Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Int J Biochem Cell Biol ; 45(8): 1921-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23732111

ABSTRACT

Osmoregulation is an essential mechanism for euryhaline fish. Gill cells undergo rapid mechanism to maintain the cellular homeostasis during osmotic stress. Reports have suggested that gill cells may be able to migrate between primary filament and secondary lamella during seawater acclimination. However, the factor that can trigger such process is not well-known. Previously, we identified the osmotic stress transcription factor 1b (Ostf1b) in medaka and found that it is an early hypertonic responsive gene and can activate the c-Jun N-terminal kinase (JNK) pathway. In this report, we aim to know if Ostf1b plays the role in the migration. Ostf1b was ectopic expressed in the human embryonic kidney cell line (HEK293) to understand the Ostf1b function. Results clearly demonstrated that Ostf1b could constitutively activate the Rho kinase 1 (ROCK1) and myosin light chain 2 (MLC2) signalling pathway that promotes cell migration, epithelial mesenchymal transition (EMT) and cytoskeletal dynamics through stress fibre formation. The study supports the notion of cell migration and cytoskeleton rearrangement theories in osmoregulation.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Transcription Factors/metabolism , Cardiac Myosins/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Myosin Light Chains/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Stress Fibers/metabolism , Tight Junction Proteins/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
2.
Mol Cell Endocrinol ; 374(1-2): 73-81, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23664860

ABSTRACT

The members of stanniocalcins (STCs: STC-1 and STC-2) family are known to be involved in tumor progression and metastasis. Although current evidences suggest the involvement of STCs in vascular biology, the functional roles of STCs in angiogenesis have not yet been elucidated. The objective of this study was to decipher the roles of STCs in angiogenesis of human umbilical vascular endothelial cells (HUVECs). We prepared STC1 or STC2 lentiviral particles to transduce the cells to reveal their effects on the processes of cell proliferation, migration and tube formation. The stimulatory effects of STCs on these processes were demonstrated, supporting the notion of STCs in angiogenesis. To dissect the molecular components involved, STC1 or STC2 transduction led to significant increases in the expression levels of cell cycle regulators (i.e. cyclin-D and phospho-retinoblastoma), matrix metalloproteinase (MMP)-2 but a decrease of tissue inhibitors of metalloproteases (TIMP)-1. The expression levels of the cell adhesion/junctional proteins vimentin and VE-cadherin, were significantly induced. Moreover the transduction induced both mRNA and protein levels of eNOS, VEGF and VEGFR2 (KDR mRNA and pKDR), highlighting the stimulatory effects of STCs on VEGF-signaling pathway. Furthermore STC2 transduction but not STC1, activated angiopoietin (Ang)-2 pathway. Taken together, STC1 and STC2 play positive roles in angiogenic sprouting. The action of STC1 was mediated via VEGF/VEGFR2 pathway while STC2 were mediated via VEGF/VEGFR2 and Ang-2 pathways.


Subject(s)
Angiopoietin-2/genetics , Glycoproteins/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Angiopoietin-2/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Movement , Cell Proliferation , Cyclin D/genetics , Cyclin D/metabolism , Gene Expression Regulation , Genetic Vectors , Glycoproteins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lentivirus/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Phosphorylation , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vimentin/genetics , Vimentin/metabolism
3.
Chemosphere ; 85(2): 277-83, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21705041

ABSTRACT

Perfluorinated compounds (PFCs) are man-made fluoro-surfactants that are identified as global pollutants and can pose health risks to humans and wildlife. Two aspects of risk assessment were conducted in this study, including exposure and response. Exposure was estimated by using the concentrations of PFCs in fish and applying standard exposure factors. Among different PFCs, PFOS, PFOA, PFNA, PFDA, PFUdA and PFTrDA were detected. Total concentrations of PFC in fish ranged from 0.27-8.4 ng g(-1) to 0.37-8.7 ng g(-1) respectively in Hong Kong and Xiamen. The calculated hazard ratio (HR) of PFOS for all fish was less than 1.0. However, the HR for mandarin fish in Hong Kong and bighead carp, grass carp and tilapia in Xiamen, had HR values of approximately 0.5, indicating that frequent consumption of these 4 more contaminated fish species might pose an unacceptable risk to human health. Our data support the notion that the released/disposed chemical pollutants into water systems make fish a source of environmental toxicants to humans. The risks and potential effects of PFCs to health of coastal population in the Pearl River Delta are of concern.


