Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 468(4): 574-9, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26549231

ABSTRACT

Parvin-ß is an adaptor protein that binds to integrin-linked kinase (ILK) and is significantly downregulated in breast tumors and breast cancer cell lines. We treated the breast cancer cell line MCF7 with 24-methylenecycloartanyl ferulate (24-MCF), a γ-oryzanol compound. We observed upregulation of parvin-ß (GenBank Accession No. AF237769) and peroxisome proliferator-activated receptor (PPAR)-γ2 (GenBank Accession No. NM_015869). Among γ-oryzanol compounds, only treatment with 24-MCF led to the formation of reverse transcription-PCR products of parvin-ß (650 and 500 bp) and PPAR-γ2 (580 bp) in MCF7 cells, but not in T47D, SK-BR-3, or MDA-MB-231 cells. 24-MCF treatment increased the mRNA and protein levels of parvin-ß in MCF7 cells in a dose-dependent manner. We hypothesized that there is a correlation between parvin-ß expression and induction of PPAR-γ2. This hypothesis was investigated by using a promoter-reporter assay, chromatin immunoprecipitation, and an electrophoretic mobility shift assay. 24-MCF treatment induced binding of PPAR-γ2 to a peroxisome proliferator response element-like cis-element (ACTAGGACAAAGGACA) in the parvin-ß promoter in MCF7 cells in a dose-dependent manner. 24-MCF treatment significantly decreased anchorage-independent growth and inhibited cell movement in comparison to control treatment with dimethyl sulfoxide. 24-MCF treatment reduced the levels of GTP-bound Rac1 and Cdc42. Evaluation of Akt1 inhibition by 24-MCF revealed that the half maximal effective concentration was 33.3 µM. Docking evaluations revealed that 24-MCF binds to the ATP-binding site of Akt1(PDB ID: 3OCB) and the compound binding energy is -8.870 kcal/mol. Taken together, our results indicate that 24-MCF treatment increases parvin-ß expression, which may inhibit ILK downstream signaling.


Subject(s)
Actinin/metabolism , Breast Neoplasms/metabolism , Coumaric Acids/administration & dosage , PPAR gamma/metabolism , Phenylpropionates/administration & dosage , Humans , MCF-7 Cells , Up-Regulation/drug effects
2.
Mol Med Rep ; 8(3): 891-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23877734

ABSTRACT

Sorghum is a principal cereal food in a number of parts of the world and is critical in folk medicine in Asia and Africa. However, its effects on bone are unknown. Growth hormone (GH) is a regulator of bone growth and bone metabolism. GH activates several signaling pathways, including the Janus kinase (Jak)/signal transducer and activator of transcription (STAT) pathways, thereby regulating expression of genes, including insulin­like growth factor (IGF)­1. Bone morphogenetic proteins (BMPs) induce the differentiation of cells of the osteoblastic lineage, increasing the pool of IGF­1 target cells, the mature osteoblasts. In the present study, the effects of Hwanggeumchal sorghum extracts (HSE) on GH signaling via the Jak/STAT pathway in osteoblasts were investigated. HSE was not observed to be toxic to osteoblastic cells and increased the expression of BMP7 and GH­related proteins, including STAT5B, p­STAT5B, IGF­1 receptor (IGF-1R), growth receptor hormone (GHR) and Jak2 in MC3T3­E1 cells. In addition, HSE increased BMP7 and GHR mRNA expression in MC3T3­E1 cells. The expression of HSE­induced BMP7 and GHR was inhibited by AG490, a Jak2 kinase inhibitor. The observations indicate that HSE­induced signaling is similar to GH signaling via the GHR­Jak2 signaling axis. Using small interference RNA (siRNA) analysis, STAT5B was found to play an essential role in HSE­induced BMP7 and GH signaling in MC3T3­E1 cells. Results of the current study indicate that HSE promotes bone growth through activation of STAT5B.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Growth Hormone/metabolism , Janus Kinase 2/metabolism , Plant Extracts/toxicity , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Sorghum/metabolism , Animals , Bone Morphogenetic Protein 7/genetics , Cell Differentiation , Cell Line , Cell Lineage , Gene Expression/drug effects , Janus Kinase 2/antagonists & inhibitors , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Plant Extracts/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/genetics , Sorghum/chemistry , Tyrphostins/pharmacology
3.
PLoS One ; 7(10): e47477, 2012.
Article in English | MEDLINE | ID: mdl-23071812

ABSTRACT

Methylsulfonylmethane (MSM) is a naturally occurring sulfur compound with well-known anti-oxidant properties and anti-inflammatory activities. But, its effects on bone are unknown. Growth hormone (GH) is regulator of bone growth and bone metabolism. GH activates several signaling pathways such as the Janus kinase (Jak)/signal transducers and activators of transcription (STAT) pathway, thereby regulating expression of genes including insulin-like growth factor (IGF)-1. GH exerts effects both directly and via IGF-1, which signals by activating the IGF-1 receptor (IGF-1R). In this study, we investigated the effects of MSM on the GH signaling via the Jak/STAT pathway in osteoblasts and the differentiation of primary bone marrow mesenchymal stem cells (MSCs). MSM was not toxic to osteoblastic cells and MSCs. MSM increased the expression of GH-related proteins including IGF-1R, p-IGF-1R, STAT5b, p-STAT5b, and Jak2 in osteoblastic cells and MSCs. MSM increased IGF-1R and GHR mRNA expression in osteoblastic cells. The expression of MSM-induced IGF-1R and GHR was inhibited by AG490, a Jak2 kinase inhibitor. MSM induced binding of STAT5 to the IGF-1R and increased IGF-1 and IGF-1R promoter activities. Analysis of cell extracts by immunoprecipitation and Western blot showed that MSM enhanced GH-induced activation of Jak2/STAT5b. We found that MSM and GH, separately or in combination, activated GH signaling via the Jak2/STAT5b pathway in UMR-106 cells. Using siRNA analysis, we found that STAT5b plays an essential role in GH signaling activation in C3H10T1/2 cells. Osteogenic marker genes (ALP, ON, OCN, BSP, OSX, and Runx2) were activated by MSM, and siRNA-mediated STAT5b knockdown inhibited MSM-induced expression of osteogenic markers. Furthermore, MSM increased ALP activity and the mineralization of MSCs. Taken together, these results indicated that MSM can promote osteogenic differentiation of MSCs through activation of STAT5b.


Subject(s)
Dimethyl Sulfoxide/pharmacology , Growth Hormone/metabolism , Janus Kinase 2/metabolism , Osteogenesis/drug effects , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Sulfones/pharmacology , Analysis of Variance , Animals , Blotting, Western , Cell Differentiation/physiology , Cell Line, Tumor , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , Immunoprecipitation , Luciferases , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteoblasts/physiology , RNA, Small Interfering/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts , Thiazoles
4.
PLoS One ; 7(7): e40531, 2012.
Article in English | MEDLINE | ID: mdl-22792362

ABSTRACT

BACKGROUND: Cancer is one of the highly virulent diseases known to humankind with a high mortality rate. Breast cancer is the most common cancer in women worldwide. Sorghum is a principal cereal food in many parts of the world, and is critical in folk medicine of Asia and Africa. In the present study, we analyzed the effects of HSE in metastatic breast cancer. METHODOLOGY/PRINCIPAL FINDINGS: Preliminary studies conducted on MDA-MB 231 and MCF-7 xenograft models showed tumor growth suppression by HSE. Western blotting studies conducted both in vivo and in vitro to check the effect of HSE in Jak/STAT pathways. Anti-metastatic effects of HSE were confirmed using both MDA-MB 231 and MCF-7 metastatic animal models. These studies showed that HSE can modulate Jak/STAT pathways, and it hindered the STAT5b/IGF-1R and STAT3/VEGF pathways not only by down-regulating the expression of these signal molecules and but also by preventing their phosphorylation. The expression of angiogenic factors like VEGF, VEGF-R2 and cell cycle regulators like cyclin D, cyclin E, and pRb were found down-regulated by HSE. In addition, it also targets Brk, p53, and HIF-1α for anti-cancer effects. HSE induced G1 phase arrest and migration inhibition in MDA-MB 231 cells. The metastasis of breast cancer to the lungs also found blocked by HSE in the metastatic animal model. CONCLUSIONS/SIGNIFICANCE: Usage of HS as a dietary supplement is an inexpensive natural cancer therapy, without any side effects. We strongly recommend the use of HS as an edible therapeutic agent as it possesses tumor suppression, migration inhibition, and anti-metastatic effects on breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Janus Kinase 2/metabolism , Plant Extracts/pharmacology , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Sorghum/chemistry , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Plant Extracts/administration & dosage , Promoter Regions, Genetic , Protein Binding/drug effects , Receptor, IGF Type 1/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
5.
Int J Oncol ; 41(1): 161-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22552610

ABSTRACT

Hsp90α is a molecular chaperone protein involved in the structural maturation of oncogenic signaling proteins. Hsp90 was recently identified as an anticancer target; various studies are ongoing to find ways for managing cancer through Hsp90α. However, this approach is limited by reported side-effects. Hypoxia is a hallmark of solid tumors, including those of breast cancer and the extent of tumor hypoxia is associated with resistance to treatment and poor prognosis. One of the major signaling pathways in cancer cells, the Jak2/STAT5b pathway, has been found to be closely correlated with hypoxia. The objective of this study was to investigate the role of Jak2/STAT5b in the regulation of Hsp90α expression so that Hsp90α targeting can be achieved indirectly by modulating the Jak2/STAT5b pathway. We examined the role of the Jak2/STAT5b pathway in the expression of Hsp90α under hypoxic conditions by immunoblotting, reporter gene assays, EMSA and RNA interference analysis. With the help of in vivo models, we also analyzed the expression of Hsp90α in different parts of solid tumor tissues. We found a close association between hypoxic stress and Hsp90α expression. We also determined that STAT5b regulates the expression of Hsp90α during hypoxic stimulation. Under hypoxic conditions the expression of Hsp90α and STAT5b were proportional. siRNA analysis and nucleotide analysis showed that the promoter of Hsp90α has a STAT5b binding domain. Our work confirmed that STAT5b is one of the transcription factors that regulate Hsp90α. We, therefore, concluded that under hypoxic conditions, the Jak2/STAT5b pathway regulates Hsp90α expression and it could serve as a promising target for the treatment of solid tumors.


Subject(s)
Gene Expression Regulation, Neoplastic , HSP90 Heat-Shock Proteins/genetics , STAT5 Transcription Factor/metabolism , Up-Regulation , Animals , Base Sequence , Cell Hypoxia , Cell Line, Tumor , Consensus Sequence , Genes, Reporter , HSP90 Heat-Shock Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Promoter Regions, Genetic , Protein Binding , STAT5 Transcription Factor/genetics
6.
PLoS One ; 7(4): e33361, 2012.
Article in English | MEDLINE | ID: mdl-22485142

ABSTRACT

Breast cancer is the most aggressive form of all cancers, with high incidence and mortality rates. The purpose of the present study was to investigate the molecular mechanism by which methylsulfonylmethane (MSM) inhibits breast cancer growth in mice xenografts. MSM is an organic sulfur-containing natural compound without any toxicity. In this study, we demonstrated that MSM substantially decreased the viability of human breast cancer cells in a dose-dependent manner. MSM also suppressed the phosphorylation of STAT3, STAT5b, expression of IGF-1R, HIF-1α, VEGF, BrK, and p-IGF-1R and inhibited triple-negative receptor expression in receptor-positive cell lines. Moreover, MSM decreased the DNA-binding activities of STAT5b and STAT3, to the target gene promoters in MDA-MB 231 or co-transfected COS-7 cells. We confirmed that MSM significantly decreased the relative luciferase activities indicating crosstalk between STAT5b/IGF-1R, STAT5b/HSP90α, and STAT3/VEGF. To confirm these findings in vivo, xenografts were established in Balb/c athymic nude mice with MDA-MB 231 cells and MSM was administered for 30 days. Concurring to our in vitro analysis, these xenografts showed decreased expression of STAT3, STAT5b, IGF-1R and VEGF. Through in vitro and in vivo analysis, we confirmed that MSM can effectively regulate multiple targets including STAT3/VEGF and STAT5b/IGF-1R. These are the major molecules involved in tumor development, progression, and metastasis. Thus, we strongly recommend the use of MSM as a trial drug for treating all types of breast cancers including triple-negative cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Dimethyl Sulfoxide/pharmacology , Down-Regulation/drug effects , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Sulfones/pharmacology , Animals , Apoptosis/drug effects , COS Cells , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Female , Gene Expression , Humans , Inhibitory Concentration 50 , Mice , Promoter Regions, Genetic , Protein Binding , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , STAT3 Transcription Factor/genetics , STAT5 Transcription Factor/genetics , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
7.
Int J Oncol ; 36(5): 1243-51, 2010 May.
Article in English | MEDLINE | ID: mdl-20372799

ABSTRACT

Cyclin D1 and insulin-like growth factor 1 receptor (IGF-1R) are key regulators of cell proliferation that are overexpressed in most breast cancers. The purpose of the present study was to investigate the molecular mechanism by which hemin exerts its inhibitory effects on aggressive breast cancer cells. We found that hemin regulates cyclin D1 and IGF-1R proteins and insulin-like growth factor-1 gene expression through STAT5b in breast cancer cells. We confirmed that STAT5b, cyclin D1, and IGF-1R is up-regulated by hypoxia, and the increased STAT5b binds strongly to the STAT5-binding sites contained within the distal 5'-flanking region of IGF-1 gene in breast cancer cells. EMSA studies showed that STAT5 binding activity to the IGF-1 and cyclin D1 promoter was distinctly decreased by hemin in STAT5b-transfected COS-7 or MDA-MB 231 cells. IGF-1 gene expression was also decreased by hemin in mammary epithelial cells. STAT5b expression was inhibited in siRNA experiments and by hemin, leading to decreased levels of IGF-1. These results provide a basis for molecular targets in cancer treatment via the STAT5b/IGF-1 or /cyclin D1 pathway in solid tumor cells. These data indicate that hemin inhibits the cyclin D1 and IGF-1 expression via STAT5b under hypoxia in ERalpha-negative breast cancer cells. These findings are valuable toward understanding the role of hemin-induced inhibition of cyclin D1 and IGF-1 expression under hypoxia in invasive and metastatic breast cancer.


Subject(s)
Cyclin D1/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Hemin/metabolism , Hypoxia , Insulin-Like Growth Factor I/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Animals , CHO Cells , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cricetinae , Cricetulus , Gene Expression Regulation, Neoplastic , Hemin/chemistry , Humans , Neoplasm Invasiveness , Neoplasm Metastasis
8.
Int J Oncol ; 33(3): 477-84, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18695876

ABSTRACT

We have shown previously that hypoxia activates the cyclin D1 promoter via the Jak2/STAT5b pathway in breast cancer cells. Most solid tumors contain hypoxic components and overexpression of cyclin D1. The purpose of the present study was to investigate the molecular mechanism by which momilactone B exerts its inhibitory effects on breast cancer cells. Momilactone B, extracted from Korean rice hulls, suppressed hypoxia-induced increases in phospho-STAT5, STAT5b, cyclin D1, and cdk4 protein levels in human breast cancer cells. STAT5b expression was inhibited by siRNA experiments leading to decreased cyclin D1. The effects of momilactone B on cell growth and apoptosis-related gene expression were investigated in breast cancer cells under hypoxic conditions (2% O2). Bax and p21 expression was found to be up-regulated, whereas ppRb and bcl-2 were down-regulated in momilactone B-treated cells under hypoxic conditions. However, the p53 protein level did not change. Flow cytometry with Annexin-FITC staining showed that the number of apoptotic cells increased in hypoxic cells treated with momilactone B compared with untreated hypoxic cells. Furthermore, caspase activity increased upon treatment with momilactone B under hypoxic conditions. These results indicate that momilactone B inhibits the growth of breast cancer cells, regulates the expression of apoptosis-related genes, and induces apoptosis through STAT5b and a caspase-3 dependent pathway. We suggest that momilactone B accelerates hypoxia-induced apoptosis of human breast cancer cells through STAT5b, and may represent an effective chemopreventive or therapeutic agent against breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Hypoxia/drug effects , Diterpenes/pharmacology , Lactones/pharmacology , STAT5 Transcription Factor/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Electrophoretic Mobility Shift Assay , Female , Flow Cytometry , Gene Expression/drug effects , Humans , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering , STAT5 Transcription Factor/metabolism , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
9.
Biochem Biophys Res Commun ; 358(3): 733-8, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17509524

ABSTRACT

Insulin-like growth factors (IGF), polypeptides that regulate growth, differentiation, and survival in cells and tissues, were found to enhance gene expression from both heterologous and homologous promoters in the presence of constitutively active STAT5. This highly conserved 700-bp DNA region contains two closely located consensus STAT5-binding sites. Hypoxia regulates the IGF-1 gene expression through the STAT5b. We confirmed STAT5b is up-regulated under hypoxic conditions, and the increased STAT5b binds strongly to the STAT5-binding sites 1 and 2 contained within the distal 5'-flanking region of IGF-1 gene in HepG2 cells. EMSA studies showed that STAT5-binding activities to the IGF-1 promoter distinctly increased under hypoxia in STAT5b-transfected COS-7 cells. The IGF-1 gene expression was also increased by hypoxia in HepG2 cells. STAT5b expression was inhibited by siRNA experiments leading to decreased IGF-1. These results provide a basis of molecular targets for cancer treatment via the STAT5b-IGF-1 pathway in solid tumor cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Hypoxia , Insulin-Like Growth Factor I/metabolism , STAT5 Transcription Factor/physiology , Animals , Binding Sites , COS Cells , Cell Line , Chlorocebus aethiops , Gene Expression Regulation , Genetic Vectors , Humans , Oxygen/metabolism , RNA Interference , RNA, Small Interfering/metabolism , STAT5 Transcription Factor/genetics
10.
Breast ; 15(2): 187-95, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16084091

ABSTRACT

Several constitutively activated signal transducers and activators of transcription (STAT) proteins have been observed in a wide number of human cancer cell lines and primary tumors. Normal cells maintain normoxic conditions but tumor cells are characteristically hypoxic. We studied the altered activation and tyrosine phosphorylation of STATs under hypoxic conditions (2% O2) or desferrioxamine (DFO) treatment in mouse mammary epithelial cells (HC11) and a human breast cancer cell line (MCF-7). STAT1, -3 and -5 proteins are especially important and are observed at elevated levels in tumorigenesis. We also investigated the serine phosphorylation of STAT1, -3, and -5 under hypoxic conditions or DFO treatment in HC11 and MCF-7 cells. Here we show that DFO or hypoxia stimulates the tyrosine and/or serine phosphorylation and the expression of STAT proteins in breast cancer cells. Our data suggest that DFO or hypoxic condition is a critical stimulator for the activation of STAT proteins in breast cancer cells. These results may provide the basis for identifying another mechanism of breast tumorigenesis via the JAK/STAT pathway in hypoxia. Also, activation of STAT proteins by hypoxia may play an important role in the physiological phenomenon of embryonic stem cells and old cells with hypoxic conditions.


Subject(s)
Breast Neoplasms/metabolism , STAT Transcription Factors/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Deferoxamine/pharmacology , Female , Humans , Mammary Glands, Animal/metabolism , Mice , Phosphorylation/drug effects , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism
11.
Exp Mol Med ; 37(4): 353-64, 2005 Aug 31.
Article in English | MEDLINE | ID: mdl-16155412

ABSTRACT

Hypoxia, a common consequence of solid tumor growth in breast cancer or other cancers, serves to propagate a cascade of molecular pathways which include angiogenesis, glycolysis, and various cell-cycle control proteins. As we have shown previously, hypoxia activates STAT5 (signal transducer and activator of transcription 5) and increases its binding activity to the GAS element in mammary epithelial cells. In this study we attempted to elucidate the mechanism by which cyclin D1 is regulated by the STAT5 protein under hypoxic conditions. Our data demonstrate that hypoxia (2% O(2)) or desferrioxamine (DFO) induces tyrosine and serine phosphorylation of STAT5 in human breast cancer cells (MCF-7) and mammary epithelial cells (HC11). Imunoprecipitation and subsequent Western analysis showed that Jak2 leads to the tyrosine phosphorylation and activation of STAT5a or STAT5b under hypoxic conditions. Using a transfected COS-7 cell model system, we demonstrate that the activity of a cyclin D1 promoter-luciferase construct increased under hypoxic conditions or DFO treatment. The activity of the STAT5b/cyclin D1 promoter increased significantly by 12 h of hypoxia, whereas the activity of the STAT5a/cyclin D1 promoter was unaffected under hypoxic conditions. These increases in promoter activity are predominantly mediated by the Jak2/STAT5b signaling pathway. We have shown by EMSA that hypoxia induces STAT5 to bind to the cyclin D1 promoter (GAS-1) in MCF-7 and HC11 cells. These data suggest that STAT5b may mediate the transcriptional activation of cyclin D1 after hypoxic stimulation.


Subject(s)
Breast Neoplasms/genetics , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Anaerobiosis/genetics , Animals , Breast Neoplasms/metabolism , COS Cells , Cell Hypoxia/genetics , Chlorocebus aethiops , Deferoxamine/pharmacology , Female , Humans , Janus Kinase 2 , Phosphorylation/drug effects , Promoter Regions, Genetic , Serine/metabolism , Tumor Cells, Cultured , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...