Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 5: 7641, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25560734

ABSTRACT

Most primary breast cancers express estrogen receptor α and can be treated via endocrine therapy using anti-estrogens such as tamoxifen; however, acquired endocrine resistance is a critical issue. To identify tamoxifen response-related microRNAs (miRNAs) in breast cancer, MCF-7 cells infected with a lentiviral miRNA library were treated with 4-hydroxytamoxifen (OHT) or vehicle for 4 weeks, and the amounts of individual miRNA precursors that had integrated into the genome were evaluated by microarray. Compared to the vehicle-treated cells, 5 'dropout' miRNAs, which were downregulated in OHT-treated cells, and 6 'retained' miRNAs, which were upregulated in OHT-treated cells, were identified. Of the dropout miRNAs, we found that miR-574-3p expression was downregulated in clinical breast cancer tissues as compared with their paired adjacent tissues. In addition, anti-miR-574-3p reversed tamoxifen-mediated suppression of MCF-7 cell growth. Clathrin heavy chain (CLTC) was identified as a miR-574-3p target gene by in silico algorithms and luciferase reporter assay using the 3' untranslated region of CLTC mRNA. Interestingly, loss and gain of miR-574-3p function in MCF-7 cells causes CLTC to be upregulated and downregulated, respectively. These results suggest that functional screening mediated by miRNA libraries can provide new insights into the genes essential for tamoxifen response in breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , Down-Regulation/drug effects , MicroRNAs/metabolism , Tamoxifen/analogs & derivatives , Up-Regulation/drug effects , 3' Untranslated Regions , Algorithms , Base Sequence , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Clathrin/antagonists & inhibitors , Clathrin/genetics , Clathrin/metabolism , Female , Gene Library , Humans , MCF-7 Cells , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotides, Antisense/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sequence Alignment , Tamoxifen/toxicity
2.
Oncogenesis ; 3: e126, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25365482

ABSTRACT

Estrogen receptor-binding fragment-associated antigen 9 (EBAG9) is a primary estrogen-responsive gene that we previously identified in MCF-7 breast cancer cells using the CpG genomic binding-site cloning technique. The expression of EBAG9 protein is often upregulated in malignant tumors, suggesting that this protein is involved in cancer pathophysiology. In the present study, we investigated the role of EBAG9 in host defense against implanted tumors in Ebag9-knockout (Ebag9KO) mice. MB-49 mouse bladder cancer cells were subcutaneously implanted into Ebag9KO and control mice. We found that tumor formation and metastasis to the lung by MB-49 cells were substantially reduced in Ebag9KO mice compared with control mice. The infiltration of CD8(+), CD3(+) and CD4(+) T cells into the generated tumors was enhanced in Ebag9KO mice compared with controls. Notably, CD8(+) T cells isolated from tumors in Ebag9KO mice exhibited substantial upregulation of immunity- and chemoattraction-related genes, including interleukin-10 receptor, interferon gamma, granzyme A, granzyme B and chemokine (C-X-C motif) receptor 3 compared with CD8(+) T cells from tumors in control mice. The CD8(+) T cells isolated from tumors in Ebag9KO mice also exhibited enhanced degranulation and increased cytolytic activity. Furthermore, the adoptive transfer of CD8(+) T cells isolated from tumors in Ebag9KO host could repress tumor growth by MB-49 cells implanted in wild-type host. These results suggest that EBAG9 modulates tumor growth and metastasis by negatively regulating the adaptive immune response in host defense. EBAG9 could be a potential target for tumor immunotherapy.

