Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 19(23): 3303-3316, 2020 12.
Article in English | MEDLINE | ID: mdl-33131417

ABSTRACT

Keloid is an extremely common and often overlooked benign neoplastic disease, but its consequences should not be underestimated. Therefore, a deep exploration of the pathological mechanism of keloid becomes very essential. After 22 samples were collected from each patient's keloid tissues and normal skin tissues, circ_0008450 and Runx3 expression was tested by qRT-PCR. When primary human keratinized epithelial cells were transfected by sh-circ_0008450 or sh-Runx3, cell proliferation, apoptosis, migration, and EMT process were assessed by CCK-8, BrdU assay, apoptosis assay, migration assay, and Western blot. Finally, transfection was performed to explore the effect of circ_0008450 on the TGF-ß/Smad signal pathway by adopting western blot. Circ_0008450 was highly expressed in keratinized epithelial tissues. After the transfection of sh-circ_0008450 into primary human keratinized epithelial cells, cell proliferation, migration, and EMT process were inhibited, and apoptosis was stimulated. Moreover, circ_0008450 silence-induced above changes were partly reversed by transfecting sh-Runx3. In addition, transfecting sh-circ_0008450 could repress TGF-ß/Smad pathway, while transfecting sh-Runx3 activated the above pathway. Circ_0008450 down-regulated Runx3 to promote the proliferation and EMT process of human keratinized epithelial cells. This discovery may be related to the activation of the TGF-ß/Smad pathway.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , RNA, Circular/antagonists & inhibitors , Adolescent , Adult , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Down-Regulation/physiology , Female , Humans , Keratinocytes/metabolism , Male , Middle Aged , RNA, Circular/genetics , Young Adult
2.
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
3.
Mol Med Rep ; 19(5): 3933-3940, 2019 May.
Article in English | MEDLINE | ID: mdl-30816536

ABSTRACT

Dysregulated microRNAs (miRNAs/miRs) directly modulate the biological functions of non­small cell lung cancer (NSCLC) cells and contribute to the initiation and progression of NSCLC; however, the specific roles and underlying mechanisms of the dysregulated miRNAs in NSCLC require further investigation. The present study reported that miRNA­629­5p (miR­629) was upregulated in NSCLC tissues and cell lines. High miR­629 expression levels were significantly associated with tumour size, clinical stage and lymph node metastasis in patients with NSCLC. Functional experiments indicated that miR­629 inhibition suppressed the viability and invasion NSCLC cells in vitro. Furthermore, bioinformatics prediction, luciferase reporter assay, reverse transcription­quantitative polymerase chain reaction and western blot analysis demonstrated that runt­related transcription factor 3 (RUNX3) was a direct target gene of miR­629 in NSCLC. Restoration of RUNX3 expression suppressed the effects of miR­629 inhibition in NSCLC cells. Rescue experiments revealed that RUNX3 knockdown partially abrogated the effects of miR­629 inhibition on NSCLC cells. In summary, miR­629 directly targeted RUNX3 to inhibit the progression of NSCLC, suggesting that this miRNA may be considered as a diagnostic and therapeutic target for patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Core Binding Factor Alpha 3 Subunit/metabolism , Lung Neoplasms/pathology , MicroRNAs/metabolism , 3' Untranslated Regions , Adult , Aged , Antagomirs/metabolism , Cell Line , Cell Movement , Cell Survival , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Down-Regulation , Female , Humans , Lymphatic Metastasis , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , RNA Interference , RNA, Small Interfering/metabolism
4.
Front Immunol ; 9: 3132, 2018.
Article in English | MEDLINE | ID: mdl-30687330

ABSTRACT

Susceptibility to ankylosing spondylitis (AS) is polygenic with more than 100 genes identified to date. These include HLA-B27 and the aminopeptidases (ERAP1, ERAP2, and LNPEPS), which are involved in antigen processing and presentation to T-cells, and several genes (IL23R, IL6R, STAT3, JAK2, IL1R1/2, IL12B, and IL7R) involved in IL23 driven pathways of inflammation. AS is also strongly associated with polymorphisms in two transcription factors, RUNX3 and T-bet (encoded by TBX21), which are important in T-cell development and function. The influence of these genes on the pathogenesis of AS and their potential for identifying drug targets is discussed here.


