Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Horm Cancer ; 9(4): 229-239, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29546532

RESUMO

Obesity is associated with increased risk of breast cancer in postmenopausal but not in premenopausal women. Many factors may be responsible for this difference. The aim of this study was to determine the mechanisms by which the genes related to the AMPK pathway, inflammation, and estrogen actions are affected by adiposity in breast tissue with the objective of identifying differences that may explain the different breast cancer risk in premenopausal and postmenopausal women. Random fine needle aspirates (rFNAs) of breast tissue were collected from 57 premenopausal and 55 postmenopausal women and were classified as normal weight, overweight, or obese. Expression levels of 21 target genes were determined using a TaqMan Low Density Array procedure. Breast tissue estradiol levels were measured by a liquid chromatography-tandem mass spectrometry procedure, and serum estradiol and follicle-stimulating hormone (FSH) were measured by a radioimmunoassay and an enzyme-linked immunosorbent assay, respectively. We found that in postmenopausal women, serum and tissue estradiol levels were increased in those who were overweight, and serum FSH levels were decreased in obese status. Interestingly, RPS6KB1, an AMPK downstream-responsive gene for protein synthesis and cell growth, and estrogen receptor α (encoded by the ESR1 gene) and its target gene GATA3 were significantly decreased in rFNA of premenopausal, obese women. In postmenopausal women, RPS6KB1, ESR1, and GATA3 expression remained unchanged in relation to adiposity. However, prostaglandin-endoperoxide synthase 2 (PTGS2), cyclin D1 (CCND1), and another ESR1 target gene, TFF1, were elevated in rFNA of obese postmenopausal women. Thus, as bodyweight increases, gene expression is indicative of increased proliferation in postmenopausal women but decreased proliferation in premenopausal women. Overall, our data reveal a novel process by which obesity promotes the risk of breast cancer in postmenopausal but not premenopausal women.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Pós-Menopausa/fisiologia , Pré-Menopausa/fisiologia , Adiposidade/fisiologia , Adulto , Feminino , Humanos , Inflamação/metabolismo , Inflamação/patologia , Pessoa de Meia-Idade , Obesidade/metabolismo , Sobrepeso/metabolismo , Fatores de Risco , Transcriptoma
2.
Oncogene ; 34(3): 323-33, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24413080

RESUMO

Aromatase inhibitors (AIs) are the standard endocrine therapy for postmenopausal breast cancer; however, currently used biomarkers, such as, estrogen receptor-alpha/progesterone receptor (ERα/PR), predict only slightly more than half of the potential responders to AI treatment. To identify novel markers of AI responsiveness, a genome-wide microarray analysis was performed using primary breast tumor samples from 50 postmenopausal women who later developed metastatic breast cancer. Sushi domain containing 3 (SUSD3) is a significantly differentially expressed gene, with 3.38-fold higher mRNA levels in AI-responsive breast tumors vs non-responders (P<0.001). SUSD3 was highly expressed in ERα-positive breast tumors and treatment with estradiol increased SUSD3 expression in ERα-positive breast cancer cells. Treatment with an antiestrogen or ERα knockdown abolished basal and estradiol-dependent SUSD3 expression. Recruitment of ERα upstream of the transcription start site of SUSD3 was demonstrated by chromatin immunoprecipitation-PCR. Flow cytometric analysis of SUSD3-knockdown cells revealed blunted estradiol effects on progression into S and M phases. SUSD3 was localized to the plasma membrane of breast cancer cells. SUSD3 knockdown decreased the appearance of actin-rich protrusions, stress fibers and large basal focal adhesions, while increasing the presence of cortical actin concomitant with a decrease in Rho and focal adhesion kinase activity. SUSD3-deficient cells demonstrated diminished cell spreading, cell-cell adhesion and motility. In conclusion, SUSD3 is a novel promoter of estrogen-dependent cell proliferation and regulator of cell-cell and cell-substrate interactions and migration in breast cancer. It may serve as a novel predictor of response to endocrine therapy and potential therapeutic target.


Assuntos
Citoesqueleto de Actina/metabolismo , Neoplasias da Mama/genética , Movimento Celular/genética , Estradiol/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Membrana/genética , Inibidores da Aromatase/farmacologia , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Estradiol/análogos & derivados , Antagonistas do Receptor de Estrogênio/farmacologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Feminino , Adesões Focais/genética , Fulvestranto , Perfilação da Expressão Gênica , Humanos , Células MCF-7 , Proteínas de Membrana/metabolismo , Microscopia Confocal , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Mol Endocrinol ; 28(8): 1304-15, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24992181

RESUMO

In endometriosis, stromal and epithelial cells from the endometrium form extrauterine lesions and persist in response to estrogen (E2) and prostaglandin E2 (PGE2). Stromal cells produce excessive quantities of estrogen and PGE2 in a feed-forward manner. However, it is unknown how estrogen stimulates cell proliferation and survival for the establishment and persistence of disease. Previous studies suggest that estrogen receptor-ß (ERß) is strikingly overexpressed in endometriotic stromal cells. Thus, we integrated genome-wide ERß binding data from previously published studies in breast cells and gene expression profiles in human endometriosis and endometrial tissues (total sample number = 81) and identified Ras-like, estrogen-regulated, growth inhibitor (RERG) as an ERß target. Estradiol potently induced RERG mRNA and protein levels in primary endometriotic stromal cells. Chromatin immunoprecipitation demonstrated E2-induced enrichment of ERß at the RERG promoter region. PGE2 via protein kinase A phosphorylated RERG and enhanced the nuclear translocation of RERG. RERG induced the proliferation of primary endometriotic cells. Overall, we demonstrated that E2/ERß and PGE2 integrate at RERG, leading to increased endometriotic cell proliferation and represents a novel candidate for therapeutic intervention.


