Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Breast Cancer Res Treat ; 143(1): 69-80, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24292869

RESUMO

Aromatase inhibitors (AIs) exert antiproliferative effects by reducing local estrogen production from androgens in postmenopausal women with hormone-responsive breast cancer. Previous reports have shown that androgen metabolites generated by the aromatase-independent enzymes, 5α-androstane-3ß, 17ß-diol (3ß-diol), androst-5-ene-3ß, and 17ß-diol (A-diol), also activate estrogen receptor (ER) α. Estradiol (E2) can also reportedly be generated from estrone sulfate (E1S) pooled in the plasma. Estrogenic steroid-producing aromatase-independent pathways have thus been proposed as a mechanism of AI resistance. However, it is unclear whether these pathways are functional in clinical breast cancer. To investigate this issue, we assessed the transcriptional activities of ER in 45 ER-positive human breast cancers using the adenovirus estrogen-response element-green fluorescent protein assay and mRNA expression levels of the ER target gene, progesterone receptor, as indicators of ex vivo and in vivo ER activity, respectively. We also determined mRNA expression levels of 5α-reductase type 1 (SRD5A1) and 3ß-hydroxysteroid dehydrogenase type 1 (3ß-HSD type 1; HSD3B1), which produce 3ß-diol from androgens, and of steroid sulfatase (STS) and 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD type 1; HSD17B1), which produce E2 or A-diol from E1S or dehydroepiandrosterone sulfate. SRD5A1 and HSD3B1 expression levels were positively correlated with ex vivo and in vivo ER activities. STS and HSD17B1 expression levels were positively correlated with in vivo ER activity alone. Elevated expression levels of these steroid-metabolizing enzymes in association with high in vivo ER activity were particularly notable in postmenopausal patients. Analysis of the expression levels of steroid-metabolizing enzymes revealed positive correlations between SRD5A1 and HSD3B1, and STS and HSD17B1. These findings suggest that the SRD5A1-HSD3B1 as well as the STS-HSD17B pathways, could contributes to ER activation, especially postmenopause. These pathways might function as an alternative estrogenic steroid-producing, aromatase-independent pathways.


Assuntos
Aromatase/metabolismo , Neoplasias da Mama/metabolismo , Redes e Vias Metabólicas , Esteroides/metabolismo , Adulto , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Inibidores da Aromatase/farmacologia , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Estradiol/sangue , Estradiol/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Modelos Biológicos , Gradação de Tumores , Estadiamento de Neoplasias , Pós-Menopausa , Receptores de Estrogênio/metabolismo , Fatores de Risco , Transdução de Sinais
2.
Breast Cancer Res Treat ; 139(3): 731-40, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23780684

RESUMO

Aromatase inhibitors (AIs) have been reported to exert their antiproliferative effects in postmenopausal women with hormone receptor-positive breast cancer not only by reducing estrogen production but also by unmasking the inhibitory effects of androgens such as testosterone (TS) and dihydrotestosterone (DHT). However, the role of androgens in AI-resistance mechanisms is not sufficiently understood. 5α-Androstane-3ß,17ß-diol (3ß-diol) generated from DHT by 3ß-hydroxysteroid dehydrogenase type 1 (HSD3B1) shows androgenic and substantial estrogenic activities, representing a potential mechanism of AI resistance. Estrogen response element (ERE)-green fluorescent protein (GFP)-transfected MCF-7 breast cancer cells (E10 cells) were cultured for 3 months under steroid-depleted, TS-supplemented conditions. Among the surviving cells, two stable variants showing androgen metabolite-dependent ER activity were selected by monitoring GFP expression. We investigated the process of adaptation to androgen-abundant conditions and the role of androgens in AI-resistance mechanisms in these variant cell lines. The variant cell lines showed increased growth and induction of estrogen-responsive genes rather than androgen-responsive genes after stimulation with androgens or 3ß-diol. Further analysis suggested that increased expression of HSD3B1 and reduced expression of androgen receptor (AR) promoted adaptation to androgen-abundant conditions, as indicated by the increased conversion of DHT into 3ß-diol by HSD3B1 and AR signal reduction. Furthermore, in parental E10 cells, ectopic expression of HSD3B1 or inhibition of AR resulted in adaptation to androgen-abundant conditions. Coculture with stromal cells to mimic local estrogen production from androgens reduced cell sensitivity to AIs compared with parental E10 cells. These results suggest that increased expression of HSD3B1 and reduced expression of AR might reduce the sensitivity to AIs as demonstrated by enhanced androgen metabolite-induced ER activation and growth mechanisms. Androgen metabolite-dependent growth of breast cancer cells may therefore play a role in AI-resistance.