Subject(s)
Core Binding Factor Alpha 3 Subunit/genetics , Gene Expression Regulation/immunology , Interleukin-23/metabolism , Spondylitis, Ankylosing/immunology , T-Box Domain Proteins/genetics , Aminopeptidases/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation/drug effects , HLA-B27 Antigen/genetics , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Interleukin-23/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Molecular Targeted Therapy/methods , Polymorphism, Single Nucleotide , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism , Spondylitis, Ankylosing/genetics , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/metabolism
5.
J Cell Mol Med ; 21(12): 3529-3539, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28699703

ABSTRACT

Irreversible pulmonary fibrosis induced by paraquat (PQ) poisoning is the major cause of death in patients with PQ poisoning. The epithelial-mesenchymal transition (EMT) is postulated to be one of the main mechanisms of pulmonary fibrosis. Here, we investigated the role of miR-210 in PQ-induced EMT and its relationship with hypoxia-inducible factor-1α (HIF-1α). Western blotting, immunofluorescence, immunoprecipitation and other methods were used in this study. We found that miR-210 expression was significantly increased after PQ poisoning, and it may be regulated by HIF-1α. Overexpression of miR-210 further increased the HIF-1α protein level and promoted EMT. Moreover, miR-210 knock-down reduced the HIF-1α protein level and decreased the degree of EMT. Runt-related transcription factor-3 (RUNX3), a direct target of miR-210, was inhibited by miR-210 in response to PQ poisoning. RUNX3 increased the hydroxylation ability of prolyl hydroxylase domain-containing protein 2 (PHD2), a key enzyme that promotes HIF-1α degradation. PHD2 immunoprecipitated with RUNX3 and its level changed similarly to that of RUNX3. The expression of the HIF-1α protein was significantly reduced when RUNX3 was overexpressed. HIF-1α protein levels were markedly increased when RUNX3 was silenced. Based on these results, a positive feedback loop may exist between miR-210 and HIF-1α. The mechanism may function through miR-210-mediated repression of RUNX3, which further decreases the hydroxylation activity of PHD2, enhances the stability of HIF-1α, and promotes PQ-induced EMT, aggravating the progression of pulmonary fibrosis. This study further elucidates the mechanism of PQ-induced pulmonary fibrosis and may provide a new perspective for the future development of therapies.


Subject(s)
Core Binding Factor Alpha 3 Subunit/genetics , Epithelial-Mesenchymal Transition/drug effects , Feedback, Physiological , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MicroRNAs/genetics , Paraquat/poisoning , Pulmonary Fibrosis/genetics , A549 Cells , Animals , Cell Line , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/metabolism , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Male , MicroRNAs/metabolism , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Protein Stability , Proteolysis , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
6.
Mol Med Rep ; 16(2): 2113-2120, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28656235