Assuntos
Proliferação de Células , Dinoprostona/fisiologia , Endometriose/metabolismo , Receptor beta de Estrogênio/fisiologia , GTP Fosfo-Hidrolases/metabolismo , Adulto , Núcleo Celular/metabolismo , Endometriose/patologia , Estradiol/fisiologia , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Transporte Proteico
4.
Horm Cancer ; 4(6): 358-70, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23943160

RESUMO

Endometrial cancer is the fourth most common malignancy among women and is a major cause of morbidity contributing to approximately 8,200 annual deaths in the USA. Despite advances to the understanding of endometrial cancer, novel interventions for the disease are necessary given that many tumors become refractory to therapy. As a strategy to identify novel therapies for endometrial carcinoma, in this study, we examined the contribution of the peroxisome proliferator-activated receptor ß/δ (PPARß/δ) to endometrial cancer cell proliferation and apoptosis. We found that when activated with the highly selective PPARß/δ agonists, GW0742 and GW501516, PPARß/δ inhibited the proliferation and markedly induced the apoptosis of three endometrial cancer cell lines. The specificity of the PPARß/δ-induced effects on cell proliferation and apoptosis was demonstrated using PPARß/δ-selective antagonists and PPARß/δ small interfering RNA in combination with PPARß/δ-selective agonists. Furthermore, we showed that PPARß/δ activation increased phosphatase and tensin homolog expression, which led to protein kinase B (AKT) and glycogen synthase kinase-3ß (GSK3ß) dephosphorylation, and increased ß-catenin phosphorylation associated with its degradation. Overall, our data suggest that the antitumorigenic effect of PPARß/δ activation in endometrial cancer is mediated through the negative regulation of the AKT/GSK3ß/ß-catenin pathway. These findings warrant further investigation of PPARß/δ as a therapeutic target in endometrial cancer.


Assuntos
Carcinoma/tratamento farmacológico , Neoplasias do Endométrio/tratamento farmacológico , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Neoplasias do Endométrio/patologia , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Terapia de Alvo Molecular , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Receptores Ativados por Proliferador de Peroxissomo/genética , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia , beta Catenina/metabolismo
5.
Mol Cell Endocrinol ; 362(1-2): 85-90, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22705581

RESUMO

In the brain, a 36-kb distal promoter (I.f) regulates the Cyp19a1 gene that encodes aromatase, the key enzyme for estrogen biosynthesis. Local estrogen production in the brain regulates critical functions such as gonadotropin secretion and sexual behavior. The mechanisms that control brain aromatase production are not well understood. Here we show that the glucocorticoid dexamethasone robustly increases aromatase mRNA and protein by up to 98-fold in mouse hypothalamic cell lines in a dose- and time-dependent fashion. Using deletion mutants of the brain-specific promoter I.f and chromatin immunoprecipitation-PCR, we isolated a distinct region (-500/-200 bp) which becomes enriched in bound glucocorticoid receptor upon dexamethasone stimulation. A glucocorticoid antagonist or siRNA based knockdown of glucocorticoid receptor ablated dexamethasone stimulation of aromatase expression. Our findings demonstrate how glucocorticoids alter aromatase expression in the hypothalamus and might indicate a mechanism whereby glucocorticoid action modifies gonadotropin pulses and the menstrual cycle.


Assuntos
Aromatase/genética , Glucocorticoides/fisiologia , Hipotálamo/enzimologia , Regiões Promotoras Genéticas , Ativação Transcricional , Animais , Aromatase/metabolismo , Linhagem Celular , Dexametasona/farmacologia , Indução Enzimática , Técnicas de Silenciamento de Genes , Genes Reporter , Glucocorticoides/farmacologia , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Camundongos , Especificidade de Órgãos , Interferência de RNA , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Regulação para Cima
6.
Oncogene ; 29(8): 1114-22, 2010 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-19935720

RESUMO

Estrogen receptor-alpha (ERalpha) and its ligand estradiol (E2) has critical roles in breast cancer growth and are key therapeutic targets. In this study, we report a novel dual role of the adenosine A1 receptor (Adora1) as an E2/ERalpha target and a regulator of ERalpha transcriptional activity. In ERalpha-positive breast cancer cells, E2 upregulated Adora1 messenger RNA (mRNA) and protein levels, an effect that was reversed by the E2 antagonist ICI 182 780. Small interference RNA ablation of Adora1 in ERalpha-positive cells reduced basal and E2-dependent proliferation, whereas Adora1 over-expression in an ERalpha-negative cell line induced proliferation. The selective Adora1 antagonist, DPCPX, reduced proliferation, establishing Adora1 as a mediator of E2/ERalpha-dependent breast cancer growth. Intriguingly, Adora1 ablation decreased both mRNA and protein levels of ERalpha and, consequently, estrogen-responsive element-dependent ERalpha transcriptional activity. Moreover, Adora1 ablation decreased binding activity of ERalpha to the promoter of its target gene TFF1 and led to reduced TFF1 promoter activity and mRNA levels, suggesting that Adora1 is required for full transcriptional activity of ERalpha on E2 stimulation. Taken together, we showed a short feed-forward loop involving E2, ERalpha and Adora1 that favors breast cancer growth. These data suggest that Adora1 may represent an important target for therapeutic intervention in hormone-dependent breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/fisiologia , Estrogênios/farmacologia , Receptor A1 de Adenosina/fisiologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Receptor A1 de Adenosina/genética , Receptores de Estrogênio/metabolismo , Fator Trefoil-1 , Proteínas Supressoras de Tumor/fisiologia
7.
Ann N Y Acad Sci ; 1155: 121-31, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19250199

RESUMO

Epithelial-stromal interactions play key roles for aromatase expression and estrogen production in breast cancer tissue. Upregulated aromatase expression in breast fibroblasts increases the tissue concentration of estradiol (E2), which then activates a large number of carcinogenic genes via estrogen receptor-alpha (ERalpha) in malignant epithelial cells. This clinically pertains, since aromatase inhibitors (AIs) are the most effective hormonal treatment of ERalpha-positive breast tumors. A single gene encodes aromatase, the key enzyme in estrogen biosynthesis, the inhibition of which by an AI effectively eliminates E2 production. Since alternative promoters regulated by distinct signaling pathways control aromatase expression, it is possible to target these pathways and inhibit estrogen production in a tissue-selective fashion. We and others previously found that the majority of estrogen production in breast cancer tissue was accounted for by the aberrant activation of the proximal promoter I.3/II region. PGE(2) that is secreted in large amounts by malignant breast epithelial cells is the most potent known natural inducer of this promoter region in breast adipose fibroblasts. Signaling effectors/transcriptional regulators that mediate PGE(2) action include the activator pathways p38/CREB-ATF and JNK/jun and the inhibitory factor BRCA1 in breast adipose fibroblasts. Selective inhibition of this promoter region may treat breast cancer while permitting aromatase expression via alternative promoters in the brain and bone and thus obviate the key side effects of the current AIs. The signaling pathways that mediate the regulation of the promoter I.3/II region in undifferentiated fibroblasts in malignant breast tumors are reviewed.


Assuntos
Aromatase/genética , Neoplasias da Mama/enzimologia , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Animais , Aromatase/metabolismo , Proteína BRCA1/metabolismo , Sequência de Bases , Neoplasias da Mama/metabolismo , DNA , Receptor alfa de Estrogênio/metabolismo , Estrogênios/biossíntese , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Transcrição Gênica , Regulação para Cima
8.
Hum Reprod Update ; 12(1): 49-56, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16123052

RESUMO

Endometriosis is a common, chronic and estrogen-dependent gynaecological disorder associated with pelvic pain and infertility. In addition to, or perhaps as a consequence of, immune, environmental and genetic factors, endometriotic lesions show high estradiol (E(2)) biosynthesis and low E(2) inactivation compared with normal endometrium. Current medical therapies of pain, which aim to lower circulating E(2) concentrations, are not effective in at least half of these patients. We and others recently demonstrated the expression of a few steroidogenic genes in endometriosis. The most important genes in this group are steroidogenic acute regulatory protein (StAR) and aromatase. Both are essential for E(2) production. Prostaglandin E(2) (PGE(2)) is the most potent known stimulator of both StAR and aromatase. PGE(2) production in endometriosis is up-regulated by increased levels of the enzyme cyclo-oxygenase-2 (COX-2) in this tissue. COX-2 in turn is stimulated by E(2), interleukin-1beta (IL-1beta) and PGE(2) itself in endometrial and endometriotic cells. Thus, there is a positive feedback loop that favours continuous formation of E(2) and PGE(2) in endometriosis. These basic findings led to recent phase-II studies employing aromatase inhibitors in the treatment of endometriosis. Aromatase inhibitors treat both postmenopausal and premenopausal endometriosis at least as effectively as the existing medical treatments. In premenopausal women, we and others administered aromatase inhibitors in combination with an ovarian-suppressant treatment. In this review, we emphasize the most recent basic studies in detail and provide a short summary of recent clinical trials.


Assuntos
Aromatase/metabolismo , Endometriose/genética , Esteroides/metabolismo , Aromatase/genética , Inibidores da Aromatase/farmacologia , Células Cultivadas , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Endometriose/tratamento farmacológico , Endometriose/metabolismo , Endometriose/patologia , Estrogênios/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , Inflamação/etiologia , Fosfoproteínas/genética , Biossíntese de Proteínas , Transdução de Sinais , Células Estromais/metabolismo
9.
J Mol Endocrinol ; 31(1): 95-104, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12914528

RESUMO

We previously reported that human malignant endometrial epithelial cell conditioned medium (MECM) up-regulated cyclooxygenase (COX)-2 mRNA and protein levels in human normal endometrial stromal cells (ESC). Here we showed that pretreatment with a selective inhibitor of the extracellularly regulated kinase (ERK)1/2 signaling pathway blocked the MECM-induced COX-2 expression in ESC. Transient transfection assays indicated critical roles of a cAMP response element (CRE,-59/-53 bp) and a nuclear factor for interleukin (IL)-6 expression (NF-IL6) site (-132/-124 bp) in the regulation of basal and MECM-induced activity of COX-2 gene promoter in ESC. Employing electrophoretic mobility shift assays, we demonstrated that increased functional binding of CCAAT/enhancer binding protein (C/EBP)alpha, C/EBPbeta and upstream stimulatory factor-2 to the CRE and C/EBPalpha and C/EBPbeta to the NF-IL6 site were, at least in part, responsible for MECM-induced COX-2 expression in ESC. Moreover, overexpression of C/EBPalpha and C/EBPbeta significantly induced COX-2 promoter activity in ESC. Collectively, these results suggest that the basal and MECM-induced transcription of the COX-2 gene in ESC is regulated through a combination of the CRE and the NF-IL6 site by functional interactions of C/EBPalpha and C/EBPbeta.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Neoplasias do Endométrio/genética , Isoenzimas/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Prostaglandina-Endoperóxido Sintases/genética , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/genética , Meios de Cultivo Condicionados , Ciclo-Oxigenase 2 , Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/patologia , Indução Enzimática , Feminino , Humanos , Isoenzimas/biossíntese , Proteínas de Membrana , Mutagênese , Sondas de Oligonucleotídeos , Prostaglandina-Endoperóxido Sintases/biossíntese , Mapeamento por Restrição , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência , Células Estromais/citologia , Células Estromais/enzimologia , Células Estromais/patologia , Transfecção , Células Tumorais Cultivadas
10.
Mol Endocrinol ; 15(12): 2093-105, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11731611

RESUMO

Progesterone stimulates the expression of 17beta-hydroxysteroid dehydrogenase (HSD) type 2, which catalyzes the conversion of the potent estrogen, E2, to an inactive form, estrone, in epithelial cells of human endometrial tissue. Various effects of progesterone on uterine epithelium have recently been shown to be mediated by stromal PRs in mice. We describe herein a critical paracrine mechanism whereby progesterone induction of 17beta-HSD type 2 enzyme activity, transcript levels, and promoter activity in human endometrial epithelial cells are mediated primarily by PR in endometrial stromal cells. Medium conditioned with progestin-pretreated human endometrial stromal cells robustly increased 17beta-HSD type 2 enzyme activity (2-fold) and mRNA levels (13.2-fold) in Ishikawa malignant endometrial epithelial cells. In contrast, direct progestin treatment of Ishikawa epithelial cells gave rise to much smaller increases in enzyme activity (1.2-fold) and mRNA levels (4-fold). These results suggest that progesterone- dependent paracrine factors arising from stromal cells are primarily responsible for the induction of epithelial 17beta-HSD type 2 expression in the endometrium. We transfected serial deletion mutants of the -1,244 bp 5'-flanking region of the 17beta-HSD type 2 gene into Ishikawa cells. No progesterone response elements could be identified upstream of the 17beta-HSD type 2 promoter. Stromal PR-dependent induction of the 17beta-HSD type 2 promoter was mediated by a critical regulatory region mapped to the -200/-100 bp sequence. Direct treatment of Ishikawa cells with progestin gave rise to a maximal increase in the activity of -200 bp/Luciferase construct only by 1.2-fold, whereas medium conditioned by progestin-pretreated endometrial stromal cells increased promoter activity up to 2.4-fold in a time- and concentration-dependent manner. The stimulatory effect of medium conditioned by progestin-pretreated stromal cells was enhanced strikingly by increasing stromal cell PR levels with the addition of estrogen. This epithelial-stromal interaction was specific for endometrial epithelial cells, since 17beta-HSD type 2 could not be induced in malignant breast epithelial cells by media conditioned with progestin-treated breast or endometrial stromal cells. In conclusion, progesterone regulates the conversion of biologically active E2 to estrone by inducing the 17beta-HSD type 2 enzyme in human endometrial epithelium primarily via PR in stromal cells, which secrete factors that induce transcription mediated primarily by the -200/-100 bp 5'-regulatory region of the 17beta-HSD type 2 promoter.


Assuntos
17-Hidroxiesteroide Desidrogenases/biossíntese , Endométrio/fisiologia , Estradiol/fisiologia , Regulação da Expressão Gênica , Receptores de Progesterona/fisiologia , 17-Hidroxiesteroide Desidrogenases/análise , Células 3T3 , Animais , Sequência de Bases , Western Blotting , Feminino , Gonanos/farmacologia , Antagonistas de Hormônios/farmacologia , Humanos , Camundongos , Dados de Sequência Molecular , Congêneres da Progesterona/farmacologia , Promegestona/farmacologia , Células Estromais/metabolismo , Células Estromais/fisiologia , Células Tumorais Cultivadas
12.
J Am Acad Dermatol ; 45(3 Suppl): S116-24, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11511861

RESUMO

Estradiol production is most commonly thought of as an endocrine product of the ovary; however, there are many tissues that have the capacity to synthesize estrogens from androgen and to use estrogen in a paracrine or intracrine fashion. In addition, other organs such as the adipose tissue can contribute significantly to the circulating pool of estrogens. There is increasing evidence that in both men and women extraglandular production of C(18) steroids from C(19) precursors is important in normal physiology as well as in pathophysiologic states. The enzyme aromatase is found in a number of human tissues and cells, including ovarian granulosa cells, the placental syncytiotrophoblast, adipose and skin fibroblasts, bone, and the brain, and it locally catalyzes the conversion of C(19) steroids to estrogens. Aromatase expression in adipose tissue and possibly the skin primarily accounts for the extraglandular (peripheral) formation of estrogen and increases as a function of body weight and advancing age. Sufficient circulating levels of the biologically active estrogen estradiol can be produced as a result of extraglandular aromatization of androstenedione to estrone that is subsequently reduced to estradiol in peripheral tissues to cause uterine bleeding and endometrial hyperplasia and cancer in obese anovulatory or postmenopausal women. Extraglandular aromatase expression in adipose tissue and skin (via increasing circulating levels of estradiol) and bone (via increasing local estrogen concentrations) is of paramount importance in slowing the rate of postmenopausal bone loss. Moreover, excessive or inappropriate aromatase expression was demonstrated in adipose fibroblasts surrounding a breast carcinoma, endometriosis-derived stromal cells, and stromal cells in endometrial cancer, giving rise to increased local estrogen concentrations in these tissues. Whether systemically delivered or locally produced, elevated estrogen levels will promote the growth of these steroid-responsive tissues. Finally, local estrogen biosynthesis by aromatase activity in the brain may be important in the regulation of various cognitive and hypothalamic functions. The regulation of aromatase expression in human cells via alternatively used promoters, which can be activated or inhibited by various hormones, increases the complexity of estrogen biosynthesis in the human body. Aromatase expression is under the control of the classically located proximal promoter II in the ovary and a far distal promoter I.1 (40 kilobases upstream of the translation initiation site) in the placenta. In skin, the promoter is I.4. In adipose tissue, 2 other promoters (I.4 and I.3) located between I.1 and II are used in addition to the ovarian-type promoter II. In addition, promoter use in adipose fibroblasts switches between promoters II/I.3 and I.4 upon treatments of these cells with PGE(2) versus glucocorticoids plus cytokines. Moreover, the presence of a carcinoma in breast adipose tissue also causes a switch of promoter use from I.4 to II/I.3. Thus there can be complex mechanisms that regulate the extraglandular production of estrogen in a tissue-specific and state-specific fashion.


Assuntos
Estrogênios/biossíntese , Tecido Adiposo/metabolismo , Aromatase/genética , Estrogênios/metabolismo , Feminino , Expressão Gênica , Humanos , Ovário/metabolismo , Pele/metabolismo
14.
Cancer Res ; 61(5): 2250-5, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11280794

RESUMO

The dense layer of fibroblasts that accumulate around malignant breast epithelial cells (i.e., desmoplastic reaction) arises from the breast adipose tissue and provides structural and biochemical support for breast cancer. We report herein a number of epithelial-stromal interactions responsible for desmoplastic reaction in breast cancer using cultured 3T3-L1 murine fibroblasts and human adipose fibroblasts, which can be activated with a mixture of hormones to differentiate to mature adipocytes. Adipocyte differentiation was inhibited by coculturing fibroblasts with various breast cancer cell lines (T47D, MCF-7, SSC202, SSC78, and SSC30) completely or by breast cancer cell conditioned media in a dose-dependent manner; on the other hand, adipocyte differentiation was not inhibited by coculturing with normal human primary mammary epithelial cell conditioned medium. This tumor effect was eliminated using neutralizing antibodies against tumor necrosis factor (TNF)-alpha or interleukin (IL)-11. TNF-alpha and IL-11 levels were 2.5-3 times higher in T47D conditioned medium compared with control medium, and TNF-alpha transcripts were detectable in T47D but not in 3T3-L1 cells in culture, indicating that the malignant epithelial cell is the major site of cytokine production. This was confirmed in vivo in mastectomy specimens, where immunoreactive TNF-alpha and IL-11 were readily detectable in malignant epithelial cells but not in the majority of the surrounding fibroblasts. Adipocyte differentiation is mediated by the expression of a cascade of adipogenic transcription factors, including CCAAT/enhancer binding protein (C/EBP)beta, C/EBPdelta, peroxisome proliferator-activated receptor (PPAR)gamma and C/EBPalpha. C/EBPalpha and PPARgamma are essential for this process. We demonstrated by Northern analysis that exposure of activated 3T3-L1 cells to T47D cell conditioned medium strikingly decreased the levels of PPARgamma and C/EBPalpha transcripts and increased the levels of C/EBPbeta and C/EBPdelta transcripts. In these 3T3-L1 cells, inhibition of differentiation was also confirmed by markedly suppressed levels of aP2 mRNA, which is an adipocyte-specific gene. These in vitro observations were confirmed in sections of human malignant breast tumors, where immunoreactive C/EBPalpha was readily detectable in adipose flbroblasts distant to the tumor but not in intratumoral fibroblasts. Treatment of 3T3-L1 cells with T47D cell conditioned medium or TNF-alpha changed neither the numbers of cells in G0-G1, S, and G2 phases nor the rate of [3H]thymidine incorporation, thus ruling out a proliferative effect of malignant cells on the surrounding fibroblasts. In summary, desmoplastic reaction primarily occurs via the action of cytokines (TNF-alpha and IL-11) secreted by the malignant epithelial cells to inhibit differentiation of adipose fibroblasts to mature adipocytes. This tumor-induced block in adipocyte differentiation is mediated by the selective inhibition of expression of the essential adipogenic transcription factors, i.e., PPARgamma and C/EBPalpha.


