Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Cancer Microenviron ; 7(1-2): 23-31, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24203105

ABSTRACT

Estrogen and various growth factors affecting tumor behavior are present in the breast cancer microenvironment, but their comprehensive effects and signal crosstalks are different in each case. However, there is no system to evaluate the factors, detected in individual breast cancer cases, that regulate ER activity and tumor progression. In this study, we analyzed the effects of individual breast cancer extracts by our original system using an estrogen-signal reporter cell line, MCF-7-E10, which we previously established. MCF-7-E10 cell line is stably transfected by an estrogen response element (ERE)-green fluorescent protein (GFP) gene; it expresses GFP when estrogen receptors (ERs) are activated by estrogen or growth factor signal-mediated ER phosphorylation. Using this cell line, we analyzed the comprehensive effects of factors derived from breast cancer tissues on ER activity and growth of MCF-7-E10 cells for each case. We also analyzed relationships between these activities and clinicopathologic characteristics of patients who provided cancer specimens. The breast cancer extracts, which reflect the combined activities of growth factors present in individual cases, stimulated MCF-7-E10 cell growth in an estrogen-independent manner, and specifically stimulated growth of other breast cancer cell lines, regardless of ER expression. High growth-promoting activities were seen in tumor regions of specimens with tumors > 10 mm in size, HER2 intrinsic subtype, and scirrhous and solid-tubular carcinoma histological subtypes. Anti-human hepatocyte growth factor (HGF) antibody and an inhibitor for insulin-like growth factor-1 (IGF-1) receptor inhibited MCF-7-E10 cell growth by the breast cancer extracts, indicating that signal pathways via HGF or IGF-1 receptor significantly affect breast cancer. These data suggest that growth factors other than estrogen in the tumor extract significantly affect breast cancer aggressiveness in an estrogen-independent manner, and could be useful therapeutic targets.

2.
J Steroid Biochem Mol Biol ; 139: 61-72, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24128438

ABSTRACT

The acquisition of estrogen-deprivation resistance and estrogen receptor (ER) signal-independence in ER-positive breast cancer is one of the crucial steps in advancing the aggressiveness of breast cancer; however, this has not yet been elucidated in detail. To address this issue, we established several estrogen-deprivation-resistant (EDR) breast cancer cell lines from our unique MCF-7 cells, which had been stably transfected with an ERE-GFP reporter plasmid. Three cell lines with high ER activity and another 3 cell lines with no ER activity were established from cell cloning by monitoring GFP expression in living cells. The former three ERE-GFP-positive EDR cell lines showed the overexpression of ER and high expression of several ER-target genes. Further analysis of intracellular signaling factors revealed a marked change in the phosphorylation status of ERα on Ser167 and Akt on Thr308 by similar mechanisms reported previously; however, we could not find any changes in MAP-kinase factors. Comprehensive phospho-proteomic analysis also indicated the possible contribution of the Akt pathway to the phosphorylation of ERα. On the other hand, constitutive activation of c-Jun N-terminal kinase (JNK) was observed in ERE-GFP-negative EDR cells, and the growth of these cells was inhibited by a JNK inhibitor. An IGF1R-specific inhibitor diminished the phosphorylation of JNK, which suggested that a novel signaling pathway, IGF1R-JNK, may be important for the proliferation of ER-independent MCF-7 cells. These results indicate that ER-positive breast cancer cells can acquire resistance by more than two mechanisms at a time, which suggests that multiple mechanisms may occur simultaneously. This finding also implies that breast cancers with different resistance mechanisms can concomitantly occur and mingle in an individual patient, and may be a cause of the recurrence of cancer.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Response Elements , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/physiology , Female , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MCF-7 Cells , Phosphorylation , Protein Processing, Post-Translational , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism
3.
Breast Cancer Res Treat ; 133(2): 427-36, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21909982

ABSTRACT

Neoadjuvant chemotherapy (NAC) has become the standard treatment for advanced breast cancer. Several prognostic markers, including estrogen receptor-α (ERα), are used to predict the response to NAC. However, the molecular significance of ERα expression in the efficacy of chemotherapy is not yet fully understood. To examine this issue, we first evaluated ERα transcriptional activity in breast cancer cells derived from pre-NAC specimens using estrogen response element-green fluorescent protein (ERE-GFP) as a reporter gene, and found that, in the cases for which ERα activities determined by GFP expression were not detected or low, pCR (pathological complete response) could be achieved even though ERα protein was expressed. Next, we examined the effects of alterations in ERα expression levels on sensitivity to paclitaxel, a key drug in NAC, by stable expression of ERα in ER-negative SKBR3 cells and by siRNA-mediated down-regulation of ERα in ER-positive MCF-7 cells, and showed that ERα expression and sensitivity to paclitaxel showed an inverse correlation. We also established paclitaxel-resistant MCF-7 cell clones and found that they have higher estrogen-induced ER activity than parent cells. Paclitaxel is a microtubule-stabilizing agent, while HDAC6 (histone deacetylase 6), which we previously identified as an estrogen-regulated gene, enhances cell motility by destabilizing microtubules via deacetylation of α-tubulin. Finally, we demonstrate herein that ERα knockdown in MCF-7 cells prevents deacetylation of α-tubulin, thereby increasing sensitivity to paclitaxel. Taken together, these results suggest that ERα expression directly regulates sensitivity to paclitaxel in NAC for breast cancer via the effect on microtubule stability.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Paclitaxel/therapeutic use , Acetylation , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Humans , Neoadjuvant Therapy , Transcription, Genetic , Treatment Outcome , Tubulin/metabolism
4.
Cancer Med ; 1(3): 328-37, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23342282

