proteasome-mediated pathway. Our results identify a novel mechanism of regulating nuclear export and protein stability of RUNX3 by the CSN complex.


Subject(s)
Core Binding Factor Alpha 3 Subunit/analysis , Core Binding Factor Alpha 3 Subunit/metabolism , Cytoplasm/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Hydrolases/metabolism , Active Transport, Cell Nucleus/physiology , COP9 Signalosome Complex , Cell Nucleus/metabolism , Cells, Cultured , HeLa Cells , Humans , Transcription, Genetic , Transfection
11.
J Cell Biochem ; 105(1): 236-44, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18570183

ABSTRACT

The adenoviral gene, termed early region 1A (E1A), is crucial for transformation and has been used very effectively as a tool to determine the molecular mechanisms that underlie the basis of cellular transformation. pRb, p107, p130, p300/CBP, p400, TRRAP, and CtBP were identified to be E1A-binding proteins and their roles in cellular transformation have been established. Although the major function of E1A is considered to be the regulation of gene expression that is critical for differentiation and cell cycle exit, one of the most significant questions relating to E1A transformation is how E1A mediates this regulation. RUNX3 is a transcription factor that was first described as a gastric cancer tumor suppressor but is now known to be involved in many different cancers. Exogenous expression of RUNX3 strongly inhibits the growth of cells. Here, we show that the adenovirus oncoprotein E1A interacts with RUNX3 in vitro and in vivo. RUNX3 interacts with the N-terminus (amino acids 2-29) of E1A, which is known to interact with p300/CBP, p400, and TRRAP. E1A interacts directly with the Runt domain of RUNX3 but does not interfere with CBFbeta-RUNX3 interactions. In addition, E1A inhibits the transactivation activity of RUNX3 on the p21(WAF1/CIP1) promoter. Consistent with these observations, the growth inhibition induced by RUNX3 is reduced by E1A. These results demonstrate that E1A specifically binds to RUNX3 and inactivates its transactivation activity. We propose that one of the mechanisms for the oncogenic activity of E1A is the inhibition of RUNX3, similar to that of RB and p300/CBP.


Subject(s)
Adenovirus E1A Proteins/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Transcriptional Activation/genetics , Adenovirus E1A Proteins/genetics , CCAAT-Binding Factor/metabolism , Cell Line , Core Binding Factor Alpha 3 Subunit/genetics , Humans , Protein Binding
12.
Mol Cell Biol ; 26(12): 4474-88, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16738314

ABSTRACT

Genes involved in the transforming growth factor beta (TGF-beta) signaling pathway are frequently altered in several types of cancers, and a gastric tumor suppressor RUNX3 appears to be an integral component of this pathway. We reported previously that apoptosis is notably reduced in Runx3-/- gastric epithelial cells. In the present study, we show that a proapoptotic gene Bim was transcriptionally activated by RUNX3 in the gastric cancer cell lines SNU16 and SNU719 treated with TGF-beta. The human Bim promoter contains RUNX sites, which are required for its activation. Furthermore, a dominant negative form of RUNX3 comprised of amino acids 1 to 187 increased tumorigenicity of SNU16 by inhibiting Bim expression. In Runx3-/- mouse gastric epithelium, Bim was down-regulated, and apoptosis was reduced to the same extent as that in Bim-/- gastric epithelium. We confirmed comparable expression of TGF-beta1 and TGF-beta receptors between wild-type and Runx3-/- gastric epithelia and reduction of Bim in TGF-beta1-/- stomach. These results demonstrate that RUNX3 is responsible for transcriptional up-regulation of Bim in TGF-beta-induced apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Base Sequence , Bcl-2-Like Protein 11 , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/deficiency , Core Binding Factor Alpha 3 Subunit/genetics , DNA/genetics , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gastric Mucosa/drug effects , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Smad Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Up-Regulation
13.
Mol Cell Biol ; 25(18): 8097-107, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16135801

ABSTRACT

RUNX3 has been suggested to be a tumor suppressor of gastric cancer. The gastric mucosa of the Runx3-null mouse develops hyperplasia due to enhanced proliferation and suppressed apoptosis accompanied by a decreased sensitivity to transforming growth factor beta1 (TGF-beta1). It is known that TGF-beta1 induces cell growth arrest by activating CDKN1A (p21(WAF1)(/Cip1)), which encodes a cyclin-dependent kinase inhibitor, and this signaling cascade is considered to be a tumor suppressor pathway. However, the lineage-specific transcription factor that cooperates with SMADs to induce p21 expression is not known. Here we show that RUNX3 is required for the TGF-beta-dependent induction of p21 expression in stomach epithelial cells. Overexpression of RUNX3 potentiates TGF-beta-dependent endogenous p21 induction. In cooperation with SMADs, RUNX3 synergistically activates the p21 promoter. In contrast, RUNX3-R122C, a mutation identified in a gastric cancer patient, abolished the ability to activate the p21 promoter or cooperate with SMADs. Furthermore, areas in mouse and human gastric epithelium where RUNX3 is expressed coincided with those where p21 is expressed. Our results suggest that at least part of the tumor suppressor activity of RUNX3 is associated with its ability to induce p21 expression.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , Gastric Mucosa/growth & development , Stomach Neoplasms/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/metabolism , Animals , Core Binding Factor Alpha 3 Subunit , Cyclin-Dependent Kinase Inhibitor p21 , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Gastric Mucosa/chemistry , Gastric Mucosa/drug effects , Gene Expression/drug effects , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice , Mutation , Promoter Regions, Genetic/drug effects , Smad Proteins , Stomach Neoplasms/genetics , Transcription Factors/analysis , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/genetics , Up-Regulation
14.
J Biol Chem ; 280(42): 35579-87, 2005 Oct 21.
Article in English | MEDLINE | ID: mdl-16115867