Subject(s)
Fishes , Fluorocarbons/analysis , Food Contamination , Fresh Water/chemistry , Water Pollutants, Chemical/analysis , Alkanesulfonic Acids/analysis , Alkanesulfonic Acids/toxicity , Animals , Aquatic Organisms , Carps , Fluorocarbons/toxicity , Hong Kong , Humans , Risk Assessment , Tilapia
4.
J Cell Biochem ; 112(8): 2089-96, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21465530

ABSTRACT

Our previous study demonstrated that, stanniocalcin-1 (STC1) was a target of histone deacetylase (HDAC) inhibitors and was involved in trichostatin A (TSA) induced apoptosis in the human colon cancer cells, HT29. In this study, we reported that the transcriptional factor, specificity protein 1 (Sp1) in association with retinoblastoma (Rb) repressed STC1 gene transcription in TSA-treated HT29 cells. Our data demonstrated that, a co-treatment of the cells with TSA and Sp1 inhibitor, mithramycin A (MTM) led to a marked synergistic induction of STC1 transcript levels, STC1 promoter (1 kb)-driven luciferase activity and an increase of apoptotic cell population. The knockdown of Sp1 gene expression in TSA treated cells, revealed the repressor role of Sp1 in STC1 transcription. Using a protein phosphatase inhibitor okadaic acid (OKA), an increase of Sp1 hyperphosphorylation and so a reduction of its transcriptional activity, led to a significant induction of STC1 gene expression. Chromatin immunoprecipitation (ChIP) assay revealed that Sp1 binding on STC1 proximal promoter in TSA treated cells. The binding of Sp1 to STC1 promoter was abolished by the co-treatment of MTM or OKA in TSA-treated cells. Re-ChIP assay illustrated that Sp1-mediated inhibition of STC1 transcription was associated with the recruitment of another repressor molecule, Rb. Collectively our findings identify STC1 is a downstream target of Sp1.


Subject(s)
Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glycoproteins/biosynthesis , Hydroxamic Acids/pharmacology , Protein Synthesis Inhibitors/pharmacology , Repressor Proteins/metabolism , Sp1 Transcription Factor/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/genetics , Glycoproteins/genetics , Humans , Okadaic Acid/pharmacology , Phosphorylation/drug effects , Phosphorylation/genetics , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Sp1 Transcription Factor/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
5.
Exp Cell Res ; 316(20): 3425-34, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20619259

ABSTRACT

The human glycoprotein, stanniocalcin-2 (STC2) is a HIF-1 target gene that is found to be associated with tumor development. The relationship of the prognostic outcome to the level of its expression in cancer tissues is controversial; however experimental evidence suggests that STC2 is a positive regulator of cancer progression. In the present study, we investigated if the expression of STC2 in hypoxic cells is associated with cancer invasion and metastasis. We studied the epithelial-mesenchymal transition (EMT) markers in STC2-silenced and over-expressed SKOV3 cells maintained in hypoxic condition. Western blot and immunocytochemical analysis revealed that the stable expression of exogenous STC2 promoted EMT, as revealed by the increase of N-cadherin/vimentin but a decrease of E-cadherin levels. This observation was further confirmed by colony formation assay where the STC2 stably transfected cells showed high degree of motility with fibroblast morphology under hypoxic condition. In conducting invasion assay in hypoxia, the STC2 stably transfected cells showed high degree of invasiveness. This observation was correlated with the significant increase of MMP2 and MMP9 expression in the STC2 stably transfected cells. In HUVEC/SKOV3 co-culture invasion study, endothelial invasion was found to be enhanced by the seeding of STC2 stably transfected cells in the lower compartment. These observations were possibly mediated by an increase of ROS and activated ERK1/2 levels in the cells. Collectively, the finding provides the first evidence that STC2 is a positive regulator in tumor progression at hypoxia.


Subject(s)
Cell Hypoxia/physiology , Epithelial-Mesenchymal Transition , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Invasiveness , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Coculture Techniques , Endothelial Cells/cytology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression/genetics , Glycoproteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/genetics , Phosphorylation , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Transfection , Vimentin/metabolism
6.
Exp Cell Res ; 316(3): 466-76, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19786016

ABSTRACT

Stanniocalcin-2 (STC2), the paralog of STC1, has been suggested as a novel target of oxidative stress response to protect cells from apoptosis. The expression of STC2 has been reported to be highly correlated with human cancer development. In this study, we reported that STC2 is a HIF-1 target gene and is involved in the regulation of cell proliferation. STC2 was shown to be up-regulated in different breast and ovarian cancer cells, following exposure to hypoxia. Using ovarian cancer cells (SKOV3), the underlying mechanism of HIF-1 mediated STC2 gene transactivation was characterized. Hypoxia-induced STC2 expression was found to be HIF-1alpha dependent and required the recruitment of p300 and HDAC7. Using STC2 promoter deletion constructs and site-directed mutagenesis, two authentic consensus HIF-1 binding sites were identified. Under hypoxic condition, the silencing of STC2 reduced while the overexpression of STC2 increased the levels of phosphorylated retinoblastoma and cyclin D in both SKOV3 and MCF7 cells. The change in cell cycle proteins correlated with the data of the serial cell counts. The results indicated that cell proliferation was reduced in STC2-silenced cells but was increased in STC2-overexpressing hypoxic cells. Solid tumor progression is usually associated with hypoxia. The identification and functional analysis of STC2 up-regulation by hypoxia, a feature of the tumor microenvironment, sheds light on a possible role for STC2 in tumors.


Subject(s)
Glycoproteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Proliferation/drug effects , Disulfides/pharmacology , E1A-Associated p300 Protein/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycoproteins/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Indole Alkaloids/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Luciferases/metabolism , Molecular Sequence Data , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Response Elements/genetics , Time Factors
7.
Exp Cell Res ; 314(8): 1823-30, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18394600

ABSTRACT

Mammalian stanniocalcin-2 (STC2) is a secreted glycoprotein hormone with a putative role in unfolded protein response and apoptosis. Here we reported that STC2 expression was sporadically abrogated in human cancer cells by transcriptional silencing associated with CpG island promoter hypermethylation. Direct sequencing of bisulfite-modified DNA from a panel of seven human cancer cell lines revealed that CpG dinucleotides in STC2 promoter was methylated in human ovarian epithelial cancer (SKOV3, OVCAR3 and CaOV3), pancreatic cancer (BxP3), colon adenoma (HT29), and leukemia (Jurkat cells). STC2 CpG island hypermethylation was accompanied with a low basal STC2 expression level. Treatment of these cancer cells with 5-aza-2'-deoxycytidine (5-aza-CdR), an inhibitor of DNA methylation significantly induced STC2 expression. Using SKOV3 cells as a model, the link between DNA demethylation and STC2 expression was consistently demonstrated with hydralazine treatment, which was shown to reduce the protein level of DNA methyltransferase 1 (DNMT1) but stimulated STC2 expression. Two human normal surface ovarian cell-lines (i.e. IOSE 29 and 398) showed no methylation at CpG dinucleotides in the examined promoter region and were accompanied with high basal STC2 levels. Hypoxia stimulated STC2 expression in SKOV3 cells was markedly increased in 5-aza-CdR pretreated cells, showing that DNA methylation may hinder the HIF-1 mediated activation. To elucidate this possibility, RNA interference studies confirmed that endogenous HIF-1 alpha was a key factor for STC2 gene activation as well as in the synergistic induction of STC2 expression in 5-aza-CdR pretreated cells. Chromatin immunoprecipitation (ChIP) assay demonstrated the binding of HIF-1 alpha to STC2 promoter. The binding was increased in 5-aza-CdR pretreated cells. Collectively, this is the first report to show that STC2 was aberrantly hypermethylated in human cancer cells. The findings demonstrated that STC2 epigenetic inactivation may interfere with HIF-1 mediated activation of STC2 expression.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Silencing , Glycoproteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Cell Hypoxia , Cell Line, Tumor , CpG Islands , DNA Methylation , Decitabine , Glycoproteins/metabolism , Humans , Hydralazine/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , RNA Interference , Transcriptional Activation
8.
Biochem Biophys Res Commun ; 356(4): 968-75, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17395153

ABSTRACT

There is growing evidence to suggest that altered patterns of STC1 gene expression relate to the process of human cancer development. Our previous study has demonstrated the involvement of HIF-1 in the regulation of STC1 expression in human cancer cells. Recently, STC1 has been implicated as a putative pro-apoptotic factor in regulating the cell-death mechanism. Thus it would be of interest to know if STC1 is regulated by a tumor suppressor protein, p53. In this study, we provide evidence to demonstrate that the induction of STC1 expression in apoptotic human nasopharyngeal cancer cells (CNE2) is mediated by the activation of p53. Our study indicated that the activation of STC1 and heat-shock protein (hsp70) accompanied iodoacetamide (IDAM)-induced apoptosis in CNE-2. In addition, cellular events such as GSH depletion, mitochondrial membrane depolarization, reduction of pAkt and procaspase-3, and the induction of total p53 protein, acetylated p53, and annexin V positive cells were observed. The activation of STC1 was found to be at the transcriptional level and was independent of prior protein synthesis. Co-treatment of IDAM exposed cells with N-acetyl cysteine (NAC) prevented cell death by restoring mitochondrial membrane potential and cellular levels of GSH. NAC co-treatment also suppressed STC1 expression but had no effect on IDAM-induced hsp70 expression. RNA interference studies demonstrated that endogenous p53 was involved in activating STC1 gene expression. Collectively, the present findings provide the first evidence of p53 regulation of STC1 expression in human cancer cells.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...