Assuntos
Androgênios/metabolismo , Inibidores da Aromatase/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos , Receptores de Estrogênio/metabolismo , Androstano-3,17-diol/metabolismo , Androstano-3,17-diol/farmacologia , Neoplasias da Mama/metabolismo , Di-Hidrotestosterona/metabolismo , Di-Hidrotestosterona/farmacologia , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Letrozol , Células MCF-7/efeitos dos fármacos , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Nitrilas/farmacologia , Progesterona Redutase/genética , Progesterona Redutase/metabolismo , Receptores Androgênicos/metabolismo , Elementos de Resposta/genética , Transdução de Sinais , Esteroide Isomerases/genética , Esteroide Isomerases/metabolismo , Testosterona/metabolismo , Testosterona/farmacologia , Triazóis/farmacologia
3.
Anticancer Res ; 42(2): 759-766, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35093874

RESUMO

BACKGROUND/AIM: Carboxyl terminus of Hsc70-interacting protein (CHIP) is a ubiquitin ligase that induces ubiquitination and degradation of its target proteins including oncoproteins. We reported that its down-regulation is associated with tumor progression and metastasis of breast cancer. However, the mechanism through which CHIP gene affects cancer cells is unclear. MATERIALS AND METHODS: We extracted RNA from 45 primary breast cancer samples and compared CHIP mRNA expression profiles, promoter DNA methylation status, and clinicopathological information. RESULTS: CHIP mRNA expression was significantly correlated with the tumor progression status. In several samples, a pinpoint CpG methylation in the CHIP gene promoter region was significantly correlated with CHIP mRNA expression. When this specific CpG was methylated in estrogen receptor (ER)-positive cases, a significant difference in 5-year recurrence was not found compared with ER-negative cases. CONCLUSION: CpG methylation contributes to the long-term prognosis of ER-positive breast cancer.


Assuntos
Neoplasias da Mama/genética , Ubiquitina-Proteína Ligases/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ilhas de CpG , Metilação de DNA , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Prognóstico , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Receptores de Estrogênio/metabolismo , Recidiva , Células Tumorais Cultivadas
4.
J Steroid Biochem Mol Biol ; 165(Pt B): 448-457, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27632897

RESUMO

The relationship between tobacco smoke and breast cancer incidence has been studied for many years, but the effect of smoking on hormonal therapy has not been previously reported. We investigated the effect of smoking on hormonal therapy by performing in vitro experiments. We first prepared tobacco smoke condensate (TSC) and examined its effect on estrogen receptor (ER) activity. The ER activity was analyzed using MCF-7-E10 cells into which the estrogen-responsive element (ERE)-green fluorescent protein (GFP) reporter gene had been stably introduced (GFP assay) and performing an ERE-luciferase assay. TSC significantly activated ERs, and upregulated its endogenous target genes. This activation was inhibited by fulvestrant but more weakly by tamoxifen. These results suggest that the activation mechanism may be different from that for estrogen. Furthermore, using E10 estrogen depletion-resistant cells (EDR cells) established as a hormonal therapy-resistant model showing estrogen-independent ER activity, ER activation and induction of ER target genes were significantly higher following TSC treatment than by estradiol (E2). These responses were much higher than those of the parental E10 cells. In addition, the phosphorylation status of signaling factors (ERK1/2, Akt) and ER in the E10-EDR cells treated with TSC increased. The gene expression profile induced by estrogenic effects of TSC was characterized by microarray analysis. The findings suggested that TSC activates ER by both ligand-dependent and -independent mechanisms. Although TSC constituents will be metabolized in vivo, breast cancer tissues might be exposed for a long period along with hormonal therapy. Tobacco smoke may have a possibility to interfere with hormonal therapy for breast cancer, which may have important implications for the management of therapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/metabolismo , Fumaça , Antineoplásicos Hormonais/farmacologia , Proliferação de Células , Estradiol/análogos & derivados , Estradiol/farmacologia , Estrogênios/farmacologia , Fulvestranto , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Hormônios/farmacologia , Humanos , Ligantes , Células MCF-7 , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia , Nicotiana
5.
J Steroid Biochem Mol Biol ; 167: 115-125, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27888136