ABSTRACT

To predict the efficacy of hormonal therapy at the individual-level, immunohistochemical methods are used to analyze expression of classical molecular biomarkers such as estrogen receptor (ER), progesterone receptor (PgR), and HER2. However, the current diagnostic standard is not perfect for the individualization of diverse cases. Therefore, establishment of more accurate diagnostics is required. Previously, we established a novel method that enables analysis of ER transcriptional activation potential in clinical specimens using an adenovirus estrogen response element-green fluorescence protein (ERE-GFP) assay system. Using this assay, we assessed the ERE transcriptional activity of 62 primary breast cancer samples. In 40% of samples, we observed that ER protein expression was not consistent with ERE activity. Comparison of ERE activity with clinicopathological information revealed that ERE activity was significantly correlated with the ER target gene, PgR, rather than ER in terms of both protein and mRNA expression. Moreover, subgrouping of Luminal A-type breast cancer samples according to ERE activity revealed that ERα mRNA expression correlated with ER target gene mRNA expression in the high-, but not the low-, ERE-activity group. On the other hand, the low-ERE-activity group showed significantly higher mRNA expression of the malignancy biomarker Ki67 in association with disease recurrence in 5% of patients. Thus, these data suggest that ER expression does not always correlate with ER transcriptional activity. Therefore, in addition to ER protein expression, determination of ERE activity as an ER functional marker will be helpful for analysis of a variety of diverse breast cancer cases and the subsequent course of treatment.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Receptors, Progesterone/biosynthesis , Transcriptional Activation , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Estrogen Receptor alpha/biosynthesis , Estrogens/genetics , Estrogens/metabolism , Female , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins , Humans , Ki-67 Antigen/genetics , Middle Aged , Neoplasm Staging , Receptors, Progesterone/genetics
5.
Anticancer Res ; 29(10): 3971-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19846938

ABSTRACT

BACKGROUND: Expression analysis of estrogen response genes (ERGs) may help to predict the effectiveness of endocrine therapies in breast cancer patients. We produced a custom-made, 3-dimensional microarray system (3DMS), using previously identified ERGs, and analyzed expression of ERGs on breast cancer tissues. MATERIALS AND METHODS: aRNA was synthesized from 27 breast cancer tumors. The aRNAs were applied to the infrastructure of a 3DMS with spotted cDNA probes to 36 ERGs. Data were analyzed by cluster analysis. RESULTS: All 27 specimens were classified clearly into one of two clusters based on whether the ERGs were up or down regulated. These groups correlated with high expression of ER (P<0.05) and Her2 (P<0.05). In some cases, ERG expressions were low even though the tumor was ER positive. CONCLUSION: 3DMS may be useful for accurately predicting prognosis and whether endocrine therapies targeting the ER would be effective in an individual breast cancer patient.


Subject(s)
Breast Neoplasms/genetics , Estrogens/genetics , Gene Expression Regulation, Neoplastic , Oligonucleotide Array Sequence Analysis/methods , Breast Neoplasms/pathology , Female , Humans , Neoplasm Staging , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/genetics , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Receptors, Progesterone/biosynthesis
6.
Nat Cell Biol ; 11(3): 312-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19198599

ABSTRACT

CHIP is a U-box-type ubiquitin ligase that induces ubiquitylation and degradation of its substrates, which include several oncogenic proteins. The relationship between CHIP and tumour progression, however, has not been elucidated. Here, we show that CHIP suppresses tumour progression in human breast cancer by inhibiting oncogenic pathways. CHIP levels were negatively correlated with the malignancy of human breast tumour tissues. In a nude mouse xenograft model, tumour growth and metastasis were significantly inhibited by CHIP expression. In contrast, knockdown of CHIP (shCHIP) in breast cancer cells resulted in rapid tumour growth and metastastic phenotypes in mice. In cell-based experiments, anchorage-independent growth and invasiveness of shCHIP cells was significantly elevated due to increased expression of Bcl2, Akt1, Smad and Twist. Proteomic analysis identified the transcriptional co-activator SRC-3 (refs 13, 14, 15, 16, 17, 18, 19) as a direct target for ubiquitylation and degradation by CHIP. Knocking down SRC-3 in shCHIP cells reduced the expression of Smad and Twist, and suppressed tumour metastasis in vivo. Conversely, SRC-3 co-expression prevented CHIP-induced suppression of metastasis formation. These observations demonstrate that CHIP inhibits anchorage-independent cell growth and metastatic potential by degrading oncogenic proteins including SRC-3.


