Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Oncogene ; 36(50): 6884-6894, 2017 12 14.
Article in English | MEDLINE | ID: mdl-28846108

ABSTRACT

The restriction (R)-point decision is fundamental to normal differentiation and the G1-S transition, and the decision-making machinery is perturbed in nearly all cancer cells. The mechanisms underlying the cellular context-dependent R-point decision remain poorly understood. We found that the R-point was dysregulated in Runx3-/-mouse embryonic fibroblasts (MEFs), which formed tumors in nude mice. Ectopic expression of Runx3 restored the R-point and abolished the tumorigenicity of Runx3-/-MEFs and K-Ras-activated Runx3-/-MEFs (Runx3-/-;K-RasG12D/+). During the R-point, Runx3 transiently formed a complex with pRb and Brd2 and induced Cdkn1a (p21Waf1/Cip1/Sdi1; p21), a key regulator of the R-point transition. Cyclin D-CDK4/6 promoted dissociation of the pRb-Runx3-Brd2 complex, thus turning off p21 expression. However, cells harboring oncogenic K-Ras maintained the pRb-Runx3-Brd2 complex and p21 expression even after introduction of Cyclin D1. Thus, Runx3 plays a critical role in R-point regulation and defense against cellular transformation.


Subject(s)
Cell Transformation, Neoplastic , Core Binding Factor Alpha 3 Subunit/physiology , Animals , Carcinogenesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Genes, ras , HEK293 Cells , Humans , Mice , Protein Serine-Threonine Kinases/physiology , Retinoblastoma Protein/physiology , Transcription Factors
2.
Oncogene ; 36(7): 999-1011, 2017 02 16.
Article in English | MEDLINE | ID: mdl-27425596

ABSTRACT

The tumor-suppressor RUNX3 has a critical role in a lineage determination, cell cycle arrest and apoptosis. Lozenge (Lz), a Drosophila homolog of mammalian RUNX family members, has integral roles in these processes and specifically in eye cell fate determination. To elucidate the genetic modifiers of Lz/RUNX3, we performed a large-scale functional screen in a fly mutant library. The screen revealed genetic interactions between the Lz, Rac and Hippo pathways. Analysis of interactions among these genes revealed that the defective phenotype resulting from activation of Yki, an end point effector of the Hippo pathway, was suppressed by Lz and enhanced by Rac-Trio. Molecular biological analysis using mammalian homologs reveled that LATS1/2-mediated YAP phosphorylation-facilitated dissociation of the YAP-TEAD4 complex and association of the YAP-RUNX3 complex. When cells were stimulated to proliferate, activated RAC-TRIO signaling inhibited LATS1/2-mediated YAP phosphorylation; consequently, YAP dissociated from RUNX3 and associated with TEAD, thereby replacing the YAP-RUNX3 complex with YAP-TEAD. RUNX3 contributed to both association and dissociation of YAP-TEAD complex, most likely through the formation of the YAP-TEAD-RUNX3 ternary complex. Ectopic expression of RUNX3 in MKN28 gastric cancer cells reduced tumorigenicity, and the tumor-suppressive activity of RUNX3 was associated with its ability to interact with YAP. These results identify a novel regulatory mechanism, mediated by the Hippo and RAC-TRIO pathways, that changes the binding partner of YAP.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/genetics , Muscle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/pathology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Biomarkers, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Proliferation , Core Binding Factor Alpha 3 Subunit/genetics , DNA-Binding Proteins/genetics , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Male , Mice , Mice, Nude , Muscle Proteins/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , TEA Domain Transcription Factors , Trans-Activators , Transcription Factors/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
3.
Oncogenesis ; 3: e101, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24777122

ABSTRACT

Transcription factors of the RUNX family (RUNXs), which play pivotal roles in normal development and neoplasia, are regulated by various post-translational modifications. To understand the molecular mechanisms underlying the regulation of RUNXs, we performed a large-scale functional genetic screen of a fly mutant library. The screen identified dPias (the fly ortholog of mammalian PIASs), an E3 ligase for the SUMO (small ubiquitin-like modifier) modification, as a novel genetic modifier of lz (the fly ortholog of mammalian RUNX3). Molecular biological analysis revealed that lz/RUNXs are sumoylated by dPias/PIAS1 at an evolutionarily conserved lysine residue (K372 of lz, K144 of RUNX1, K181 of RUNX2 and K148 of RUNX3). PIAS1-mediated sumoylation inhibited RUNX3 transactivation activity, and this modification was promoted by the AKT1 kinase. Importantly, PIAS1 failed to sumoylate some RUNX1 mutants associated with breast cancer. In nude mice, tumorigenicity was promoted by RUNX3 bearing a mutation in the sumoylation site, but suppressed by wild-type RUNX3. Our results suggest that RUNXs are sumoylated by PIAS1, and that this modification could play a critical role in the regulation of the tumor-suppressive activity of these proteins.