RESUMO

Approximately 70% of breast cancers express estrogen receptor α (ERα), which plays critical roles in breast cancer development. Fulvestrant has been effectively used to treat ERα-positive breast cancer, although resistance remains a critical problem. To elucidate the mechanism of resistance to fulvestrant, we established fulvestrant-resistant cell-lines named MFR (MCF-7 derived fulvestrant resistance) and TFR (T-47D derived fulvestrant resistance) from the ERα-positive luminal breast cancer cell lines MCF-7 and T-47D, respectively. Both fulvestrant-resistant cell lines lost sensitivity to estrogen and anti-estrogens. We observed diminished ERα expression at both the protein and mRNA levels. To address the mechanism of gene expression regulation, we examined epigenetic alteration, especially the DNA methylation level of ERα gene promoters. MFR cells displayed high methylation levels upstream of the ERα gene, whereas no change in DNA methylation was observed in TFR cells. Hence, we examined the gene expression plasticity of ERα, as there are differences in its reversibility following fulvestrant withdrawal. ERα gene expression was not restored in MFR cells, and alternative intracellular phosphorylation signals were activated. By contrast, TFR cells exhibited plasticity of ERα gene expression and ERα-dependent growth; moreover, these cells were resensitized to estrogen and anti-estrogens. The difference in epigenetic regulation among individual cells might explain the difference in the plasticity of ERα expression. We also identified an MFR cell-activating HER/Src-Akt/MAPK pathway; thus, the specific inhibitors effectively blocked MFR cell growth. This finding implies the presence of multiple fulvestrant resistance mechanisms and suggests that the optimal therapies differ among individual tumors as a result of differing epigenetic mechanisms regulating ERα gene expression.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética/efeitos dos fármacos , Estradiol/análogos & derivados , Receptor alfa de Estrogênio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Fulvestranto , Regulação Neoplásica da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Regiões Promotoras Genéticas , Transdução de Sinais
6.
J Steroid Biochem Mol Biol ; 171: 209-217, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28412323

RESUMO

Hormone therapy is the most effective treatment for patients with estrogen receptor α-positive breast cancers. However, although resistance occurs during treatment in some cases and often reflects changed estrogen receptor α status, the relationship between changes in estrogen receptor α expression and resistance to therapy are poorly understood. In this study, we identified a mechanism for altered estrogen receptor α expression during disease progression and acquired hormone therapy resistance in aromatase inhibitor-resistant breast cancer cell lines. Subsequently, we investigated promoter switching and DNA methylation status of the estrogen receptor α promoter, and found marked changes of methylation at a single CpG site (CpG4) in resistant cells. In addition, luciferase reporter assays showed reduced transcriptional activity from this methylated CpG site. This CpG region was also completely conserved among species, suggesting that it acts as a methylation-sensitive Ets-2 transcription factor binding site, as confirmed using chromatin immunoprecipitation assays. In estrogen receptor α-positive tumors, CpG4 methylation levels were inversely correlated with estrogen receptor α expression status, suggesting that single CpG site plays an important role in the regulation of estrogen receptor α transcription.