ABSTRACT

Two major isoforms of the Runx2 gene are expressed by alternative promoter usage: Runx2 type I (Runx2-I) is derived from the proximal promoter (P2), and Runx2 type II (Runx2-II) is produced by the distal promoter (P1). Our previous results indicate that Dlx5 mediates BMP-2-induced Runx2 expression and osteoblast differentiation (Lee, M.-H., Kim, Y-J., Kim, H-J., Park, H-D., Kang, A-R., Kyung, H.-M., Sung, J-H., Wozney, J. M., Kim, H-J., and Ryoo, H-M. (2003) J. Biol. Chem. 278, 34387-34394). However, little is known of the molecular mechanisms by which Dlx5 up-regulates Runx2 expression in BMP-2 signaling. Here, Runx2-II expression was found to be specifically stimulated by BMP-2 treatment or by Dlx5 overexpression. In addition, BMP-2, Dlx5, and Runx2-II were found to be expressed in osteogenic fronts and parietal bones of the developing cranial vault and Runx2-I and Msx2 in the sutural mesenchyme. Furthermore, Runx2 P1 promoter activity was strongly stimulated by Dlx5 overexpression, whereas Runx2 P2 promoter activity was not. Runx2 P1 promoter deletion analysis indicated that the Dlx5-specific response is due to sequences between -756 and -342 bp of the P1 promoter, where three Dlx5-response elements are located. Dlx5 responsiveness to these elements was confirmed by gel mobility shift assay and site-directed mutagenesis. Moreover, Msx2 specifically suppressed the Runx2 P1 promoter, and the responsible region overlaps with that recognized by Dlx5. In summary, Dlx5 specifically transactivates the Runx2 P1 promoter, and its action on the P1 promoter is antagonized by Msx2.


Subject(s)
Core Binding Factor Alpha 1 Subunit/biosynthesis , Homeodomain Proteins/physiology , Promoter Regions, Genetic , Transcription Factors/physiology , Animals , Base Sequence , Binding Sites , Blotting, Northern , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA/chemistry , DNA Primers/chemistry , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization , Luciferases/metabolism , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotides/chemistry , Osteoblasts/cytology , Osteoblasts/metabolism , Polymerase Chain Reaction , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemistry , Response Elements , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Transforming Growth Factor beta/metabolism , Up-Regulation
15.
Cell ; 109(1): 113-24, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11955451

ABSTRACT

Runx3/Pebp2alphaC null mouse gastric mucosa exhibits hyperplasias due to stimulated proliferation and suppressed apoptosis in epithelial cells, and the cells are resistant to growth-inhibitory and apoptosis-inducing action of TGF-beta, indicating that Runx3 is a major growth regulator of gastric epithelial cells. Between 45% and 60% of human gastric cancer cells do not significantly express RUNX3 due to hemizygous deletion and hypermethylation of the RUNX3 promoter region. Tumorigenicity of human gastric cancer cell lines in nude mice was inversely related to their level of RUNX3 expression, and a mutation (R122C) occurring within the conserved Runt domain abolished the tumor-suppressive effect of RUNX3, suggesting that a lack of RUNX3 function is causally related to the genesis and progression of human gastric cancer.


Subject(s)
Cell Division/genetics , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/deficiency , Epithelium/metabolism , Gastric Mucosa/metabolism , Stomach Neoplasms/genetics , Transcription Factors/deficiency , Tumor Suppressor Proteins/deficiency , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Division/drug effects , Cell Transformation, Neoplastic/drug effects , Core Binding Factor Alpha 3 Subunit , DNA Methylation , DNA-Binding Proteins/genetics , Epithelium/pathology , Epithelium/physiopathology , Exons/genetics , Female , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Targeting , Humans , Hyperplasia/genetics , Hyperplasia/pathology , Hyperplasia/physiopathology , Male , Mice , Mice, Knockout , Protein Structure, Tertiary/genetics , Stomach/pathology , Stomach/physiopathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/physiopathology , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...