Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Cell Res ; 19(4): 487-96, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19308091

ABSTRACT

MicroRNAs (miRNAs), which are small noncoding RNA molecules, play important roles in the post-transcriptional regulation process. The microRNA-21 gene (miR-21) has been reported to be highly expressed in various solid tumors, including breast cancer. Bone morphogenetic protein-6 (BMP-6) has been identified as an inhibitor of breast cancer epithelial-mesenchymal transition (EMT) through rescuing E-cadherin expression. We initiated experiments to identify the relationships between miR-21 and BMP-6 in breast cancer progression. Real-time PCR analysis showed that miR-21 expression was very high in MDA-MB-231 cells that expressed little BMP-6. A reverse correlation between BMP-6 and miR-21 was also determined in breast cancer tissue samples. Moreover, BMP-6 inhibited miR-21 transcription in MDA-MB-231 cells. In order to investigate how BMP-6 inhibited the miR-21 promoter (miPPR-21), we constructed a series of miPPR-21 reporters. Luciferase assay results indicated that BMP-6 inhibited miPPR-21 activity through the E2-box and AP-1-binding sites. We also demonstrated that both deltaEF1 and TPA induced miR-21 expression. Using site-directed mutation and CHIP assay, we found that deltaEF1 induced miPPR-21 activity by binding to the E2-box on miPPR-21. Moreover, TPA triggered miPPR-21 activity through the AP-1 binding sites. BMP-6 treatment significantly reduced the binding of these factors to miPPR-21 by decreasing the expression of deltaEF1 and c-Fos/c-Jun. We also demonstrated that BMP-6-induced downregulation of miR-21 modified the activity of PDCD4 3'UTR and inhibited MDA-MB-231 cell invasion. deltaEF1 overexpression and TPA induction blocked this inhibitory effect of BMP-6. In conclusion, BMP-6-induced inhibition of miR-21 suggests that BMP-6 may function as an anti-metastasis factor by a mechanism involving transcriptional repression of miR-21 in breast cancer.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Breast Neoplasms/genetics , Homeodomain Proteins/antagonists & inhibitors , MicroRNAs/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Bone Morphogenetic Protein 6/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Down-Regulation , Homeodomain Proteins/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/pharmacology , Mutagenesis, Site-Directed , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Tetradecanoylphorbol Acetate/chemistry , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Transfection , Zinc Finger E-box-Binding Homeobox 1
2.
Free Radic Biol Med ; 46(9): 1275-82, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19245827

ABSTRACT

Oxidative stress is involved in a variety of kidney diseases, and heme oxygenase 1 (HO-1) induction is a protective response to oxidative stress. Downregulation of bone morphogenetic protein 6 (BMP6) is associated with renal damage in intrauterine growth-restricted newborns. However, it is unknown whether BMP6 has a renoprotective effect or HO-1 induction property. In this study, we demonstrate that BMP6 effectively protects renal proximal tubule cells (HK-2) against hydrogen peroxide (H(2)O(2))-induced cell injury. BMP6 also increased HO-1 gene expression and activity of HO. Inhibition of de novo gene expression, the HO inhibitor ZnPPIX, HO-1 knockdown, or the carbon monoxide (CO) scavenger hemoglobin attenuated the cytoprotective effect of BMP6, whereas HO-1 constitutive expression, the HO-1 inducer hemin, or the hemin metabolites bilirubin and CO ameliorated H(2)O(2)-induced cell injury. Stimulation of HK-2 cells with BMP6 activated Smad signaling but not mitogen-activated protein kinases. In addition, BMP6-mediated induction of HO-1 expression and increase in HO activity were inhibited by Smad5 knockdown. Furthermore, deletion or mutation of the Smad-binding element in the HO-1 promoter also inhibited BMP6-induced luciferase activity. In summary, these findings suggest that induction of HO-1 through a Smad-dependent mechanism is responsible for the cytoprotective effect of BMP6 in H(2)O(2)-mediated renal cell injury.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Enzyme Induction/genetics , Heme Oxygenase-1/genetics , Kidney Tubules, Proximal/metabolism , Smad5 Protein/metabolism , Bone Morphogenetic Protein 6/genetics , Cell Line, Transformed , Cell Survival/drug effects , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Enzyme Induction/drug effects , Feedback, Physiological , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/biosynthesis , Humans , Hydrogen Peroxide/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Oxidative Stress , Protein Synthesis Inhibitors/pharmacology , RNA, Small Interfering/genetics , Response Elements/genetics , Smad5 Protein/genetics , Transfection , Transgenes
3.
Zhonghua Xue Ye Xue Za Zhi ; 29(5): 325-8, 2008 May.
Article in Chinese | MEDLINE | ID: mdl-18844071