3.
Oncogene ; 30(5): 619-30, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-20890304

ABSTRACT

The androgen receptor (AR) is a critical transcriptional factor that contributes to the development and the progression of prostate cancer (PCa) by regulating the transcription of various target genes. Genome-wide screening of androgen target genes provides useful information to understand a global view of AR-mediated gene network in PCa. In this study, we performed 5'-cap analysis of gene expression (CAGE) to determine androgen-regulated transcription start sites (TSSs) and chromatin immunoprecipitation (ChIP) on array (ChIP-chip) analysis to identify AR binding sites (ARBSs) and histone H3 acetylated (AcH3) sites in the human genome. CAGE determined 13 110 distinct, androgen-regulated TSSs (P<0.01), and ChIP-chip analysis identified 2872 androgen-dependent ARBSs (P<1e-5) and 25 945 AcH3 sites (P<1e-4). Both androgen-regulated coding genes and noncoding RNAs, including microRNAs (miRNAs) were determined as androgen target genes. Besides prototypic androgen-regulated TSSs in annotated gene promoter regions, there are many androgen-dependent TSSs that are widely distributed throughout the genome, including those in antisense (AS) direction of RefSeq genes. Several pairs of sense/antisense promoters were newly identified within single RefSeq gene regions. The integration of CAGE and ChIP-chip analyses successfully identified a cluster of androgen-inducible miRNAs, as exemplified by the miR-125b-2 cluster on chromosome 21. Notably, the number of androgen-upregulated genes was larger in LNCaP cells treated with R1881 for 24 h than for 6 h, and the percentage of androgen-upregulated genes accompanied with adjacent ARBSs was also much higher in cells treated with R1881 for 24 h than 6 h. On the basis of the Oncomine database, the majority of androgen-upregulated genes containing adjacent ARBSs and CAGE tag clusters in our study were previously confirmed as androgen target genes in PCa. The integrated high-throughput genome analyses of CAGE and ChIP-chip provide useful information for elucidating the AR-mediated transcriptional network that contributes to the development and progression of PCa.


Subject(s)
Chromatin Immunoprecipitation/methods , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Receptors, Androgen/genetics , Acetylation , Androgens/pharmacology , Binding Sites/genetics , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human/genetics , Genomics/methods , Histones/metabolism , Humans , Male , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction/genetics , Transcription Initiation Site
4.
Prostate Cancer Prostatic Dis ; 13(4): 356-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20820187

ABSTRACT

Recent advances in cancer biology reveal that microRNAs (miRNAs) are involved in the regulation of cancer-related genes, or they function as tumor suppressors or oncogenes. In prostate cancer, evidence has accumulated for the contribution of the androgen-dependent gene network to tumor growth, although the precise functions of miRNAs in prostate cancer remain to be investigated. Here, we identified androgen-responsive miRNAs by the short RNA sequencing analysis in LNCaP prostate cancer cells. Among 10 miRNAs with known sequences, we have determined that miR-148a reduces the expression of cullin-associated and neddylation-dissociated 1 (CAND1), a negative regulator of SKP1-Cullin1-F-box (SCF) ubiquitin ligases, by binding to the 3'-untranslated region of CAND1 mRNA. CAND1 knockdown by small interfering RNA promoted the proliferation of LNCaP cells. Our study indicates the potential contribution of miR-148a to the growth of human prostate cancer.


Subject(s)
Adenocarcinoma/genetics , Cell Proliferation , MicroRNAs/genetics , MicroRNAs/physiology , Prostatic Neoplasms/genetics , Transcription Factors/genetics , 3' Untranslated Regions , Adenocarcinoma/pathology , Androgens/pharmacology , Binding Sites/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Male , MicroRNAs/metabolism , Prostatic Neoplasms/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transfection
5.
Cancer Gene Ther ; 17(9): 624-32, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20467453

ABSTRACT

The estrogen-responsive gene Efp promotes the growth of breast cancer cells by stimulating the degradation of a negative cell-cycle regulator, 14-3-3sigma, and is hence considered a suitable molecular target for breast cancer therapy. The use of small interfering RNA (siRNA) and its derivatives to silence cancer-related genes is being investigated with the aim of identifying clinical applications for these molecules. Recently, it has been shown that DNA-modified siRNA (chimeric siRNA) has good potential in clinical applications, because it induces fewer off-target effects or immune responses in mammalian cells. In the present study, we identified the most specific and effective siRNA (siEfp-1) for silencing Efp expression in MCF-7 breast cancer cells. For this purpose, we used an algorithm that primarily eliminates off-target effects. siEfp-1 considerably suppressed the in vitro proliferation and cell-cycle progression of MCF-7 cells, as well as the in vivo growth of MCF-7 tumors, in athymic mice. DNA-modified siEfp-1 (chimeric siEfp) significantly inhibited the expression of Efp, proliferation of cultured cells and the in vivo growth of MCF-7-derived tumors in athymic mice. In addition, the silencing of Efp expression by siEfp-1 and chimeric siEfp increased the expression of the 14-3-3sigma protein. These results suggest that siEfp-1 and chimeric siEfp could be useful in breast cancer therapy. Chimeric siEfp, in particular, has a high specificity and induces few side effects and is therefore expected to be used as a novel nucleic acid-based therapeutic agent.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , DNA/chemistry , RNA, Small Interfering/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Cell Cycle , Cell Line, Tumor , DNA/administration & dosage , DNA/genetics , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
6.
J Mol Endocrinol ; 39(4): 239-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17909264