ABSTRACT

Lung cancer is the primary cause of cancer­associated mortality in men and women worldwide. Increasing evidence indicates that abnormal microRNA (miRNA) expression contributes to the carcinogenesis and progression of multiple human cancers, including non­small cell lung cancer (NSCLC). Therefore, miRNAs exhibit the potential to act as biomarkers for the diagnosis, treatment and prognosis of human malignancies. miRNA­661 (miR­661) has previously been demonstrated to be important in the development of various human cancer types. However, the expression levels, functions and underlying mechanisms of miR­661 in NSCLC remain to be elucidated. The present study demonstrated that miR­661 was upregulated in NSCLC tissues and cell lines. In addition, miR­661 expression levels were significantly correlated with differentiation and tumor stage lymph node metastasis of NSCLC patients. Functional experiments demonstrated that miR-661 downregulation inhibited NSCLC cell proliferation and invasion in vitro. Furthermore, runt­related transcription factor 3 (RUNX3) was identified as a direct target of miR­661 in NSCLC. RUNX3 was expressed at a low level in NSCLC tissues and was negatively correlated with the miR­661 expression level. Further experiments revealed that RUNX3 knockdown significantly rescued the effects of miR­661 underexpression on NSCLC cell proliferation and invasion. In conclusion, the present findings indicated a role for miR­661 as an oncogene in NSCLC via direct targeting of RUNX3, thus suggesting that miR­661 may be used to develop novel therapies for NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Core Binding Factor Alpha 3 Subunit/metabolism , Lung Neoplasms/pathology , MicroRNAs/metabolism , 3' Untranslated Regions , Aged , Antagomirs/metabolism , Base Sequence , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Disease Progression , Female , Humans , Lung Neoplasms/metabolism , Lymphatic Metastasis , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , RNA Interference , RNA, Small Interfering/metabolism , Sequence Alignment
7.
Leukemia ; 31(10): 2219-2227, 2017 10.
Article in English | MEDLINE | ID: mdl-28119527

ABSTRACT

RUNX3, runt-domain transcription factor, is a master regulator of gene expression in major developmental pathways. It acts as a tumor suppressor in many cancers but is oncogenic in certain tumors. We observed upregulation of RUNX3 mRNA and protein expression in nasal-type extranodal natural killer (NK)/T-cell lymphoma (NKTL) patient samples and NKTL cell lines compared to normal NK cells. RUNX3 silenced NKTL cells showed increased apoptosis and reduced cell proliferation. Potential binding sites for MYC were identified in the RUNX3 enhancer region. Chromatin immunoprecipitation-quantitative PCR revealed binding activity between MYC and RUNX3. Co-transfection of the MYC expression vector with RUNX3 enhancer reporter plasmid resulted in activation of RUNX3 enhancer indicating that MYC positively regulates RUNX3 transcription in NKTL cell lines. Treatment with a small-molecule MYC inhibitor (JQ1) caused significant downregulation of MYC and RUNX3, leading to apoptosis in NKTL cells. The growth inhibition resulting from depletion of MYC by JQ1 was rescued by ectopic MYC expression. In summary, our study identified RUNX3 overexpression in NKTL with functional oncogenic properties. We further delineate that MYC may be an important upstream driver of RUNX3 upregulation and since MYC is upregulated in NKTL, further study on the employment of MYC inhibition as a therapeutic strategy is warranted.


Subject(s)
Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 3 Subunit/physiology , Gene Expression Regulation, Neoplastic , Lymphoma, Extranodal NK-T-Cell/genetics , Nose Neoplasms/genetics , Proto-Oncogene Proteins c-myc/physiology , Transcription, Genetic/genetics , Apoptosis , Azepines/pharmacology , Binding Sites , Cell Division , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Enhancer Elements, Genetic , Genes, Reporter , Genetic Vectors , Humans , Lymphoma, Extranodal NK-T-Cell/etiology , Lymphoma, Extranodal NK-T-Cell/metabolism , Lymphoma, Extranodal NK-T-Cell/pathology , Molecular Targeted Therapy , Nose Neoplasms/etiology , Nose Neoplasms/metabolism , Nose Neoplasms/pathology , Protein Interaction Mapping , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/metabolism , Triazoles/pharmacology , Up-Regulation
8.
Cell Biol Int ; 41(3): 296-308, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28067449

ABSTRACT

Although 13cRA is involved in the regulation of cellular proliferation and differentiation, its physiological roles in chondrocyte proliferation and differentiation still remain unknown. Here, we showed that 13cRA could induce the proliferation of sika deer antler chondrocytes and expression of Ccnd3 and Cdk6. Administration of 13cRA to antler chondrocytes resulted in an obvious increase in the expression of chondrocyte marker Col II and hypertrophic chondrocyte marker Col X. Silencing of Crabp2 expression by specific siRNA could prevent the 13cRA-induced up-regulation of Col X, whereas overexpression of Crabp2 showed the opposite effects. Further study found that Crabp2 mediated the regulation of 13cRA on the expression of Runx3 which was highly expressed in the antler cartilage and inhibited the differentiation of antler chondrocytes. Moreover, attenuation of Runx3 expression greatly raised 13cRA-induced chondrocyte differentiation. Simultaneously, 13cRA could stimulate the expression of Cyp26a1 and Cyp26b1 in the antler chondrocytes. Inhibition of Cyp26a1 and/or Cyp26b1 reinforced the effects of 13cRA on the expression of Col X and Runx3, while overexpression of Cyp26b1 rendered the antler chondrocytes hyposensitive to 13cRA. Collectively, 13cRA may play an important role in the differentiation of antler chondrocytes through targeting Runx3. Crabp2 enhances the effects of 13cRA on chondrocyte differentiation, while Cyp26a1 and Cyp26b1 weaken the sensitivity of antler chondrocytes to 13cRA.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/physiology , Chondrogenesis/physiology , Core Binding Factor Alpha 3 Subunit/biosynthesis , Isotretinoin/pharmacology , Animals , Antlers , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Chondrocytes/drug effects , Chondrogenesis/drug effects , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Deer , Drug Delivery Systems/methods , Isotretinoin/metabolism
9.
Biochem Biophys Res Commun ; 466(3): 592-8, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26392314

ABSTRACT

This study investigated the effect of HCV core protein on the proliferation of hepatocytes and hepatocellular carcinoma cells (HCC), the influence of HCV core protein on HCC apoptosis induced by the chemotherapeutic agent cisplatin, and the mechanism through which HCV core protein acts as a potential oncoprotein in HCV-related HCC by measuring the levels of NR4A1 and Runt-related transcription factor 3 (RUNX3), which are associated with tumor suppression and chemotherapy resistance. In the present study, PcDNA3.1-core and RUNX3 siRNA were transfected into LO2 and HepG2 cells using Lipofectamine 2000. LO2-core, HepG2-core, LO2-RUNX3 (low) and control cells were treated with different concentrations of cisplatin for 72 h, and cell proliferation and apoptosis were assayed using the CellTiter 96(®)Aqueous Non-Radioactive Cell Proliferation Assay Kit. Western blot and real time PCR analyses were used to detect NR4A1, RUNX3, smad7, Cyclin D1 and BAX. Confocal microscopy was used to determine the levels of NR4A1 in HepG2 and HepG2-core cells. The growth rate of HepG2-core cells was considerably greater than that of HepG2 cells. HCV core protein increased the expression of cyclin D1 and decreased the expressions of NR4A1 and RUNX3. In LO2 - RUNX3 (low), the rate of cell proliferation and the level of cisplatin resistance were the same as in the LO2 -core. These results suggest that HCV core protein decreases the sensitivity of hepatocytes to cisplatin by inhibiting the expression of NR4A1 and promoting the expression of smad7, which negatively regulates the TGF-ß pathway. This effect results in down regulation of RUNX3, a target of the TGF-ß pathway. Taken together, these findings indicate that in hepatocytes, HCV core protein increases drug resistance and inhibits cell apoptosis by inhibiting the expressions of NR4A1 and RUNX3.


Subject(s)
Hepacivirus/physiology , Hepacivirus/pathogenicity , Hepatocytes/pathology , Hepatocytes/virology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Viral Core Proteins/physiology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line , Cell Proliferation/drug effects , Cisplatin/pharmacology , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Cyclin D1/metabolism , Drug Resistance, Viral , Hep G2 Cells , Hepatocytes/metabolism , Host-Pathogen Interactions , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Smad7 Protein/metabolism
10.
Respir Res ; 16: 75, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26104385