4.
Oncogene ; 29(23): 3349-61, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20228843

ABSTRACT

Human lung adenocarcinoma, the most prevalent form of lung cancer, is characterized by many molecular abnormalities. K-ras mutations are associated with the initiation of lung adenocarcinomas, but K-ras-independent mechanisms may also initiate lung tumors. Here, we find that the runt-related transcription factor Runx3 is essential for normal murine lung development and is a tumor suppressor that prevents lung adenocarcinoma. Runx3-/- mice, which die soon after birth, exhibit alveolar hyperplasia. Importantly, Runx3-/- bronchioli exhibit impaired differentiation, as evidenced by the accumulation of epithelial cells containing specific markers for both alveolar (that is SP-B) and bronchiolar (that is CC10) lineages. Runx3-/- epithelial cells also express Bmi1, which supports self-renewal of stem cells. Lung adenomas spontaneously develop in aging Runx3+/- mice ( approximately 18 months after birth) and invariably exhibit reduced levels of Runx3. As K-ras mutations are very rare in these adenomas, Runx3+/- mice provide an animal model for lung tumorigenesis that recapitulates the preneoplastic stage of human lung adenocarcinoma development, which is independent of K-Ras mutation. We conclude that Runx3 is essential for lung epithelial cell differentiation, and that downregulation of Runx3 is causally linked to the preneoplastic stage of lung adenocarcinoma.


Subject(s)
Core Binding Factor Alpha 3 Subunit/physiology , Lung Neoplasms/prevention & control , Lung/cytology , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Animals , Cell Differentiation , Cell Proliferation , Core Binding Factor Alpha 3 Subunit/deficiency , Core Binding Factor Alpha 3 Subunit/genetics , Epithelial Cells/cytology , Humans , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Nuclear Proteins/analysis , Nuclear Proteins/physiology , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Pulmonary Surfactant-Associated Protein B/analysis , Repressor Proteins/analysis , Repressor Proteins/physiology , Urethane/toxicity , Uteroglobin/analysis
5.
Mol Cell Biol ; 20(23): 8783-92, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11073979

ABSTRACT

When C2C12 pluripotent mesenchymal precursor cells are treated with transforming growth factor beta1 (TGF-beta1), terminal differentiation into myotubes is blocked. Treatment with bone morphogenetic protein 2 (BMP-2) not only blocks myogenic differentiation of C2C12 cells but also induces osteoblast differentiation. The molecular mechanisms governing the ability of TGF-beta1 and BMP-2 to both induce ligand-specific responses and inhibit myogenic differentiation are not known. We identified Runx2/PEBP2alphaA/Cbfa1, a global regulator of osteogenesis, as a major TGF-beta1-responsive element binding protein induced by TGF-beta1 and BMP-2 in C2C12 cells. Consistent with the observation that Runx2 can be induced by either TGF-beta1 or BMP-2, the exogenous expression of Runx2 mediated some of the effects of TGF-beta1 and BMP-2 but not osteoblast-specific gene expression. Runx2 mimicked common effects of TGF-beta1 and BMP-2 by inducing expression of matrix gene products (for example, collagen and fibronectin), suppressing MyoD expression, and inhibiting myotube formation of C2C12 cells. For osteoblast differentiation, an additional effector, BMP-specific Smad protein, was required. Our results indicate that Runx2 is a major target gene shared by TGF-beta and BMP signaling pathways and that the coordinated action of Runx2 and BMP-activated Smads leads to the induction of osteoblast-specific gene expression in C2C12 cells.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , DNA-Binding Proteins/metabolism , Mesoderm/cytology , Neoplasm Proteins , Osteoblasts/cytology , Phosphoproteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Bone Morphogenetic Protein 2 , Cell Differentiation , Cell Line , Core Binding Factor Alpha 1 Subunit , Gene Expression Regulation, Developmental , Mesoderm/drug effects , Mice , Models, Biological , Osteogenesis/physiology , Protein Binding , Response Elements , Smad5 Protein , Stem Cells/cytology , Stem Cells/drug effects , Transforming Growth Factor beta1
6.
Mol Cells ; 10(1): 90-5, 2000 Feb 29.
Article in English | MEDLINE | ID: mdl-10774753

ABSTRACT

The chromosomal translocation t(12;21) (p12;q22) which results in the TEL-AML1 fusion gene is the most frequent genetic rearrangement in childhood B-lineage acute lymphoblastic leukemia (ALL). The rearrangement in this locus, however, is only rarely observed by routine karyotypic analysis. We established a nested-reverse transcriptase-polymerase chain reaction (nested-RT-PCR) technique for the detection of the TEL-AML1 transcript, and used this to investigate the incidence of the rearrangement, and to characterize the disease present in TEL-AML1-positive B-lineage ALL patients. The TEL-AML1 fusion transcript was detected in nine of fourteen patients. These patients were relatively homogeneous in that they were young and had low presenting leukocyte counts, both features of which are associated with a favorable prognosis. Furthermore, we could detect the TEL-AML1 transcript in the peripheral blood of t(12;21)-positive patients and we used this to assess minimal residual disease (MRD) in patients during chemotherapy. The data demonstrate that nested-RT-PCR is a suitable tool for diagnosing t(12;21)-positive ALL, that these patients constitute a clinically distinct subgroup of ALL patients, and that the method could also be used to monitor MRD in these patients.


Subject(s)
Leukemia, B-Cell/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit , Cytogenetic Analysis , Female , Humans , Karyotyping , Leukemia, B-Cell/diagnosis , Male , Neoplasm, Residual/diagnosis , Neoplasm, Residual/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , RNA, Neoplasm/analysis , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Survival Analysis , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL