Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Anal Chim Acta ; 1138: 181-190, 2020 Nov 22.
Article in English | MEDLINE | ID: mdl-33161980

ABSTRACT

Human Pituitary Tumour Transforming Gene 1 (PTTG1) is an oncoprotein involved in maintaining chromosome stability and acts as a biomarker for a panel of cancers. In this study, we endeavoured to generate an RNA aptamer against PTTG1. The RNA aptamer, SECURA-3 has an estimated equilibrium dissociation constant of 16.41 ± 6.4 nM. The aptamer was successfully harnessed in several diagnostic platforms including ELASA, aptamer-based dot blot and aptamer-based western blot. SECURA-3 was also unveiled as a potential probe that could replace its counterpart antibody in the histostaining-based detection of PTTG1 in HeLa and MCF-7 formalin-fixed paraffin-embedded cell blocks. In the aspect of therapeutics, SECURA-3 RNA aptamer demonstrates an antagonistic effect by antagonizing the interaction between PTTG1 and CXCR2, as revealed in the in vitro competitive nitrocellulose filter binding assay and dual-luciferase reporter assay in HeLa cells. As the first anti-PTTG1 aptamer, SECURA-3 RNA aptamer has immense diagnostic and therapeutic properties.


Subject(s)
Aptamers, Nucleotide , Securin/genetics , HeLa Cells , Humans , Oncogenes
2.
PLoS One ; 8(8): e71282, 2013.
Article in English | MEDLINE | ID: mdl-23977008

ABSTRACT

The aberrant expression of proto-oncogenes is involved in processes that are responsible for cellular proliferation and the inhibition of myeloid differentiation in acute myeloid leukemia (AML). Pituitary Tumor-Transforming gene 1 (PTTG1), an oncogenic transcription factor, is abundantly expressed in various human cancers and hematopoietic malignancies. However, its expression in normal leukocytes and most normal tissues is very low or undetectable. The mechanism by which PTTG1 overexpression modifies myeloid cell development and promotes leukemogenesis remain unclear. To investigate the mechanistic links between PTTG1 overexpression and leukemia cell differentiation, we utilized phorbol 12-myristate 13-acetate (PMA), a well-known agent that triggers monocyte/macrophage differentiation, to analyze the expression patterns of PTTG1 in PMA-induced myeloid differentiation. We found that PTTG1 is down-regulated at the transcriptional level in PMA-treated HL-60 and THP1 cells. In addition, we identified a binding site for a tumor suppressor protein, Kruppel-like factor 6 (KLF6), in the PTTG1 promoter. We found that KLF6 could directly bind and repress PTTG1 expression. In HL-60 and THP1 cells, KLF6 mRNA and protein levels are up-regulated with a concordant reduction of PTTG1 expression upon treatment with PMA. Furthermore, KLF6 knockdown by shRNA abolished the suppression of PTTG1 and reduced the activation of the differentiation marker CD11b in PMA-primed cells. The protein kinase C (PKC) inhibitor and the MAPK/ERK kinase (MEK) inhibitor significantly blocked the potentiation of PMA-mediated KLF6 induction and the down-regulation of PTTG1, indicating that PTTG1 is suppressed via the activation of PKC/ERK/KLF6 pathway. Our findings suggest that drugs that increase the KLF6 inhibition of PTTG1 may have a therapeutic application in AML treatment strategies.


Subject(s)
Gene Expression Regulation, Leukemic/drug effects , Kruppel-Like Transcription Factors/genetics , Proto-Oncogene Proteins/genetics , Securin/genetics , Tetradecanoylphorbol Acetate/pharmacology , Binding Sites , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cell Differentiation , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , HL-60 Cells , Humans , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Securin/metabolism , Signal Transduction
3.
Breast Cancer Res ; 14(4): R106, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22789011

ABSTRACT

INTRODUCTION: hPTTG1 (human pituitary tumor-transforming gene 1) is an oncogene overexpressed in breast cancer and several other types of cancer. Increased hPTTG1 expression has been shown to be associated with poor patient outcomes in breast cancer. Although hPTTG1 overexpression plays important roles in promoting the proliferation, invasion, and metastasis of cancer cells, it also has been suggested to induce cellular senescence. Deciphering the mechanism by which hPTTG1 overexpression induces these contradictory actions in breast cancer cells is critical to our understanding of the role of hPTTG1 in breast cancer development. METHODS: MCF-10A and MCF-7 cells were used to identify the mechanism of hPTTG1-induced senescence. The interplay between hPTTG1 overexpression and chemokine C-X-C motif receptor 2 (CXCR2)/p21-dependent senescence in tumor growth and metastasis of MCF-7 cells was investigated by orthotopic transplantation of severe combined immunodeficiency (SCID) mice. Additionally, human invasive ductal carcinoma (IDC) tissue arrays were used to confirm the hPTTG1/CXCR2/p21 axis established in vitro. RESULTS: In this study, we investigated the mechanism of hPTTG1-induced senescence as well as its role in breast cancer progression and metastasis. Herein, we showed that hPTTG1 overexpression reinforced senescence through the CXCR2/p21 signaling. Furthermore, hPTTG1 overexpression activated NF-κB signaling to transactivate the expression of interleukin (IL)-8 and growth-regulated oncogene alpha (GROα) to execute CXCR2 signaling in MCF-7 cells. When CXCR2 expression was knocked down in hPTTG1-overexpressing MCF-7 cells, hPTTG1-induced senescence was abrogated by alleviating CXCR2-induced p21 expression. In a mouse model, CXCR2-mediated senescence limited hPTTG1-induced tumor growth and metastasis. Moreover, CXCR2 knockdown in hPTTG1-overexpressing MCF-7 tumors dramatically accelerated tumor growth and metastasis. Our in vitro and in vivo results demonstrated that hPTTG1 overexpression reinforces senescence through CXCR2 signaling, and the evasion of CXCR2/p21-dependent senescence was critical to hPTTG1 exerting its oncogenic potential. Interestingly, although CXCR2-dependent senescence restrained hPTTG1-induced tumor progression, when MCF-7 cells and hPTTG1-overexpressing MCF-7 cells were co-transplanted into the mammary fat pads of SCID mice, hPTTG1-overexpressing senescent cells created a metastasis-promoting microenvironment that promoted lung metastasis of the MCF-7 cells. Immunohistochemical analysis of human breast tumor samples also confirmed the importance of the hPTTG1/CXCR2 axis in promoting breast cancer metastasis. CONCLUSIONS: Our findings provide novel molecular insights into hPTTG1-induced senescence and identify a novel mechanism by which hPTTG1 promotes metastasis by regulating the senescence-associated microenvironment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Interleukin-8B/metabolism , Securin/genetics , Signal Transduction , Breast Neoplasms/pathology , Cell Line, Tumor , Cellular Senescence/genetics , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Ligands , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/metabolism
4.
Int J Radiat Oncol Biol Phys ; 77(2): 566-74, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20457353

ABSTRACT

PURPOSE: Radiotherapy is one of the best choices for cancer treatment. However, various tumor cells exhibit resistance to irradiation-induced apoptosis. The development of new strategies to trigger cancer cell death besides apoptosis is necessary. This study investigated the role of securin in radiation-induced apoptosis and senescence in human cancer cells. METHODS AND MATERIALS: Cell survival was determined using clonogenic assays. Western blot analysis was used to analyze levels of securin, caspase-3, PARP, p53, p21, Rb, gamma-H2AX, and phospho-Chk2. Senescent cells were analyzed using a beta-galactosidase staining assay. A securin-expressed vector (pcDNA-securin) was stably transfected into securin-null HCT116 cells. Securin gene knockdown was performed by small interfering RNA and small hairpin RNA in HCT116 and MDA-MB-231 cells, respectively. RESULTS: Radiation was found to induce apoptosis in securin wild type HCT116 cells but induced senescence in securin-null cells. Restoration of securin reduced senescence and increased cell survival in securin-null HCT116 cells after irradiation. Radiation-induced gamma-H2AX and Chk2 phosphorylation were induced transiently in securin-wild-type cells but exhibited sustained activation in securin-null cells. Securin gene knockdown switches irradiation-induced apoptosis to senescence in both HCT116 p53-null and MDA-MB-231 cells. CONCLUSIONS: Our results demonstrated that the level of securin expression plays a determining role in the radiosensitivity and fate of cells. Depletion of securin impairs DNA repair after irradiation, increasing DNA damage and promoting senescence in the residual surviving cells regardless of functional p53 expression. The knockdown of securin may contribute to a novel radiotherapy protocol for the treatment of human cancer cells that are resistant to irradiation.


Subject(s)
Apoptosis/physiology , Cellular Senescence/physiology , Neoplasm Proteins/metabolism , Radiation Tolerance/physiology , Tumor Suppressor Protein p53/metabolism , Apoptosis/radiation effects , Blotting, Western , Caspase 3/metabolism , Cell Line, Tumor/radiation effects , Cellular Senescence/radiation effects , Checkpoint Kinase 2 , Colony-Forming Units Assay/methods , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Repair , Gene Knockdown Techniques/methods , HCT116 Cells/radiation effects , Histones/metabolism , Humans , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phosphorylation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism , Securin
5.
Mol Cell Biol ; 28(23): 7096-108, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18794357

ABSTRACT

The loss of E-cadherin gene expression can cause the dysfunction of the cell-cell junction to trigger tumor metastasis. Members of the Snail family of transcription factors are repressors of the expression of the E-cadherin gene. In this study, we showed that the activated androgen receptor (AR) is a novel repressor of E-cadherin gene expression and can promote metastasis. Our results demonstrated that the activated AR could bind to the E-cadherin promoter in vitro and in vivo. The activated AR and HDAC1 had synergistic effects in downregulating E-cadherin gene expression. Treating cells with the AR ligand, dihydrotestosterone (DHT), triggered the reduction of E-cadherin expression and induced changes in cell morphology from an epithelial-like to a mesenchymal-like appearance. When nonmetastatic breast cancer cells expressing cytoplasmic AR were transplanted into mice and the mice were treated with DHT, tumors were detected at metastatic sites, whereas no tumors were detected in transplanted mice without DHT treatment. Furthermore, clinical data from breast cancer patients with invasive ductal carcinomas showed high levels of AR expression in the nuclei and low levels of E-cadherin expression. These results suggest that, similarly to Snail and Twist, the activated AR can downregulate E-cadherin expression to promote the activation of epithelial-mesenchymal transition and tumor metastasis.


Subject(s)
Cadherins/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis/pathology , Receptors, Androgen/physiology , Animals , Cadherins/analysis , Carcinoma, Ductal, Breast/chemistry , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Down-Regulation , Epithelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Mice , Neoplasm Proteins/analysis , Neoplasms, Experimental/pathology , Promoter Regions, Genetic , Protein Binding , Receptors, Androgen/analysis , Receptors, Androgen/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
6.
Biochem Biophys Res Commun ; 367(2): 235-41, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18062917

ABSTRACT

Twist is a basic helix loop helix protein that plays a role both in human development and in cancer biogenesis. While characterizing the effects of Twist on breast epithelial cell transformation, we identified E-cadherin as a target gene that is down-regulated by Twist. In this study, we demonstrate that Twist can transcriptionally repress E-cadherin in breast cancer cells. Using transient promoter assays, we show that Twist can down-regulate E-cadherin promoter activity by up to two folds. This is further supported by immunoblot analyses which indicates that over-expression of Twist decreases E-cadherin protein levels in breast cancer cell lines. Subsequently, chromatin immunoprecipitation performed on MCF-7/Twist and Hs578 T (high level of endogenous Twist expression) confirmed Twist binding to the E-cadherin promoter. Finally, the functional relevance of this regulation was verified by quantitative real-time PCR and immunohistochemistry on a cohort of breast cancer samples.


Subject(s)
Breast Neoplasms/metabolism , Cadherins/metabolism , Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcriptional Activation , Twist-Related Protein 1/metabolism , Cell Line, Tumor , Humans
7.
Cancer Res ; 67(22): 11064-73, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006853

ABSTRACT

The potassium chloride cotransporter (KCC) is a major determinant of osmotic homeostasis and plays an emerging role in tumor biology. Here, we investigate if KCC is involved in the regulation of epithelial-mesenchymal transition (EMT), a critical cellular event of malignancy. E-cadherin and beta-catenin colocalize in the cell-cell junctions, which becomes more obvious in a time-dependent manner by blockade of KCC activity in cervical cancer SiHa and CaSki cells. Real-time reverse transcription-PCR on the samples collected from the laser microdissection indicates that KCC3 is the most abundant KCC isoform in cervical carcinoma. The characteristics of EMT appear in KCC3-overexpressed, but not in KCC1- or KCC4-overexpressed cervical cancer cells, including the elongated cell shape, increased scattering, down-regulated epithelial markers (E-cadherin and beta-catenin), and up-regulated mesenchymal marker (vimentin). Some cellular functions are enhanced by KCC3 overexpression, such as increased invasiveness and proliferation, and weakened cell-cell association. KCC3 overexpression decreases mRNA level of E-cadherin. The promoter activity assays of various regulatory sequences confirm that KCC3 expression is a potent negative regulator for human E-cadherin gene expression. The proteosome inhibitor restores the decreased protein abundance of beta-catenin by KCC3 overexpression. In the surgical specimens of cervical carcinoma, the decreased E-cadherin amount was accompanied by the increased KCC3 abundance. Vimentin begins to appear at the invasive front and becomes significantly expressed in the tumor nest. In conclusion, KCC3 down-regulates E-cadherin/beta-catenin complex formation by inhibiting transcription of E-cadherin gene and accelerating proteosome-dependent degradation of beta-catenin protein. The disruption of E-cadherin/beta-catenin complex formation promotes EMT, thereby stimulating tumor progression.


Subject(s)
Epithelium/metabolism , Gene Expression Regulation, Neoplastic , Mesoderm/metabolism , Symporters/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Humans , Models, Biological , Neoplasm Invasiveness , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms , Reverse Transcriptase Polymerase Chain Reaction , beta Catenin/metabolism
8.
Oncogene ; 24(56): 8277-90, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16116478

ABSTRACT

In cancer cells, loss of E-cadherin gene expression caused dysfunction of the cell-cell junction system, triggering cancer invasion and metastasis. Therefore, E-cadherin is an important tumor-suppressor gene. To understand how E-cadherin gene expression is regulated in cancer cells, we have used E-cadherin-positive and -negative expressing cells to find out the possible up- or down regulating transcription factors in human E-cadherin regulatory sequences. Functional analysis of human E-cadherin regulatory sequences constructs indicated that AML1, Sp1, and p300 may play important roles in promoting E-cadherin expression. In addition, we found there are four HNF3-binding sites in human E-cadherin regulatory sequences. The exogenous HNF3 can enhance the E-cadherin promoter activity in metastatic breast cancer cells and the metastatic breast cancer cells stably transfected with HNF3 showed re-expression of E-cadherin. The HNF3 stable transfectants changed from mesenchymal-like into epithelial morphology. The transwell assays showed the re-expressed E-cadherin reduced cell motility of metastatic breast cancer cells. These results suggested HNF3 may play important roles in the upregulation of the E-cadherin promoter, with the consequent re-expression of E-cadherin, thus reducing the metastatic potential of breast cancer cells. These findings suggested HNF3 plays important roles in the upregulation of the E-cadherin gene and may be able to reduce the motility of metastatic breast cancer cells.


Subject(s)
Cadherins/biosynthesis , Cadherins/genetics , Gene Expression Regulation/physiology , Hepatocyte Nuclear Factors/physiology , Base Sequence , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Chromatin/physiology , Core Binding Factor Alpha 2 Subunit/metabolism , DNA Methylation , HCT116 Cells , Hepatocyte Nuclear Factors/genetics , Humans , Molecular Sequence Data , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Promoter Regions, Genetic , Snail Family Transcription Factors , Transcription Factors/metabolism
9.
Oncogene ; 21(30): 4673-9, 2002 Jul 11.
Article in English | MEDLINE | ID: mdl-12096343

ABSTRACT

Tumorigenesis is a multi-step process involving a series of changes of cellular genes. Most solid tumors and hematopoietic malignancies often show abnormal chromosome numbers, the aneuploidy. The chromosomal aneuploidy keeps cells in the state of chromosomal instability (CIN) that will increase the mutation rate of cells affected and thus push them deeper into the process of tumorigenesis. The yeast genetic studies showed that normal distribution of chromosome during mitosis is under the surveillance of a set of genes, the spindle assembly checkpoint genes, that include the BUB and MAD gene groups and MPS. In some colorectal cancers with CIN it was found to have hBUB1 gene mutated and the mutated gene functions dominantly. We have examined a series of breast cancer cell lines with or without CIN for the hBUB1 gene mutation and found none. However, we detected various degrees of deletion in the coding sequences of the hBUB1 gene in cells from T lymphoblastic leukemia cell lines, Molt3 and Molt4, and cells from some acute lymphoblastic leukemia and Hodgkin's lymphoma patients. So far the lesions of deletion are in the kinetochore localization domain of the hBUB1 gene that may explain why the deletion lesions in the BUB1 gene cause aneuploidy in lymphoma and lymphoma cells. The deletions are heterozygous in nature. Like the mutated hBUB1 gene in colorectal cancer, the mutant hBUB1 cDNA from lymphoblastic leukemia cells behaves dominantly.


Subject(s)
Aneuploidy , Leukemia/genetics , Lymphoma/genetics , Protein Kinases/genetics , Breast Neoplasms/genetics , Cell Cycle Proteins/metabolism , DNA Mutational Analysis , Genes, Dominant/genetics , Humans , Mitosis , Poly-ADP-Ribose Binding Proteins , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...