ABSTRACT

Vitamin K is known as a critical nutrient required for bone homeostasis and blood coagulation, and it is clinically used as a therapeutic agent for osteoporosis in Japan. Besides its enzymatic action as a cofactor of vitamin K-dependent gamma-glutamyl carboxylase (GGCX), we have previously shown that vitamin K(2) is a transcriptional regulator of bone marker genes and extracellular matrix-related genes, by activating the steroid and xenobiotic receptor (SXR). To explore a novel action of vitamin K in osteoblastic cells, we identified genes up-regulated by a vitamin K(2) isoform menaquinone-4 (MK-4) using oligonucleotide microarray analysis. Among these up-regulated genes by MK-4, growth differentiation factor 15 (GDF15) and stanniocalcin 2 (STC2) were identified as novel MK-4 target genes independent of GGCX and SXR pathways in human and mouse osteoblastic cells. The induction of GDF15 and STC2 is likely specific to MK-4, as it was not exerted by another vitamin K(2) isoform MK-7, vitamin K(1), or the MK-4 side chain structure geranylgeraniol. Investigation of the involved signaling pathways revealed that MK-4 enhanced the phosphorylation of protein kinase A (PKA), and the MK-4-dependent induction of both GDF15 and STC2 genes was reduced by the treatment with a PKA inhibitor H89 or siRNA against PKA. These results suggest that vitamin K(2) modulates its target gene expression in osteoblastic cells through the PKA-dependent mechanism, which may be distinct from the previously known vitamin K signaling pathways.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Vitamin K 2/pharmacology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Carbon-Carbon Ligases/genetics , Carbon-Carbon Ligases/metabolism , Cells, Cultured , Gene Expression Profiling , Glycoproteins/genetics , Glycoproteins/metabolism , Growth Differentiation Factor 15 , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Pregnane X Receptor , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Vitamin K 2/analogs & derivatives
7.
Oncogene ; 26(30): 4453-63, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17297473

ABSTRACT

The androgen receptor (AR) plays a key role as a transcriptional factor in prostate development and carcinogenesis. Identification of androgen-regulated genes is essential to elucidate the AR pathophysiology in prostate cancer. Here, we identified androgen target genes that are directly regulated by AR in LNCaP cells, by combining chromatin immunoprecipitation (ChIP) with tiling microarrays (ChIP-chip). ChIP-enriched or control DNAs from the cells treated with R1881 were hybridized with the ENCODE array, in which a set of regions representing approximately 1% of the whole genome. We chose 10 bona fide AR-binding sites (ARBSs) (P<1e-5) and validated their significant AR recruitment ligand dependently. Eight upregulated genes by R1881 were identified in the vicinity of the ARBSs. Among the upregulated genes, we focused on UGT1A and CDH2 as AR target genes, because the ARBSs close to these genes (in UGT1A distal promoter and CDH2 intron 1) were most significantly associated with acetylated histone H3/H4, RNA polymerase II and p160 family co-activators. Luciferase reporter constructs including those two ARBSs exhibited ligand-dependent transcriptional regulator/enhancer activities. The present study would be powerful to extend our knowledge of the diversity of androgen genetic network and steroid action in prostate cancer cells.


Subject(s)
Androgens/pharmacology , Chromatin Immunoprecipitation/methods , Oligonucleotide Array Sequence Analysis/methods , Prostatic Neoplasms/genetics , Response Elements , Acetylation , Antigens, CD/genetics , Binding Sites , Cadherins/genetics , Cell Line, Tumor , Glucuronosyltransferase/genetics , Histones/metabolism , Humans , Male , RNA Polymerase II/metabolism , Receptors, Androgen/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...