Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Med ; 1(1): 5-16, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23342249

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a critical process providing tumor cells with the ability to migrate and escape from the primary tumor and metastasize to distant sites. Recently, EMT was shown to be associated with the cancer stem cell (CSC) phenotype in breast cancer. Snail is a transcription factor that mediates EMT in a number of tumor types, including colorectal cancer (CRC). Our study was done to determine the role of Snail in mediating EMT and CSC function in CRC. Human CRC specimens were stained for Snail expression, and human CRC cell lines were transduced with a retroviral Snail construct or vector control. Cell proliferation and chemosensitivity to oxaliplatin of the infected cells were determined by the MTT (colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. Migration and invasion were determined in vitro using modified Boyden chamber assays. EMT and putative CSC markers were analyzed using Western blotting. Intravenous injection of tumor cells was done to evaluate their metastatic potential in mice. Snail was overexpressed in human CRC surgical specimens. This overexpression induced EMT and a CSC-like phenotype in human CRC cells and enhanced cell migration and invasion (P < 0.002 vs. control). Snail overexpression also led to an increase in metastasis formation in vivo (P < 0.002 vs. control). Furthermore, the Snail-overexpressing CRC cells were more chemoresistant to oxaliplatin than control cells. Increased Snail expression induces EMT and the CSC-like phenotype in CRC cells, which enhance cancer cell invasion and chemoresistance. Thus, Snail is a potential therapeutic target in metastatic CRC.


Subject(s)
Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , Phenotype , Transcription Factors/genetics , Animals , Cell Line , Cell Movement/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Gene Expression , Heterografts , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Snail Family Transcription Factors , Transcription Factors/metabolism
2.
PLoS One ; 6(10): e23208, 2011.
Article in English | MEDLINE | ID: mdl-22028766

ABSTRACT

NRP-2 is a high-affinity kinase-deficient receptor for ligands belonging to the class 3 semaphorin and vascular endothelial growth factor families. NRP-2 has been detected on the surface of several types of human cancer cells, but its expression and function in gastrointestinal (GI) cancer cells remains to be determined. We sought to determine the function of NRP-2 in mediating downstream signals regulating the growth and survival of human gastrointestinal cancer cells. In human gastric cancer specimens, NRP-2 expression was detected in tumor tissues but not in adjacent normal mucosa. In CNDT 2.5 cells, shRNA mediated knockdown NRP-2 expression led to decreased migration and invasion in vitro (p<0.01). Focused gene-array analysis demonstrated that loss of NRP-2 reduced the expression of a critical metastasis mediator gene, S100A4. Steady-state levels and function of ß-catenin, a known regulator of S100A4, were also decreased in the shNRP-2 clones. Furthermore, knockdown of NRP-2 sensitized CNDT 2.5 cells in vitro to 5FU toxicity. This effect was associated with activation of caspases 3 and 7, cleavage of PARP, and downregulation of Bcl-2. In vivo growth of CNDT 2.5 cells in the livers of nude mice was significantly decreased in the shNRP-2 group (p<0.05). Intraperitoneal administration of NRP-2 siRNA-DOPC decreased the tumor burden in mice (p = 0.01). Collectively, our results demonstrate that tumor cell-derived NRP-2 mediates critical survival signaling in gastrointestinal cancer cells.


Subject(s)
Gastrointestinal Neoplasms/pathology , Neuropilin-2/metabolism , Signal Transduction , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neuropilin-2/deficiency , Neuropilin-2/genetics , Protein Stability , Protein Transport , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , beta Catenin/chemistry , beta Catenin/genetics
3.
Gastroenterology ; 141(5): 1728-37, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21806944

ABSTRACT

BACKGROUND & AIMS: Metastatic gastrointestinal neuroendocrine tumors (NETs) frequently are refractory to chemotherapy. Chemoresistance in various malignancies has been attributed to cancer stem cells (CSCs). We sought to identify gastrointestinal neuroendocrine CSCs (N-CSCs) in surgical specimens and a NET cell line and to characterize novel N-CSC therapeutic targets. METHODS: Human gastrointestinal NETs were evaluated for CSCs using the Aldefluor (Stemcell Technologies, Vancouver, Canada) assay. An in vitro, sphere-forming assay was performed on primary NET cells. CNDT2.5, a human midgut carcinoid cell line, was used for in vitro (sphere-formation) and in vivo (tumorigenicity assays) CSC studies. N-CSC protein expression was characterized using Western blotting. In vivo, systemic short interfering RNA administration targeted Src. RESULTS: By using the Aldefluor assay, aldehyde dehydrogenase-positive (ALDH+) cells comprised 5.8% ± 1.4% (mean ± standard error of the mean) of cells from 19 patient samples. Although many primary cell lines failed to grow, CNDT96 ALDH+ cells formed spheres in anchorage-independent conditions, whereas ALDH- cells did not. CNDT2.5 ALDH+ cells formed spheres, whereas ALDH- cells did not. In vivo, ALDH+ CNDT2.5 cells generated more tumors, with shorter latency than ALDH- or sham-sorted cells. Compared with non-CSCs, ALDH+ cells demonstrated increased expression of activated Src, Erk, Akt, and mammalian target of rapamycin (mTOR). In vivo, anti-Src short interfering RNA treatment of ALDH+ tumors reduced tumor mass by 91%. CONCLUSIONS: CSCs are present in NETs, as shown by in vitro sphere formation and in vivo tumorigenicity assays. Src was activated in N-CSCs and represents a potential therapeutic target in gastrointestinal NETs.


Subject(s)
Carcinoid Tumor/pathology , Gastrointestinal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Neuroendocrine Tumors/pathology , Aldehyde Dehydrogenase/metabolism , CSK Tyrosine-Protein Kinase , Carcinogenicity Tests , Carcinoid Tumor/metabolism , Cell Line, Tumor , Cells, Cultured , Gastrointestinal Neoplasms/metabolism , Humans , In Vitro Techniques , Neuroendocrine Tumors/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Sirolimus/metabolism , src-Family Kinases
5.
Clin Cancer Res ; 15(22): 6763-70, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19887479

ABSTRACT

Class 3 semaphorins (SEMA3) were first identified as glycoproteins that negatively mediate neuronal guidance by binding to neuropilin and repelling neurons away from the source of SEMA3. However, studies have shown that SEMA3s are also secreted by other cell types, including tumor cells, where they play an inhibitory role in tumor growth and angiogenesis (specifically SEMA3B and SEMA3F). SEMA3s primarily inhibit the cell motility and migration of tumor and endothelial cells by inducing collapse of the actin cytoskeleton via neuropilins and plexins. Besides binding to SEMA3s, neuropilin also binds the protumorigenic and proangiogenic ligand vascular endothelial growth factor (VEGF). Although some studies attribute the antitumorigenic and antiangiogenic properties of SEMA3s to competition between SEMA3s and VEGF for binding to neuropilin receptors, several others have shown that SEMA3s display growth-inhibitory activity independent of competition with VEGF. A better understanding of these molecular interactions and the role and signaling of SEMA3s in tumor biology will help determine whether SEMA3s represent potential therapeutic agents. Herein, we briefly review (a) the role of SEMA3s in mediating tumor growth, (b) the SEMA3 receptors neuropilins and plexins, and (c) the potential competition between SEMA3s and VEGF family members for neuropilin binding.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic , Semaphorins/physiology , Angiogenesis Inhibitors/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Movement , Endothelial Cells/cytology , Humans , Ligands , Models, Biological , Neoplasm Metastasis , Nerve Tissue Proteins/metabolism , Neuropilins/metabolism , Vascular Endothelial Growth Factor A/metabolism
6.
Semin Oncol ; 36(2 Suppl 1): S12-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19393831

ABSTRACT

Our understanding of the process of tumor angiogenesis has changed significantly since the late 1970s, when vascular endothelial growth factor (VEGF) was first identified as vascular permeability factor and later found to be the major mediator of physiologic and pathologic angiogenesis. Since then, several additional VEGF-related ligands, VEGF receptors (VEGFRs), and complementary/alternative pathways that regulate tumor angiogenesis have been identified. Over the last decade, several antiangiogenic agents have been developed with the aim to inhibit new blood vessel growth, and we have learned that VEGF inhibition does far more than simply block new blood vessel growth. Clinical studies have demonstrated an improvement of progression-free and overall survivals with anti-VEGF therapy (with or without chemotherapy) in patients with advanced-stage malignancies. Unfortunately, even when anti-VEGF therapy is effective, the benefit of therapy is short-lived, with the development of tumor growth. We now recognize the presence of numerous complementary and redundant pathways that regulate tumor vasculature. For example, VEGF/VEGFR and angiopoietin/Tie-2 axes are two redundant, complementary components regulating tumor angiogenesis and vascular maintenance. The current clinical challenge is to identify: (1) factors that predict efficacy, and (2) markers of tumor response to anti-VEGF therapy, which can be achieved only by developing a thorough understanding of the biology of the VEGF system and the role of complementary pathways that may mediate resistance to anti-VEGF therapy.


Subject(s)
Endothelial Cells/physiology , Neovascularization, Pathologic/physiopathology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/physiology , Drug Resistance, Neoplasm , Humans , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/physiology , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/physiology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Cancer Res ; 69(5): 1951-7, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19244128

ABSTRACT

5-Fluorouracil (5FU) and oxaliplatin are standard therapy for metastatic colorectal cancer (CRC), but the development of chemoresistance is inevitable. Because cancer stem cells (CSC) are hypothesized to be chemoresistant, we investigated CSC properties in newly developed chemoresistant CRC cell lines and sought to identify targets for therapy. The human CRC cell line HT29 was exposed to increasing doses of 5FU (HT29/5FU-R) or oxaliplatin (HT29/OxR) to achieve resistance at clinically relevant doses. Western blotting and flow cytometry were done to determine molecular alterations. The insulin-like growth factor-I receptor (IGF-IR) monoclonal antibody (mAb) AVE-1642 was used to inhibit signaling in vitro and in vivo using murine xenograft models. HT29/5FU-R and HT29/OxR showed 16- to 30-fold enrichment of CD133(+) cells and 2-fold enrichment of CD44(+) cells (putative CRC CSC markers). Resistant cells were enriched 5- to 22-fold for double-positive (CD133(+)/CD44(+)) cells. Consistent with the CSC phenotype, resistant cells exhibited a decrease in cellular proliferation in vitro (47-59%; P < 0.05). Phosphorylated and total IGF-IR levels were increased in resistant cell lines. HT29/5FU-R and HT29/OxR cells were approximately 5-fold more responsive to IGF-IR inhibition relative to parental cells (P < 0.01) in vitro. Tumors derived from HT29/OxR cells showed significantly greater growth inhibition in response to an IGF-IR mAb than did parental cells (P < 0.05). Chemoresistant CRC cells are enriched for CSC markers and the CSC phenotype. Chemotherapy-induced IGF-IR activation provided for enhanced sensitivity to IGF-IR-targeted therapy. Identification of CSC targets presents a novel therapeutic approach in this disease.


Subject(s)
Colorectal Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Receptor, IGF Type 1/antagonists & inhibitors , AC133 Antigen , Animals , Antigens, CD/analysis , Apoptosis/drug effects , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Glycoproteins/analysis , HT29 Cells , Humans , Hyaluronan Receptors/analysis , Male , Mice , Peptides/analysis , Phenotype , Receptor, IGF Type 1/analysis , Receptor, IGF Type 1/physiology , Signal Transduction
8.
Clin Cancer Res ; 14(24): 8052-60, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19088020