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia (BPD) in premature infants is a predominantly secondary occurrence to intrauterine inflammation/infection and postpartum mechanical ventilation; in recent years, an association with epigenetics has also been found. DNA methylation, catalyzed by DNA methyl transferases (DNMTs), and tri-methylation of lysine 27 on histone H3 (H3K27me3), mediated by the methyltransferase, Enhancer of Zeste Homolog 2 (EZH2), are some of the most commonly found modifications in epigenetics. Runt-related transcription factor 3 (RUNX3) is associated with pulmonary epithelial and vascular development and regulates expression at the post-transcriptional level by DNA methylation through DNMT1 or DNMT3b. However, the involvements of these epigenetic factors in the occurrence of BPD are, as yet, unclear. METHODS: Newborn rats were randomly assigned to a model, hyperoxia (85 % O2) or control, normoxia group (21 % O2). Lung tissues and alveolar type 2 (AT2) epithelial cells were collected between 1-14 days. The expression of DNMTs, and EZH2 was detected by immunohistochemistry, Western blot and real-time PCR. The percentage of DNA methylation and H3K27me3 levels in the RUNX3 promoter region was measured by bisulfite sequencing PCR and chromatin immunoprecipitation assay. RUNX3 protein and mRNA expression in AT2 cells was also measured after inhibition using the DNA methylation inhibitor, 5-Aza-2'-deoxycytidine, the H3K27me3 inhibitor, JMJD3, and the EZH2 inhibitor, DZNep. RESULTS: Compared with the control group, RUNX3 protein was downregulated and DNMT3b and EZH2 were highly expressed in lung tissues and AT2 cells of the model group (P < 0.05), while high DNA methylation and H3K27me3 modifications were present in the RUNX3 promoter region, in lung tissues of the model group (P < 0.05). Following hyperoxia in the model group, JMJD3 and DZNep significantly reversed the hyperoxia-induced down-regulation of RUNX3 expression in AT2 cells (P < 0.05), more so than 5-Aza-2'-deoxycytidine (P < 0.05). CONCLUSIONS: 1) DNA methylation and H3K27 trimethylation are present in the BPD model; 2) RUNX3 down-regulation is attributed to both DNMT3b-catalyzed DNA methylation and EZH2-catalyzed histone methylation.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Core Binding Factor Alpha 3 Subunit/biosynthesis , DNA Methylation/physiology , Disease Models, Animal , Hyperoxia/metabolism , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/pathology , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Hyperoxia/pathology , Rats , Rats, Sprague-Dawley
11.
PLoS One ; 9(1): e86917, 2014.
Article in English | MEDLINE | ID: mdl-24475196

ABSTRACT

RUNX3 (runt-related transcription factor-3) has been reported to suppress tumor tumorigenesis and metastasis in different human cancers. In this study, we used tissue microarray (TMA) to determine the significance of RUNX3 in prostate cancer progession. Our results showed ectopic expression of RUNX3 in prostate cancer tissues when compared with tumor adjacent normal prostate tissues, and reduced RUNX3 staining was significantly correlated with TNM stage. Moreover, we demonstrated that RUNX3 overexpression inhibited prostate cancer cell migration and invasion resulting from the elevated upregulation of tissue inhibitor of matrix metalloproteinase-2 (TIMP-2), which subsequently inhibited metalloproteinase-2 (MMP-2) expression and activity in vitro. Knock down of RUNX3 expression broke up the balance of TIMP-2/MMP-2, whereas silence of TIMP-2 resulted in the inhibition of MMP-2 expression in prostate cells. We also showed that restoration of RUNX3 decreased vascular endothelial growth factor (VEGF) secretion and suppressed endothelial cell growth and tube formation. Strikingly, RUNX3 was demonstrated to inhibit tumor metastasis and angiogenesis in vivo. Altogether, our results support the tumor suppressive role of RUNX3 in human prostate cancer, and provide insights into development of targeted therapy for this disease.


Subject(s)
Carcinoma/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Prostatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Animals , Carcinoma/blood supply , Carcinoma/metabolism , Carcinoma/secondary , Cell Movement , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/metabolism , Disease Progression , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Neoplasm Transplantation , Neovascularization, Pathologic , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tissue Array Analysis , Tissue Inhibitor of Metalloproteinase-2/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tumor Microenvironment , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
ChemMedChem ; 9(3): 649-56, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24376239

ABSTRACT

Expression and stability of the tumor suppressor runt-related transcription factor 3 (RUNX3) are regulated by histone deacetylase (HDAC). HDAC inhibition alters epigenetic and posttranslational stability of RUNX3, leading to tumor suppression. However, HDAC inhibitors can nonselectively alter global gene expression through chromatin remodeling. Thus, lactam-based HDAC inhibitors were screened to identify potent protein stabilizers that maintain RUNX3 stability by acetylation. RUNX activity and HDAC inhibition were determined for 111 lactam-based analogues through a cell-based RUNX activation and HDAC inhibition assay. 3-[1-(4-Bromobenzyl)-2-oxo-2,5-dihydro-1H-pyrrol-3-yl]-N-hydroxypropanamide (11-8) significantly increased RUNX3 acetylation and stability with relatively low RUNX3 mRNA expression and HDAC inhibitory activity. This compound showed significant antitumor effects, which were stronger than SAHA, in an MKN28 xenograft model. Thus, we propose a novel strategy, in which HDAC inhibitors serve as antitumor chemotherapeutic agents that selectively target epigenetic regulation and protein stability of RUNX3.


Subject(s)
Antineoplastic Agents/pharmacology , Core Binding Factor Alpha 3 Subunit/metabolism , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Lactams/pharmacology , Neoplasms, Experimental/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/chemistry , Core Binding Factor Alpha 3 Subunit/genetics , Dose-Response Relationship, Drug , Gene Expression Profiling , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Lactams/chemical synthesis , Lactams/chemistry , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Stability/drug effects , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
13.
Int J Oncol ; 43(5): 1511-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24042352

ABSTRACT

We recently reported that the tumor suppressor Runt-related transcription factor 3 (RUNX3) is silenced in colorectal cancer cells via oxidative stress-induced hypermethylation of its promoter. The resulting downregulation of RUNX3 expression influences cell proliferation. Activation of the Akt signaling pathway is also associated with cell survival and proliferation; however, the effects of oxidative stress on the relationship between RUNX3 and Akt signaling are largely unknown. Therefore, this study investigated the mechanisms involved in cell proliferation caused by oxidative stress-induced silencing of RUNX3. The levels of RUNX3 mRNA and protein were downregulated in response to treatment of the human colorectal cancer cell line SNU-407 with H2O2. Treatment of the cells with H2O2 also upregulated Akt mRNA and protein expression, and inhibited the binding of RUNX3 to the Akt promoter. The inverse correlation between the expression levels of RUNX3 and Akt in H2O2-treated cells was also associated with nuclear translocation of ß-catenin and upregulation of cyclin D1 expression, which induced cell proliferation. H2O2 treatment also increased the binding of ß-catenin to the cyclin D1 promoter. The results presented here demonstrate that reactive oxygen species silence the tumor suppressor RUNX3, enhance the Akt-mediated signaling pathway, and promote the proliferation of colorectal cancer cells.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/pathology , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Oxidative Stress , Proto-Oncogene Proteins c-akt/genetics , Blotting, Western , Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Cyclin D1 , Humans , Hydrogen Peroxide/pharmacology , Immunoenzyme Techniques , Oxidants/pharmacology , Promoter Regions, Genetic/genetics , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , beta Catenin/genetics , beta Catenin/metabolism
14.
Clin Exp Metastasis ; 30(2): 165-76, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22886570

ABSTRACT

The function of tropomyosin receptor kinase (Trk) family including TrkA, TrkB, and TrkC in cancer remains unknown. The role of Trks in oral squamous cell carcinoma (OSCC) was examined. Knockdown of Trks provided inhibition of growth or invasion and decrease of apoptosis in OSCC cells, which expressed Trks at high levels. VEGF expression was associated with TrkA and TrkB expression; a decrease of VEGF-C and VEGF-D was observed in OSCC cells with TrkB knockdown. TrkC did not affect the expression of VEGF family. An immunohistochemical analysis of 102 OSCCs showed that TrkB expression was related to microvessel density (MVD), lymph vessel density (LVD), and poor prognosis. TrkC expression was correlated with clinical stage, lymph node metastasis, MVD, LVD, and poor prognosis. TrkA expression was associated with VEGF expression, whereas TrkB expression was associated with the expressions of VEGF, VEGF-C and VEGF-D. No significant association was found between the expression of TrkC and genes of the VEGF family. Expression of Trks was not associated with RUNX3 silencing by methylation in OSCC cells. Trks expression was inversely correlated with RUNX3 expression in the OSCC cases. These results suggested that Trks enhances progression of OSCC through angiogenesis and lymphangiogenesis.


Subject(s)
Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/pathology , Neovascularization, Pathologic , Receptor, trkA/metabolism , Adult , Aged , Apoptosis , Blotting, Western , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cell Adhesion , Cell Movement , Cell Proliferation , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , DNA/genetics , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics , Survival Rate
15.
J Immunol ; 189(2): 832-40, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22685315

ABSTRACT

A transcription factor network that includes STAT4, T-bet, and Runx3 promotes the differentiation of Th1 cells and inflammatory immune responses. How additional transcription factors regulate the function of Th1 cells has not been defined. In this study we show that the negative regulatory factor Twist1 decreases expression of T-bet, Runx3, and IL-12Rß2 as it inhibits IFN-γ production. Ectopic expression of Runx3, but not T-bet or IL-12Rß2, compensates for the effects of Twist1 on IFN-γ production, and Twist1 regulation of Ifng depends on complex formation with Runx3. Twist1 decreases Runx3 and T-bet binding at the Ifng locus, and it decreases chromatin looping within the Ifng locus. These data define an IL-12/STAT4-induced negative regulatory loop that impacts multiple components of the Th1 transcriptional network and provide further insight into regulation of Th1 differentiation.


Subject(s)
Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/physiology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Nuclear Proteins/physiology , Th1 Cells/immunology , Th1 Cells/metabolism , Twist-Related Protein 1/physiology , Animals , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/prevention & control , Interferon-gamma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , STAT4 Transcription Factor/antagonists & inhibitors , STAT4 Transcription Factor/deficiency , STAT4 Transcription Factor/physiology , T-Box Domain Proteins/antagonists & inhibitors , T-Box Domain Proteins/genetics , T-Box Domain Proteins/physiology , Twist-Related Protein 1/genetics
16.
Cell Cycle ; 11(10): 1938-47, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22544322

ABSTRACT

RUNX family proteins are critical regulators of lineage differentiation during development. The high prevalence of RUNX mutation/epigenetic inactivation in human cancer indicates a causative role for dysfunctional RUNX in carcinogenesis. This is supported by well-documented evidence of functional interaction of RUNX with components of major oncogenic or tumor suppressive signaling pathways such as TGFß and Wnt. Here, we explore the binding partners of RUNX3 proteins to further define the scope of RUNX3 function. Using a mass spectrometry-based approach, we found that RUNX3 binds to centrosomal protein rootletin. This led us to uncover the presence of RUNX proteins at the centrosome. Our findings suggest a potential function for RUNX3 during mitosis.


Subject(s)
Centrosome/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Cytoskeletal Proteins/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , HCT116 Cells , HeLa Cells , Humans , Mass Spectrometry , Mitosis , Nocodazole/pharmacology , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Tubulin/metabolism , Wnt Proteins/metabolism
17.
Gynecol Oncol ; 122(2): 410-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21612813

ABSTRACT

OBJECTIVE: The Runt domain transcription factor, RUNX3, has been shown to be a tumor suppressor in a variety of cancers including gastric, colon and breast cancer. Interestingly, an oncogenic role for RUNX3 has also been suggested in basal cell carcinoma and head and neck cancer. Here, we explore the role of RUNX3 in ovarian cancer. METHODS: Expression of RUNX3 mRNA and protein was evaluated in human ovarian cancer cell lines. In addition, subcellular localization of RUNX3 was also examined in cell lines and ovarian cancer tissues. Effect of exogenous RUNX3 expression and knockdown on cell proliferation was investigated by proliferation assays and a soft agar assay. RESULTS: Expression of RUNX3 was detected in the nucleus of ovarian cancer cell lines and ovarian cancer tissues and was found to play a growth stimulatory role. RUNX3 knockdown resulted in a decrease in cell proliferation in liquid media as well as in soft agar. Despite the fact that exogenous expression of RUNX3 strongly inhibits cell growth in many cell types, RUNX3 promoted cell growth in ovarian cancer cell lines not expressing RUNX3. CONCLUSION: RUNX3 is frequently expressed in the nuclei of ovarian cancer cell lines and plays an oncogenic role in ovarian cancer.


Subject(s)
Core Binding Factor Alpha 3 Subunit/physiology , Oncogenes , Ovarian Neoplasms/etiology , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Female , Gene Expression Profiling , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
18.
Cancer Sci ; 102(4): 776-83, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21205092

ABSTRACT

RUNX3 is a candidate tumor suppressor gene localized in 1p36, a region frequently inactivated through hypermethylation, histone modulation, and other processes in various human tumors. In this study, to elucidate a causal link between RUNX3 expression and biliary tract cancer, we investigated 17 human biliary cancer specimens. In addition, to examine roles of RUNX3 in biliary tract cancer, we restored silenced RUNX3 in the human biliary cancer cell line Mz-ChA-2 using a histone deacetylase inhibitor. Thirteen of 17 human cancer specimens exhibited suppressed RUNX3 expression compared with normal biliary ducts. Moreover, the decreased RUNX3 expression was related to a lower accumulation of acetylated histone H3 associated with RUNX3. In in vitro experiments, vorinostat, a member of a new class of highly potent histone deacetylase inhibitors, restored RUNX3 expression in Mz-ChA-2 cells. Furthermore, vorinostat-induced RUNX3 significantly enhanced p21 expression and growth inhibition of Mz-ChA-2 cells through restoration of TGF-ß signaling. These data suggest the significance of histone deacetylation-associated suppression of RUNX3 expression in biliary tract carcinogenesis. Furthermore, vorinostat might hold promise for treating biliary tract cancer through enhancement of TGF-ß signaling by restoration of RUNX3.


Subject(s)
Biliary Tract Neoplasms/genetics , Biliary Tract Neoplasms/pathology , Core Binding Factor Alpha 3 Subunit/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Histones/genetics , Acetylation , Biliary Tract Neoplasms/drug therapy , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Immunoenzyme Techniques , Luciferases/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Vorinostat
19.
Oncogene ; 29(18): 2605-15, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20348954

ABSTRACT

The study of RUNX3 in tumor pathogenesis is a rapidly expanding area of cancer research. Functional inactivation of RUNX3-through mutation, epigenetic silencing, or cytoplasmic mislocalization-is frequently observed in solid tumors of diverse origins. This alone indicates that RUNX3 inactivation is a major risk factor in tumorigenesis and that it occurs early during progression to malignancy. Conversely, RUNX3 has also been described to have an oncogenic function in a subset of tumors. Although the mechanism of how RUNX3 switches from tumor suppressive to oncogenic activity is unclear, this is of clinical relevance with implications for cancer detection and prognosis. Recent developments have significantly contributed to our understanding of the pleiotropic tumor suppressive properties of RUNX3 that regulate major signaling pathways. This review summarizes the important findings that link RUNX3 to tumor suppression.


Subject(s)
Core Binding Factor Alpha 3 Subunit/physiology , Neoplasms/etiology , Animals , Cellular Senescence , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Cytoplasm/metabolism , DNA Methylation , DNA Repair , Gene Deletion , Gene Silencing , Humans , Mutagenesis, Insertional , Protein Transport , Signal Transduction
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...