Subject(s)
Breast Neoplasms/enzymology , Ubiquitin-Protein Ligases/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Transformation, Neoplastic , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases/metabolism , Humans , Lung Neoplasms/secondary , Mice , Nuclear Receptor Coactivator 3 , Protein Processing, Post-Translational , Trans-Activators/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
7.
Endocr Relat Cancer ; 15(2): 451-63, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18508998

ABSTRACT

The estrogen pathway plays an important role in the etiology of human endometrial carcinoma (EC). We examined whether estrogen biosynthesis in the tumor microenvironment promotes endometrial cancer. To examine the contribution of stromal cells to estrogen signaling in EC, we used reporter cells stably transfected with the estrogen response element (ERE) fused to the destabilized green fluorescent protein (GFP) gene. In this system, the endometrial cancer stromal cells from several patients activated the ERE of cancer cells to a variable extent. The GFP expression level increased when testosterone, a substrate for aromatase, was added. The effect was variably inhibited by aromatase inhibitors (AIs), although the response to AIs varied among patients. These results suggest that GFP expression is driven by estrogen synthesized by aromatase in the endometrial cancer stromal cells. In a second experiment, we constructed an adenovirus reporter vector containing the same construct as the reporter cells described above, and visualized endogenous ERE activity in primary culture cancer cells from 15 EC specimens. The GFP expression levels varied among the cases, and in most primary tissues, ERE activities were strongly inhibited by a pure anti-estrogen, fulvestrant. Interestingly, a minority of primary tissues in endometrial cancer showed ERE activity independent of the estrogen-ER pathway. These results suggest that AI may have some therapeutic value in EC; however, the hormonal microenvironment must be assessed prior to initiating therapy.


Subject(s)
Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Estrogens/metabolism , Signal Transduction/physiology , Stromal Cells/metabolism , 17-Hydroxysteroid Dehydrogenases/metabolism , Adenoviridae/genetics , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , Breast Neoplasms , Cell Communication/physiology , Cell Line, Tumor , Estrogens/biosynthesis , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , Middle Aged , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Stromal Cells/cytology , Testosterone/pharmacology
8.
Toxicol In Vitro ; 21(4): 741-52, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17369015

ABSTRACT

It is important to know the difference as well as the similarity in estrogen responsiveness among cell lines for understanding the effects of estrogenic chemicals. Here, using 120 estrogen responsive genes, we examined comparative expression profiles between the profile in breast cancer MCF-7 cells treated with 17beta-estradiol and the profiles in other cell lines derived from breast (T-47D and HBC-4 cells), endometrium (Ishikawa cells) and kidney (RXF-631L cells) treated with estrogenic chemicals. First, comparative profiling between MCF-7 and T-47D cells showed similar (correlation coefficient or R value=0.49-0.87) profiles for all chemicals examined: 17beta-estradiol, estrone, estriol, diethylstilbestrol, bisphenol A, nonylphenol and genistein. The analysis using other cell lines indicated that significant correlations to the profile in MCF-7 cells treated with 17beta-estradiol were observed for the profiles in Ishikawa cells treated with 17beta-estradiol, diethylstilbestrol and bisphenol A, and HBC-4 cells treated with 17beta-estradiol. The profiles for diethylstilbestrol and bisphenol A in HBC-4 cells and all three chemicals in RXF-631L cells did not show significant correlation with those in MCF-7 cells. Hierarchical cluster analysis revealed that there are cell-specific responses to estrogenic chemicals (T-47D and HBC-4 cells for example). Correlation analysis using six (proliferation, transcription, transport, enzymes, signaling and others) functionally-categorized gene groups indicated that the genes related to enzymes showed greater correlations for all chemicals tested in T-47D cells and some chemicals in Ishikawa and HBC-4 cells while those related to transcription contributed to variations.


Subject(s)
Estrogens, Non-Steroidal/pharmacology , Gene Expression Profiling , Biological Transport, Active/drug effects , Biological Transport, Active/genetics , Cell Line , Cell Proliferation/drug effects , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Data Interpretation, Statistical , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Oligonucleotide Array Sequence Analysis , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...