ABSTRACT

PURPOSE: Neuropilin-2 (NRP-2) is a coreceptor for vascular endothelial growth factor (VEGF) on endothelial cells. NRP-2 is overexpressed in pancreatic ductal adenocarcinoma (PDAC) cells relative to nonmalignant ductal epithelium. This study determined the role of NRP-2 in PDAC cells. EXPERIMENTAL DESIGN: NRP-2 expression was reduced in PDAC cells with stable short-hairpin RNA (shRNA) transfection. Western blotting was done to evaluate signaling intermediates. Migration and invasion studies were carried out in Boyden chambers. Anchorage-independent growth was assessed by soft-agar colony formation. In vivo growth was evaluated using murine subcutaneous and orthotopic xenograft models. Immunohistochemical analysis evaluated in vivo proliferation and angiogenesis. RESULTS: shRNA-NRP-2 decreased NRP-2 levels without affecting neuropilin-1 levels. Akt activation was decreased in clones with reduced NRP-2 (shRNA-NRP-2). shRNA-NRP-2 cells showed decreased migration, invasion, and anchorage-independent growth compared with control cells. In vitro proliferation rates were similar in control- and shRNA-transfected cells. Subcutaneous and orthotopic xenografts from shRNA-transfected cells were significantly smaller than those resulting from control-transfected cells (P < 0.05). Furthermore, shRNA-NRP-2 tumors exhibited less cellular proliferation and decreased microvascular area relative to control tumors (P < 0.05). Constitutive expression of the angiogenic mediator Jagged-1 was reduced in shRNA-NRP-2 cells, whereas vascular endothelial growth factor levels were unchanged. CONCLUSION: Reduction of NRP-2 expression in PDAC cells decreased survival signaling, migration, invasion, and ability to grow under anchorage-independent conditions. In vivo, reduction of NRP-2 led to decreased growth of xenograft tumors and decreased vascular area, which was associated with decreased Jagged-1 levels. NRP-2 is a potential therapeutic target on PDAC cells.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Neovascularization, Pathologic/etiology , Neuropilin-2/physiology , Pancreatic Neoplasms/pathology , Adenocarcinoma/blood supply , Calcium-Binding Proteins/analysis , Carcinoma, Pancreatic Ductal/blood supply , Cell Line, Tumor , Cell Movement , Humans , Intercellular Signaling Peptides and Proteins/analysis , Jagged-1 Protein , Membrane Proteins/analysis , Neoplasm Invasiveness , Neuropilin-2/analysis , Pancreatic Neoplasms/blood supply , Serrate-Jagged Proteins
9.
Clin Cancer Res ; 14(7): 1931-7, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18381930

ABSTRACT

Endoglin (CD105) is an accessory protein of the transforming growth factor-beta receptor system expressed on vascular endothelial cells. Mutation of the endoglin gene is associated with hereditary hemorrhagic telangiectasias, or Osler-Weber-Rendu syndrome, and has been studied extensively in the context of this disease. The expression of endoglin is elevated on the endothelial cells of healing wounds, developing embryos, inflammatory tissues, and solid tumors. Endoglin is a marker of activated endothelium, and its vascular expression is limited to proliferating cells. Recent studies identified endoglin expression in several solid tumor types, with the level of expression correlating with various clinicopathologic factors including decreased survival and presence of metastases. Attempts to target endoglin and the cells that express this protein in tumor-bearing mice have yielded promising results.


Subject(s)
Antigens, CD/physiology , Biomarkers, Tumor/analysis , Endothelial Cells/metabolism , Neoplasms/blood supply , Neoplasms/metabolism , Receptors, Cell Surface/physiology , Animals , Endoglin , Humans , Neovascularization, Pathologic/physiopathology , Transforming Growth Factor beta/metabolism
10.
Mol Carcinog ; 47(1): 34-46, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17626252

ABSTRACT

Transcriptional regulation via activator protein-1 (AP-1) protein binding to AP-1 binding sites within gene promoter regions of AP-1 target genes plays a key role in controlling cellular invasion, proliferation, and oncogenesis, and is important to pathogenesis of arthritis and cardiovascular disease. To identify new proteins that interact with the AP-1 DNA binding site, we performed the DNA affinity chromatography-based Nucleotide Affinity Preincubation Specificity TEst of Recognition (NAPSTER) assay, and discovered a 97 kDa protein that binds in vitro to a minimal AP-1 DNA sequence element. Mass spectrometric fragmentation sequencing determined that p97 is nucleolin. Immunoblotting of DNA affinity-purified material with anti-nucleolin antibodies confirmed this identification. Nucleolin also binds the AP-1 site in gel shift assays. Nucleolin interacts in NAPSTER with the AP-1 site within the promoter sequence of the metalloproteinase-13 gene (MMP-13), and binds in vivo in chromatin immunoprecipitation assays in the vicinity of the AP-1 site in the MMP-13 promoter. Overexpression of nucleolin in human HeLa cervical carcinoma cells significantly represses AP-1 dependent gene transactivation of a minimal AP-1 reporter construct and of an MMP-13 promoter reporter sequence. This is the first report of nucleolin binding and transregulation at the AP-1 site.


Subject(s)
DNA/metabolism , Matrix Metalloproteinase 13/genetics , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Transcription Factor AP-1/genetics , Transcriptional Activation , Adenocarcinoma , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , DNA/chemistry , DNA/genetics , Humans , Mass Spectrometry , Molecular Sequence Data , Peptide Fragments/chemistry , Plasmids , Protein Binding , Transcription Factor AP-1/metabolism , Nucleolin
11.
Biochim Biophys Acta ; 1769(9-10): 525-31, 2007.
Article in English | MEDLINE | ID: mdl-17822788

ABSTRACT

Matrix metalloproteinases (MMPs) are key enzymes that implement degradation of the extracellular matrix during cellular invasion in development, tissue remodeling, and pathogenic disease states. MMP-13 has pivotal roles in the pathogenesis of invasive cancers and arthritis. Here we report the identification of Y-box binding protein-1 (YB-1) as a new repressor of MMP-13 transactivation. YB-1 binds in vitro in DNA affinity chromatography to the activator protein-1 (AP-1) DNA sequence within the MMP-13 promoter. Chromatin immunoprecipitation assays reveal that YB-1 binds in living cells to the MMP-13 gene promoter to a region of the MMP-13 promoter containing the AP-1 site. YB-1 represses tumor promoter-induced MMP-13 promoter transactivation at the AP-1 site. This is the first report demonstrating YB-1 binding in vitro and in living cells to a mammalian AP-1 target gene, and the first report of YB-1 regulation of the MMP-13 promoter.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic/physiology , Matrix Metalloproteinase 13/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Response Elements/physiology , Transcription Factor AP-1/metabolism , Transcriptional Activation/physiology , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Matrix Metalloproteinase 13/genetics , Nuclear Proteins/genetics , Protein Binding , Repressor Proteins/genetics , Transcription Factor AP-1/genetics , Y-Box-Binding Protein 1
12.
Am J Pathol ; 170(5): 1594-606, 2007 May.
Article in English | MEDLINE | ID: mdl-17456765

ABSTRACT

Homeobox genes control developmental patterning and are increasingly being found to be deregulated in tumors. The DLX4 homeobox gene maps to the 17q21.3-q22 region that is amplified in some epithelial ovarian cancers. Because amplification of this region correlates with poor prognosis, we investigated whether DLX4 overexpression contributes to aggressive behavior of this disease. DLX4 was not detected in normal ovary and cystadenomas, whereas its expression in ovarian carcinomas was strongly associated with high tumor grade and advanced disease stage. Overexpression of DLX4 in ovarian cancer cells promoted growth in low serum and colony formation. Imaging of mice bearing intraperitoneal tumors revealed that DLX4 overexpression substantially increased tumor burden. Tumors that overexpressed DLX4 were more vascularized than vector-control tumors. Conditioned medium of DLX4-overexpressing tumor cells was more effective than medium conditioned by vector-control cells in stimulating endothelial cell growth. These observations were associated with the ability of DLX4 to induce expression of vascular endothelial growth factor as well as intracellular and secreted isoforms of fibroblast growth factor-2. Moreover, increased levels of these fibroblast growth factor-2 isoforms induced vascular endothelial growth factor expression in tumor cells. This study reveals a novel role for a homeobox gene in ovarian tumorigenicity by its induction of a proangiogenic, growth-stimulatory molecular program.


Subject(s)
Fibroblast Growth Factor 2/biosynthesis , Homeodomain Proteins/metabolism , Ovarian Neoplasms/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Apoptosis , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Genes, Homeobox , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection
13.
Mol Cell Endocrinol ; 245(1-2): 77-85, 2005 Dec 21.
Article in English | MEDLINE | ID: mdl-16298470

ABSTRACT

Border cell migration is a process that occurs during Drosophila ovarian development in which cells derived from a simple epithelium migrate and invade neighboring tissue. This process resembles the behavior of cancerous cells that derive from the simple epithelium of the human ovary. One important regulator of border cell migration is Taiman, a homolog of steroid receptor coactivator-3 (SRC-3). Because increasing evidence indicates that similarities exist between the molecular control of migration of border cells and of cancer cells, we investigated whether SRC-3 controls ovarian cancer cell migration. Little or no SRC-3 expression was detected in normal ovarian surface epithelium, ovarian cysts and borderline ovarian tumors that lack stromal invasion. In contrast, SRC-3 was abundantly expressed in high-grade ovarian carcinomas. Inhibiting SRC-3 expression in ovarian cancer cells markedly reduced cell spreading and migration, and altered intracellular localization of focal adhesion kinase. This inhibitory effect on cell migration was independent of the estrogen receptor (ER) status of the cells. These studies reveal a novel role for SRC-3 in ovarian cancer progression by promoting cell migration, independently of its role in estrogen receptor signaling.


Subject(s)
Acetyltransferases/physiology , Cell Movement , Oncogene Proteins/physiology , Ovarian Neoplasms/pathology , Trans-Activators/physiology , Acetyltransferases/chemistry , Acetyltransferases/genetics , Animals , Cell Line , Cell Line, Tumor , Drosophila Proteins , Female , Focal Adhesion Protein-Tyrosine Kinases/analysis , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/physiology , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases , Humans , Nuclear Receptor Coactivator 3 , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovary/cytology , Ovary/physiology , RNA, Small Interfering , Receptors, Estrogen/analysis , Receptors, Estrogen/physiology , Sequence Homology, Amino Acid , Signal Transduction/physiology , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factors/chemistry
14.
Eur J Cancer ; 41(16): 2428-37, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16199152

ABSTRACT

Homeobox genes encode transcription factors that play essential roles in controlling cell growth and differentiation during embryonic development. Many homeobox genes are aberrantly expressed in a wide variety of solid tumours, and their deregulation appears to enhance cell survival and proliferation and to inhibit differentiation. In hematologic malignancies, deregulated homeobox genes profoundly perturb self-renewal and proliferation of hematopoietic stem cells and progenitors. It is increasingly recognised that solid tumours, like hematologic malignancies, could arise from cancer stem cells, and that targeting these cells could be the most effective means of inhibiting tumour progression and disease recurrence. Studying the biological effects and mechanisms of homeobox genes in cancers could provide valuable insights into identifying cancer stem cells and targeting the self-renewal pathways in these cell populations.


Subject(s)
Genes, Homeobox/physiology , Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression/genetics , Genes, Homeobox/genetics , Genes, Tumor Suppressor/physiology , Humans , Stem Cells/physiology
15.
Biochem J ; 388(Pt 3): 921-8, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15702969

ABSTRACT

Involvement of the AP-1 (activator protein-1) transcription factor has been demonstrated previously in the regulation of cell proliferation and cell-cycle progression, in the control of cell migration, invasion and metastasis, and in signal transduction, stress responsiveness, DNA replication and DNA repair. YB-1 (Y-box-binding protein-1) has also been implicated in many of these processes. However, the mechanism by which YB-1 mediates these processes is poorly understood. In the present study, we report that overexpression of a transfected gene encoding YB-1 in human HeLa cervical carcinoma cells significantly represses the transactivation of a minimal AP-1 reporter construct in response to the tumour promoter PMA. YB-1 also represses mRNA expression and PMA-induced promoter transactivation of the endogenous AP-1 target gene encoding matrix metalloproteinase-12 (metalloelastase). YB-1 transrepression of both the minimal and matrix metalloproteinase-12 promoter reporter constructs is dependent on the AP-1 sequence. To identify new nuclear proteins that bind specifically to the AP-1 DNA-binding site, we devised a DNA-affinity-chromatography-based assay termed NAPSTER (nucleotide-affinity preincubation specificity test of recognition) and discovered a 49 kDa protein from human cancer cells that binds in a sequence-specific manner to the AP-1 DNA sequence. By tandem MS fragmentation sequencing analyses we determined that p49 is a YB-1. Immunoblotting of the NAPSTER-purified p49 protein using anti-YB-1 antibodies confirmed YB-1 binding to the AP-1 DNA sequence, as did gel mobility-supershift assays using YB-1 antibodies. This is the first report of YB-1 transrepression and interaction at the AP-1 DNA-binding site.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/genetics , DNA/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcriptional Activation/genetics , Base Sequence , Cell Nucleus , Chromatography, Affinity , DNA-Binding Proteins/genetics , Down-Regulation , Electrophoretic Mobility Shift Assay , Genes, Reporter/genetics , HT29 Cells , HeLa Cells , Humans , Matrix Metalloproteinase 12 , Metalloendopeptidases/genetics , Mutation/genetics , Nuclear Proteins , Protein Subunits , Protein Transport , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Substrate Specificity , Transcription Factor AP-1/chemistry , Transcription Factor AP-1/metabolism , Y-Box-Binding Protein 1
16.
Mol Carcinog ; 39(1): 34-60, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14694446

ABSTRACT

Relative expression levels of 9500 genes were determined by cDNA microarray analyses in mouse skin JB6 cells susceptible (P+) and resistant (P-) to 12-O-tetradecanoyl phorbol-13 acetate (TPA)-induced neoplastic transformation. Seventy-four genes in 6 functional classes were differentially expressed: (I) extracellular matrix (ECM) and basement membrane (BM) proteins (20 genes). P+ cells express higher levels than P- cells of several collagens and proteases, and lower levels of protease inhibitors. Multiple genes encoding adhesion molecules are expressed preferentially in P- cells, including six genes implicated in axon guidance and adhesion. (II) Cytoskeletal proteins (13 genes). These include actin isoforms and regulatory proteins, almost all preferentially expressed in P- cells. (III) Signal transduction proteins (12 genes). Among these are Ras-GTPase activating protein (Ras-GAP), the deleted in oral cancer-1 and SLIT2 tumor suppressors, and connexin 43 (Cx43) gap junctional protein, all expressed preferentially in P- cells. (IV) Interferon-inducible proteins (3 genes). These include interferon-inducible protein (IFI)-16, an Sp1 transcriptional regulator expressed preferentially in P- cells. (V) Other transcription factors (4 genes). Paired related homeobox gene 2 (Prx2)/S8 homeobox, and retinoic acid (RA)-regulated nur77 and cellular retinoic acid-binding protein II (CRABPII) transcription factors are expressed preferentially in P- cells. The RIN-ZF Sp-transcriptional suppressor exhibits preferential P+ expression. (VI) Genes of unknown functions (22 sequences). Numerous mesenchymal markers are expressed in both cell types. Data for multiple genes were confirmed by real-time PCR. Overall, 26 genes were newly implicated in cancer. Detailed analyses of the functions of the genes and their interrelationships provided converging evidence for their possible roles in implementing genetic programs mediating cancer susceptibility and resistance. These results, in conjunction with cell wounding and phalloidin staining data, indicated that concerted genetic programs were implemented that were conducive to cell adhesion and tumor suppression in P- cells and that favored matrix turnover, cell motility, and abrogation of tumor suppression in P+ cells. Such genetic programs may in part be orchestrated by Sp-, RA-, and Hox-transcriptional regulatory pathways implicated in this study.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , Genetic Predisposition to Disease , Interferons/pharmacology , Oligonucleotide Array Sequence Analysis , Animals , Biomarkers, Tumor/metabolism , Carcinogens/pharmacology , Cell Adhesion/physiology , Cell Movement/physiology , Gene Expression Profiling , Mice , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...