Assuntos
Neoplasias da Mama/metabolismo , Metilação de DNA , Fosfatos de Dinucleosídeos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Regiões Promotoras Genéticas , Proteína Proto-Oncogênica c-ets-2/metabolismo , Transcrição Gênica , Antineoplásicos Hormonais/farmacologia , Inibidores da Aromatase/farmacologia , Sequência de Bases , Neoplasias da Mama/tratamento farmacológico , Sequência Conservada , Metilação de DNA/efeitos dos fármacos , DNA Recombinante/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/genética , Feminino , Genes Reporter/efeitos dos fármacos , Humanos , Células MCF-7 , Pessoa de Meia-Idade , Mutação , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteína Proto-Oncogênica c-ets-2/genética , Proteínas Recombinantes/metabolismo , Elementos de Resposta/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
7.
PLoS One ; 11(5): e0155844, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27228187

RESUMO

Aromatase inhibitors (AIs) effectively treat hormone receptor-positive postmenopausal breast cancer, but some patients do not respond to treatment or experience recurrence. Mechanisms of AI resistance include ligand-independent activation of the estrogen receptor (ER) and signaling via other growth factor receptors; however, these do not account for all forms of resistance. Here we present an alternative mechanism of AI resistance. We ectopically expressed aromatase in MCF-7 cells expressing green fluorescent protein as an index of ER activity. Aromatase-overexpressing MCF-7 cells were cultured in estrogen-depleted medium supplemented with testosterone and the AI, letrozole, to establish letrozole-resistant (LR) cell lines. Compared with parental cells, LR cells had higher mRNA levels of steroid sulfatase (STS), which converts estrone sulfate (E1S) to estrone, and the organic anion transporter peptides (OATPs), which mediate the uptake of E1S into cells. LR cells proliferated more in E1S-supplemented medium than did parental cells, and LR proliferation was effectively inhibited by an STS inhibitor in combination with letrozole and by ER-targeting drugs. Analysis of ER-positive primary breast cancer tissues showed a significant correlation between the increases in the mRNA levels of STS and the OATPs in the LR cell lines, which supports the validity of this AI-resistant model. This is the first study to demonstrate the contribution of STS and OATPs in E1S metabolism to the proliferation of AI-resistant breast cancer cells. We suggest that E1S metabolism represents a new target in AI-resistant breast cancer treatment.


Assuntos
Inibidores da Aromatase/farmacologia , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Feminino , Humanos , Células Tumorais Cultivadas
8.
Cancer Med ; 5(8): 1873-82, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27334118

RESUMO

The carboxyl terminus of the Hsc70-interacting protein (CHIP) is considered to induce the ubiquitination and degradation of several oncogenic proteins, and play a role in the inhibition of tumor progression and invasion under experimental conditions. However, the impact of CHIP expression on the prognosis of breast cancer patients has not yet been established. In this study, using an immunohistochemical method, 272 patients with invasive breast cancer were assessed for the expression of CHIP (graded scores 0-3) and the statuses of biomarkers, such as estrogen receptor (ER), progesterone receptor (PgR), and HER2. The relationships between the statuses of CHIP and biomarkers as well as clinical features were also evaluated, and that between the expression of CHIP and patient prognosis was analyzed. We revealed that the strong expression of CHIP correlated with positive ER (P < 0.001), positive PgR (P < 0.001), and negative HER2 (P = 0.02). In postmenopausal patients, relapse-free survival (RFS) was significantly better in the high CHIP group than in the low CHIP group (P = 0.042). In addition, RFS and cancer-specific survival (CSS) were significantly better in patients with ER-positive/CHIP score 3 tumors than in those with ER-negative/CHIP score 0 tumors (RFS: P = 0.038, CSS: P = 0.0098). The methylation status of CHIP gene promoter did not always account for the down-regulation of its expression. In conclusion, the overexpression of CHIP is a potent prognostic factor of a good prognosis in ER-positive breast cancer patients in the postmenopausal phase.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Mastectomia/métodos , Pessoa de Meia-Idade , Gradação de Tumores , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Pós-Menopausa , Prognóstico , Regiões Promotoras Genéticas , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
9.
Clin Breast Cancer ; 14(4): 249-57, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24332559