Assuntos
Adipócitos/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteína alfa Estimuladora de Ligação a CCAAT/biossíntese , Comunicação Celular/fisiologia , Interleucina-11/metabolismo , Receptores Citoplasmáticos e Nucleares/biossíntese , Fatores de Transcrição/biossíntese , Fator de Necrose Tumoral alfa/metabolismo , Células 3T3 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Mama/citologia , Mama/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/antagonistas & inibidores , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Diferenciação Celular/fisiologia , Técnicas de Cocultura , Proteínas de Ligação a DNA/biossíntese , Regulação para Baixo/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Interleucina-11/fisiologia , Camundongos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Células Estromais/metabolismo , Células Estromais/patologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/fisiologia
15.
Cancer Res ; 61(5): 2328-34, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11280806

RESUMO

Expression of aromatase P450 (P450arom), which catalyzes the formation of estrogens, is aberrantly increased in adipose fibroblasts surrounding breast carcinomas, giving rise to proliferation of malignant cells. Aromatase in human adipose tissue is primarily expressed in undifferentiated fibroblasts under the control of several distinct and alternatively used P450arom promoters. In tumor-free breast adipose tissue, P450arom is usually expressed at low levels via a distal promoter (I.4), whereas in the breast adipose tissue bearing a tumor, P450arom is increased through the activation of two proximal promoters, II and I.3. Because the in vivo activation of P450arom promoter II is a key event responsible for aberrantly high P450arom expression in breast tumors, we studied the molecular basis for the enhancement of P450arom promoter II using human adipose fibroblasts (HAFs) in primary culture treated with T47D breast cancer cell-conditioned medium (TCM) as a model system. Upon treatment with TCM, HAFs displayed a striking induction of P450arom mRNA levels via promoter II usage. This effect appeared to be specific for malignant breast epithelial cells, because conditioned media from breast cancer cell lines T47D and MCF-7 induced promoter II activity, whereas normal breast epithelial cells or liver or prostate cancer cell lines did not produce such an effect. Although treatment with a cyclic AMP analogue also caused a switch in the promoter use from I.4 to II in cultured HAFs, TCM-induced promoter II use was found to be mediated via a cyclic AMP-independent pathway. Use of serial deletion mutants of the promoter II 5'-flanking sequence revealed the presence of critical cis-acting elements in the -517/-278 bp region, which regulate the baseline activity. TCM caused a 5.7-fold induction of the -517-bp promoter II construct, whereas site-directed mutagenesis of a CCAAT/enhancer binding protein (C/EBP) binding site (-317/-304 bp) abolished both baseline and TCM-induced activities. Ectopic expressions of C/EBPalpha and C/EBPbeta, but not C/EBPdelta, significantly induced promoter II activity. Moreover, we demonstrated the presence of both C/EBPbeta and C/EBPdelta but not C/EBPalpha in a DNA-protein complex formed by the nuclear extract from TCM-treated HAFs and a probe containing this critical C/EBP binding element (-317/-304 bp). Finally, treatment of HAFs with TCM strikingly induced C/EBPbeta expression, whereas this did not affect the levels of C/EBPalpha or C/EBPdelta transcripts. In conclusion, malignant breast epithelial cells secrete factors, which induce aromatase expression in adipose fibroblasts via promoter II. This is, at least in part, mediated by a TCM-induced up-regulation and enhanced binding of C/EBPbeta to a promoter II regulatory element.


Assuntos
Tecido Adiposo/enzimologia , Aromatase/biossíntese , Neoplasias da Mama/patologia , Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Comunicação Celular/fisiologia , Fibroblastos/enzimologia , Regiões Promotoras Genéticas/fisiologia , Tecido Adiposo/citologia , Aromatase/genética , Sequência de Bases , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Células Cultivadas , Meios de Cultivo Condicionados , AMP Cíclico/biossíntese , AMP Cíclico/fisiologia , Células Epiteliais/citologia , Feminino , Fibroblastos/citologia , Regulação Neoplásica da Expressão Gênica , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Células Estromais/citologia , Regulação para Cima
16.
J Steroid Biochem Mol Biol ; 79(1-5): 19-25, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11850203

RESUMO

Aromatase is the key enzyme for estrogen biosynthesis. It is normally expressed in the human ovary, skin, adipose tissue and brain. Aromatase activity is not detectable in normal endometrium. In contrast, aromatase is expressed aberrantly in endometriosis and is stimulated by PGE2. This results in local production of estrogen, which induces PGE2 formation and establishes a positive feedback cycle. Another abnormality in endometriosis, i.e. deficient 17beta-hydroxysteroid dehydrogenase (17beta-HSD) type 2 expression, impairs the inactivation of estradiol to estrone. These molecular aberrations collectively favor accumulation of increasing quantities of estradiol and PGE2 in endometriosis. The clinical relevance of these findings was exemplified by the successful treatment of an unusually aggressive case of post-menopausal endometriosis using an aromatase inhibitor.


Assuntos
Aromatase/metabolismo , Endometriose/enzimologia , Aromatase/genética , Inibidores da Aromatase , Dinoprostona/metabolismo , Endometriose/tratamento farmacológico , Endometriose/genética , Endometriose/metabolismo , Inibidores Enzimáticos/uso terapêutico , Estradiol/metabolismo , Estrogênios/biossíntese , Estrogênios/metabolismo , Estrona/metabolismo , Feminino , Expressão Gênica , Hormônio Liberador de Gonadotropina/agonistas , Humanos , Modelos Biológicos , Ductos Paramesonéfricos/enzimologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Estromais/enzimologia
17.
Hum Reprod Update ; 6(5): 413-8, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11045871

RESUMO

Although treatment of one unusually aggressive case of postmenopausal endometriosis with an aromatase inhibitor has been strikingly successful, large clinical trials are required to establish whether aromatase inhibitors will have a significant role in the medical management of endometriosis. Introduction of aromatase inhibitors into the treatment of endometriosis underscores the importance of basic research leading to the development of novel strategies in reproductive disorders. It was shown earlier that aromatase activity was not detectable in normal endometrium. Aromatase, however, is expressed inappropriately in endometriosis and stimulated by prostaglandin E2. Aromatase activity gives rise to local biosynthesis of oestrogen, which, in turn, stimulates prostaglandin E2 production, thus establishing a positive feedback cycle. This favours accumulation of oestrogen and prostaglandins in endometriosis, which is an inflammatory disorder dependent on oestrogen for growth.


Assuntos
Inibidores da Aromatase , Endometriose/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Aromatase/genética , Fator I de Transcrição COUP , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dinoprostona/biossíntese , Endometriose/etiologia , Endometriose/metabolismo , Estrogênios/biossíntese , Feminino , Fatores de Transcrição Fushi Tarazu , Expressão Gênica , Proteínas de Homeodomínio , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares , Fator Esteroidogênico 1 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Útero/enzimologia
18.
J Clin Endocrinol Metab ; 85(8): 2897-902, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10946900

RESUMO

We previously demonstrated that 17beta hydroxysteroid dehydrogenase type 2, the enzyme that inactivates estradiol to estrone, is expressed in luteal eutopic endometrium in response to progesterone but not in simultaneously biopsied peritoneal endometriotic tissue. This molecular evidence of progesterone resistance, together with the clinical observation of resistance of endometriosis to treatment with progestins, led us to determine the levels of progesterone receptor (PR) isoforms PR-A and PR-B in eutopic endometrial and extra-ovarian endometriotic tissues. It was proposed that progesterone action on target genes is mediated primarily by homodimers of PR-B, whereas the truncated variant PR-A acts as a repressor of PR-B function. Immunoprecipitation, followed by Western blot analysis, was performed to detect bands specific for PR-A and PR-B in paired samples of endometriotic and eutopic endometrial tissues simultaneously biopsed from 18 women undergoing laparoscopy during various phases of the menstrual cycle. PR-B was present in 17 of 18 eutopic endometrial samples, and its level increased in the preovulatory phase, as expected, whereas PR-A was detected in all samples (n = 18) with a similar, but less prominent, cyclic variation in its levels. In endometriotic samples, however, no detectable PR-B could be demonstrated, whereas PR-A was detected in all samples (n = 18), albeit in much lower levels and without any cyclic variation in contrast with the eutopic endometrium. Levels of PR-A and PR-B in endometriotic and eutopic endometrial tissues were determined and compared after normalization to total protein and estrogen receptor-alpha levels. Using RNase protection assay, we also demonstrated indirectly that only PR-A transcripts were present in endometriotic tissue samples (n = 8), whereas both PR-A and PR-B transcripts were readily detectable in all eutopic endometrial samples (n = 8). This was indicative that failure to detect PR-B protein in endometriotic tissues is due to the absence of PR-B transcripts. We conclude that progesterone resistance in endometriotic tissue from laboratory and clinical observations may be accounted for by the presence of the inhibitory PR isoform PR-A and the absence of the stimulatory isoform PR-B.


Assuntos
Endometriose/genética , Endométrio/metabolismo , Ciclo Menstrual , Receptores de Progesterona/genética , Adulto , Western Blotting , Dimerização , Endometriose/patologia , Endométrio/química , Endométrio/patologia , Feminino , Humanos , RNA Mensageiro/análise , Receptores de Progesterona/análise , Transcrição Gênica
19.
J Mol Endocrinol ; 25(1): 35-42, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10915216

RESUMO

Conversion of C(19) steroids to estrogens is catalyzed by aromatase in human ovary, placenta and extraglandular tissues such as adipose tissue, skin and the brain. Aromatase activity is not detectable in normal endometrium. In contrast, aromatase is expressed aberrantly in endometriosis and is stimulated by prostaglandin E(2) (PGE(2)).( )This results in local production of estrogen, which induces PGE(2) formation and establishes a positive feedback cycle. Another abnormality in endometriosis, i.e. deficient hydroxysteroid dehydrogenase (17beta-HSD) type 2 expression, impairs the inactivation of estradiol to estrone. These molecular aberrations collectively favor accumulation of increasing quantities of estradiol and PGE(2 )in endometriosis. The clinical relevance of these findings was exemplified by the successful treatment of an unusually aggressive case of postmenopausal endometriosis using an aromatase inhibitor.


Assuntos
Endometriose/metabolismo , Estrogênios/biossíntese , Anastrozol , Aromatase/genética , Aromatase/metabolismo , Inibidores da Aromatase , Dinoprostona/metabolismo , Endometriose/tratamento farmacológico , Endometriose/genética , Inibidores Enzimáticos/uso terapêutico , Estrona/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Ductos Paramesonéfricos/enzimologia , Nitrilas/uso terapêutico , Gravidez , Triazóis/uso terapêutico
20.
Am J Obstet Gynecol ; 182(4): 767-75, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10764452

RESUMO

OBJECTIVE: Although an association between dioxin exposure and endometriosis has been proposed, the effects of this environmental toxin on human endometriosis are not known. To understand the potential underlying molecular mechanisms we studied the expressions of cytochrome P-450 genes (CYP1A1, CYP1A2, and CYP1B1 ), which are induced by dioxin, and the expressions of cytosolic receptor for dioxin, aryl hydrocarbon receptor, and its nuclear translocator, aryl hydrocarbon receptor nuclear translocator protein, in endometriotic and eutopic endometrial tissues. STUDY DESIGN: Levels of transcripts of CYP1A1, CYP1A2, CYP1B1, aryl hydrocarbon receptor, and aryl hydrocarbon receptor nuclear translocator protein were determined by a quantitative reverse transcriptase-polymerase chain reaction and Southern blot assay in total ribonucleic acid samples from endometriotic and eutopic endometrial tissues. Eutopic endometrial tissue samples (n = 33) and endometriotic tissue samples (n = 10) were obtained at the time of uterine curettage and laparoscopy from disease-free women and from patients with endometriosis. Portions of these eutopic endometrial and endometriotic tissues were obtained simultaneously from the same patients (n = 8 pairs of samples). Levels of transcripts of CYP1A1, CYP1A2, CYP1B1, aryl hydrocarbon receptor, and aryl hydrocarbon receptor nuclear translocator protein were determined in endometrial and endometriotic tissues during follicular and luteal phases of the cycle and in cultured endometriotic stromal cells treated with forskolin, phorbol diacetate, medroxyprogesterone acetate,and serum. RESULTS: Transcripts of dioxin receptor, its nuclear translocator, and two dioxin-induced target genes (CYP1A2 and CYP1B1) were demonstrated during follicular and luteal phases of the cycle in both eutopic endometrial tissues and tissues affected by pelvic endometriosis, with no readily detectable differences between these tissues. On the other hand, levels of transcripts of another dioxin-induced gene, CYP1A1, were found to be strikingly higher in endometriotic tissues than in the eutopic endometrium. Mean levels in endometriotic tissues were 8.7 times those found in eutopic endometrium. Various hormonal treatments of endometriotic stromal cells did not significantly alter these levels. CONCLUSION: We demonstrated for the first time the expression of dioxin-related transcription factors aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator protein and target genes CYP1A1, CYP1A2, and CYP1B1 in endometriotic tissues and stromal cells. Strikingly elevated CYP1A1 transcripts in endometriosis may give rise to significantly increased P-4501A1 enzyme activity and thus promote the development and growth of endometriosis by either activating procarcinogens or inducing the formation of catechol estrogens or both. In fact, the proposed link between dioxin exposure and endometriosis may be explained in part by the up-regulation of the CYP1A1 gene expression in endometriotic tissues.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Sistema Enzimático do Citocromo P-450/genética , Proteínas de Ligação a DNA , Dioxinas/farmacologia , Endometriose/genética , Endometriose/metabolismo , Endométrio/fisiologia , Expressão Gênica , Transativadores/metabolismo , Translocador Nuclear Receptor Aril Hidrocarboneto , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1B1 , Endométrio/metabolismo , Feminino , Humanos , Família Multigênica , Receptores de Hidrocarboneto Arílico/genética , Valores de Referência , Fatores de Transcrição/genética , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...