ABSTRACT

OBJECTIVE: To identify the interaction partners of a new splicing product of LMO2 gene (LMO2-C), and study its function in K562 cells. METHODS: Maltose binding protein (MBP) pull down and mammalian two-hybrid assay (MTHA) were used to identify the interaction partners of LMO2-C in K562 cells. Semiquantitative RT-PCR was used to detect the expression of hematopoietic specific gene glycoprotein (GPA) in K562 cells. RESULTS: MBP-LMO2-C fusion protein was expressed and purified in soluble form successfully. Endogenous GATA1 and LDB1 proteins were confirmed to bind to LMO2-C by MBP pull down analysis. The MTHA also showed that LMO2-C had comparable binding affinities to LDB1 with LMO2-L, and over expression of LMO2-C prevented LMO2-L from binding to LDB1, the inhibition rate being (81.13 +/- 0.68)%. RT-PCR results showed that the expression level of GPA was reduced [(51.00 +/- 1.58)%] in K562 cells while LMO2-C overexpressed. CONCLUSION: LMO2-C can bind endogenous GATA1 and LDB1 protein in K562 cells and down regulates the expression of GPA.


Subject(s)
DNA-Binding Proteins/metabolism , Metalloproteins/metabolism , RNA Splicing , Adaptor Proteins, Signal Transducing , DNA-Binding Proteins/genetics , GATA1 Transcription Factor/metabolism , Humans , K562 Cells , LIM Domain Proteins , Maltose-Binding Proteins , Metalloproteins/genetics , Periplasmic Binding Proteins , Proto-Oncogene Proteins , Transcription Factors/metabolism , Two-Hybrid System Techniques
4.
Sheng Wu Gong Cheng Xue Bao ; 24(5): 862-6, 2008 May.
Article in Chinese | MEDLINE | ID: mdl-18724709

ABSTRACT

Bone morphogenetic protein 2 (BMP2), which belongs to the transforming growth factor-beta (TGF-beta) superfamily, is a multifunctional molecule with distinct abilities to induce bone formation. BMP2 has been identified to have eminent pharmaceutical importance for clinical application. We previously constructed stable cell line in Chinese hamster ovary cells (CHO) that highly expressed recombinant human BMP2 (rhBMP2). For large-scale production of the recombinant protein used in clinical application, it is critical to have both high expression and stability of the protein. In the present study, the stability of the cell line (rCHO(hBMP2)-C8) with the highest expression, as well as the stability of rhBMP2 protein were investigated systematically. We cultured the rCHO (hBMP2)-C8 cell line in the presence or absence of MTX for two months, the cell growth and rhBMP2 production characteristics were examined during the culture; we found the duration that the rCHO(hBMP2)-C8 cell line could secret rhBMP2 continually into the serum-free medium. Moreover, we detected the temperature sensitivity of rhBMP2 in culture medium. This study will contribute to our understanding for further producing rhBMP2 by large-scale culture technology.


Subject(s)
Bone Morphogenetic Protein 2/biosynthesis , Cell Culture Techniques/methods , Recombinant Proteins/biosynthesis , Animals , Bone Morphogenetic Protein 2/genetics , CHO Cells , Cricetinae , Cricetulus , Culture Media , Genetic Vectors/genetics , Humans , Recombinant Proteins/genetics
5.
Sheng Wu Gong Cheng Xue Bao ; 24(5): 887-91, 2008 May.
Article in Chinese | MEDLINE | ID: mdl-18724713

ABSTRACT

Pulldown assay is an in vitro method for studies of protein-protein interactions, in which tagged proteins are usually expressed as the bait to enrich other proteins that could bind to them. In this technology, the GST tag is broadest used for its modest size and hydrophilic property. In most cases, the GST tag could increase the hydrophility of the fusion protein and help to avoid the formation of inclusion bodies. However, in the other few cases, the target protein may be strongly hydrophobic or have complicated structures that were hard to fold and assemble in correct conformations without champerons, and even the existence of GST tag could not make them soluble. These proteins were always expressed as inclusion bodies and had no functions. LMO2 was a small molecular weight and insoluble protein, in this study, GST system and MBP system were used to express GST-LMO2 and MBP-LMO2 fusion proteins, respectively. We found that GST-LMO2 fusion protein was expressed as inclusion bodies whereas MBP-LMO2 fusion protein was expressed in soluble form. Moreover, the production rate of MBP-LMO2 was also much higher than GST-LMO2. Then MBP-LMO2 fusion proteins and renatured GST-LMO2 fusion proteins were used as bait in pulldown assay to study the interaction between LMO2 and endogenous GATA1 in K562 cells. Western blot analyses showed that both of these proteins could bind to endogenous GATA1 in K562 cells, but recovered GATA1 protein by MBP-LMO2 fusion protein was much more than GST-LMO2 fusion protein. These results suggest that using of MBP system is a helpful attempt in the case of studying small molecular weight, strong hydrophobic proteins.


Subject(s)
Carrier Proteins/chemistry , DNA-Binding Proteins/chemistry , GATA1 Transcription Factor/chemistry , Glutathione Transferase/chemistry , Metalloproteins/chemistry , Recombinant Fusion Proteins/metabolism , Adaptor Proteins, Signal Transducing , Chemical Precipitation , Genetic Vectors , Humans , K562 Cells , LIM Domain Proteins , Maltose-Binding Proteins , Protein Binding , Protein Interaction Domains and Motifs , Protein Renaturation , Proto-Oncogene Proteins/chemistry , Recombinant Fusion Proteins/genetics
6.
J Cell Biochem ; 103(5): 1584-97, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-17879955

ABSTRACT

Breast carcinoma is one of the most common malignant tumors and has become a more common cancer in women. BMP6 was abnormally expressed in breast cancer specimens and cell lines. However, the contribution of BMP6 in promoting breast cancer progression remains unknown. The purpose of our study was to establish whether expression of BMP6 in breast cancer cells affect their proliferation or apoptosis and the mechanism. We found that BMP6 inhibited proliferation of MDA-MB-231 cells and blocked cell cycle at G(0)/G(1) stage. BMP6 also inhibited serum deprivation induced apoptosis in MDA-MB-231 cells. At the 4 days of serum starvation, BMP6 reduced the percentage of caspase-3 positive cells from 49% to 21%, BMP6 also reduced sub-G(1) peak induced by serum starvation. In contrast, BMP6 significantly enhanced survivin expression both at mRNA and protein levels. Dominant negative-survivin and Antisense-survivin impaired BMP6 induced antiapoptotic effect. BMP6 enhanced survivin expression at the transcription level in a Smad-dependent manner. BMP6 also played its antiapoptotic effect through activation p38 MAPK signal pathway, independent of smad/survivin pathway. These results suggested that BMP6 induced cell cycle arrest in estrogen-insensitive breast cancer cells. BMP6 inhibits stress-induced apoptosis via both Smad and p38 signal pathways.


Subject(s)
Apoptosis , Bone Morphogenetic Proteins/metabolism , Breast Neoplasms/metabolism , MAP Kinase Signaling System , Smad Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Bone Morphogenetic Protein 6 , Bone Morphogenetic Proteins/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Female , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins , MAP Kinase Signaling System/drug effects , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , RNA, Messenger/biosynthesis , Resting Phase, Cell Cycle/drug effects , Survivin , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...