RESUMO

INTRODUCTION: Estrogen receptor (ER)-α expression offers a critical characterization of breast cancer, but risk of recurrence is difficult to predict using only ERα status. The ERα gene has at least 6 transcription start sites, 6 distinct first exons, and probably 6 promoters. To examine whether these promoters have differential effects in breast cancer, we quantified expression of promoter-specific ERα messenger RNA (mRNA), using real-time polymerase chain reaction (PCR) and statistical assessment. PATIENTS AND METHODS: We examined variations in the use of breast cancer cell lines and 43 ERα positive (ERα(+)) breast cancer tissue samples by quantifying promoter-specific mRNA of ERα with real-time PCR analysis using primers and probes specially designed for this study. Moreover, we correlated the results of quantified the promoter-specific mRNA with mRNA of total ERα and related them to clinicopathological factors statistically. We also examined multiregression analyses for promoter-specific mRNAs of ERα. RESULT: We found the promoters to be used at almost similar ratios among ERα(+) breast cancer cell lines and ERα(+) breast cancer tissues. Clinicopathological variations were associated with identical ERα promoter choices. When we examined the contribution of mRNA from 3 promoters in breast cancer tissues to total ERα using multiple regression analysis, we found that only promoter A showed a significant (P < .05) transcript coefficient. CONCLUSION: Our findings imply that the use of ERα promoters as prognostic biomarkers is unfeasible. However, our results suggest that promoter usage of ERα may contribute to its expression in normal development and differentiation of individual or carcinogenesis of breast cancer.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/genética , Regulação Neoplásica da Expressão Gênica , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Feminino , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
10.
J Steroid Biochem Mol Biol ; 139: 61-72, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24128438

RESUMO

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.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Elementos de Resposta , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Proliferação de Células , Moduladores de Receptor Estrogênico/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/fisiologia , Feminino , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células MCF-7 , Fosforilação , Processamento de Proteína Pós-Traducional , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo
11.
J Steroid Biochem Mol Biol ; 144 Pt B: 513-22, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25178713

RESUMO

Aromatase inhibitors (AI) are commonly used to treat postmenopausal estrogen-receptor (ER)-positive breast carcinoma. However, resistance to AI is sometimes acquired, and the molecular mechanisms underlying such resistance are largely unclear. Recent studies suggest that AI treatment increases androgen activity during estrogen deprivation in breast carcinoma, but the role of the androgen receptor (AR) in breast carcinoma is still a matter of controversy. The purpose of this study is to examine the potential correlation between the AR- and AI-resistant breast carcinoma. To this end, we performed immunohistochemical analysis of 21 pairs of primary breast carcinoma and corresponding AI-resistant recurrent tissue samples and established two stable variant cell lines from ER-positive T-47D breast carcinoma cell line as AI-resistance models and used them in in vitro experiments. Immunohistochemical analysis demonstrated that the expression of prostate-specific antigen (PSA) and Ki-67 were significantly higher and ER and progesterone receptor (PR) were lower in recurrent lesions compared to the corresponding primary lesions. Variant cell lines overexpressed AR and PSA and exhibited neither growth response to estrogen nor expression of ER. Androgen markedly induced the proliferation of these cell lines. In addition, the expression profile of androgen-induced genes was markedly different between variant and parental cell lines as determined by microarray analysis. These results suggest that in some cases of ER-positive breast carcinoma, tumor cells possibly change from ER-dependent to AR-dependent, rendering them resistant to AI. AR inhibitors may thus be effective in a selected group of patients.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptores Androgênicos/metabolismo , Idoso , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Humanos , Calicreínas/metabolismo , Antígeno Ki-67/metabolismo , Pessoa de Meia-Idade , Antígeno Prostático Específico/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo
12.
Cancer Med ; 1(3): 328-37, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23342282

RESUMO

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.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/genética , Receptores de Progesterona/biossíntese , Ativação Transcricional , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/biossíntese , Estrogênios/genética , Estrogênios/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Fluorescência Verde , Humanos , Antígeno Ki-67/genética , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Receptores de Progesterona/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA