Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Mol Carcinog ; 56(2): 580-593, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27341075

RESUMO

Zinc (Zn) is an essential trace mineral that contributes to the regulation of several cellular functions; however, it may be also implicated in the progression of breast cancer through different mechanisms. It has been largely reported that the classical estrogen receptor (ER), as well as the G protein estrogen receptor (GPER, previously known as GPR30) can exert a main role in the development of breast tumors. In the present study, we demonstrate that zinc chloride (ZnCl2 ) involves GPER in the activation of insulin-like growth factor receptor I (IGF-IR)/epidermal growth factor receptor (EGFR)-mediated signaling, which in turn triggers downstream pathways like ERK and AKT in breast cancer cells, and main components of the tumor microenvironment namely cancer-associated fibroblasts (CAFs). Further corroborating these findings, ZnCl2 stimulates a functional crosstalk of GPER with IGF-IR and EGFR toward the transcription of diverse GPER target genes. Then, we show that GPER contributes to the stimulatory effects induced by ZnCl2 on cell-cycle progression, proliferation, and migration of breast cancer cells as well as migration of CAFs. Together, our data provide novel insights into the molecular mechanisms through which zinc may exert stimulatory effects in breast cancer cells and CAFs toward tumor progression. © 2016 Wiley Periodicals, Inc.


Assuntos
Neoplasias da Mama/metabolismo , Fibroblastos Associados a Câncer/patologia , Cloretos/metabolismo , Receptores ErbB/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Somatomedina/metabolismo , Transdução de Sinais , Compostos de Zinco/metabolismo , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Feminino , Humanos
2.
J Biol Chem ; 287(52): 43234-45, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23135268

RESUMO

Activation of lipid metabolism is an early event in carcinogenesis and a central hallmark of many tumors. Fatty acid synthase (FASN) is a key lipogenic enzyme catalyzing the terminal steps in the de novo biogenesis of fatty acids. In cancer cells, FASN may act as a metabolic oncogene, given that it confers growth and survival advantages to these cells, whereas its inhibition effectively and selectively kills tumor cells. Hormones such as estrogens and growth factors contribute to the transcriptional regulation of FASN expression also through the activation of downstream signaling and a cross-talk among diverse transduction pathways. In this study, we demonstrate for the first time that 17ß-estradiol (E2) and the selective GPER ligand G-1 regulate FASN expression and activity through the GPER-mediated signaling, which involved the EGF receptor/ERK/c-Fos/AP1 transduction pathway, as ascertained by using specific pharmacological inhibitors, performing gene-silencing experiments and ChIP assays in breast SkBr3, colorectal LoVo, hepatocarcinoma HepG2 cancer cells, and breast cancer-associated fibroblasts. In addition, the proliferative effects induced by E2 and G-1 in these cells involved FASN as the inhibitor of its activity, named cerulenin, abolished the growth response to both ligands. Our data suggest that GPER may be included among the transduction mediators involved by estrogens in regulating FASN expression and activity in cancer cells and cancer-associated fibroblasts that strongly contribute to cancer progression.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Ácido Graxo Sintase Tipo I/biossíntese , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Cerulenina/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Ácido Graxo Sintase Tipo I/antagonistas & inibidores , Ácido Graxo Sintase Tipo I/genética , Inibidores da Síntese de Ácidos Graxos/farmacologia , Feminino , Fibroblastos/patologia , Células Hep G2 , Humanos , Neoplasias/genética , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
3.
Ann Vasc Surg ; 27(6): 762-6, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23809843

RESUMO

BACKGROUND: Breast cancer (BC) and chronic venous disease (CVD) are in some way related to hormonal effects, and often the clinical manifestations of CVD intersect with the clinical course of BC. This article describes the correlations between these clinical conditions. METHODS: A total of 1138 female patients with BC were retrospectively reviewed in a 5-year period to obtain clinical information about the frequency and characteristics of contemporary CVD and the relative correlations with estrogen and progesterone receptor status. RESULTS: The presence of BC was associated with concomitant CVD clinical manifestations in patients with positive estrogen receptor status, whereas no association was found in patients with negative estrogen receptor status. The presence of negative estrogen receptor status associated with positive progesterone receptor status seemed to be even protective against CVD. Patients with more severe manifestations of CVD had positive estrogen receptor status. CONCLUSIONS: BC and CVD seem to be strongly associated. Positive estrogen receptor status may predispose to a more severe clinical course of venous disease when it occurs in patients with BC.


Assuntos
Neoplasias da Mama/complicações , Receptores de Estrogênio/sangue , Receptores de Progesterona/sangue , Insuficiência Venosa/complicações , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Neoplasias da Mama/sangue , Neoplasias da Mama/epidemiologia , Feminino , Seguimentos , Humanos , Incidência , Itália/epidemiologia , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Insuficiência Venosa/sangue , Insuficiência Venosa/epidemiologia
4.
Breast Cancer Res Treat ; 133(3): 1025-35, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22147081

RESUMO

Early growth response-1 (Egr-1) is an immediate early gene involved in relevant biological events including the proliferation of diverse types of cell tumors. In a microarray analysis performed in breast cancer cells, 17ß-estradiol (E2) and the estrogen receptor antagonist 4-hydroxitamoxifen (OHT) up-regulated Egr-1 through the G protein-coupled receptor named GPR30/GPER. Hence, in this study, we aimed to provide evidence regarding the ability of E2, OHT and the selective GPER ligand G-1 to regulate Egr-1 expression and function through the GPER/EGFR/ERK transduction pathway in both Ishikawa (endometrial) and SkBr3 (breast) cancer cells. Interestingly, we demonstrate that Egr-1 is involved in the transcription of genes regulating cell proliferation like CTGF and cyclin D1 and required for the proliferative effects induced by E2, OHT, and G-1 in both Ishikawa and SkBr3 cells. In addition, we show that GPER mediates the expression of Egr-1 also in carcinoma-associated fibroblasts (CAFs). Our data suggest that Egr-1 may represent an important mediator of the biological effects induced by E2 and OHT through GPER/EGFR/ERK signaling in breast and endometrial cancer cells. The results obtained in CAFs provide further evidence regarding the potential role exerted by the GPER-dependent Egr-1 up-regulation in tumor development and progression. Therefore, Egr-1 may be included among the bio-markers of estrogen and antiestrogen actions and may be considered as a further therapeutic target in both breast and endometrial tumors.


Assuntos
Neoplasias da Mama/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Neoplasias do Endométrio/genética , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Tamoxifeno/análogos & derivados , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo/genética , Ciclina D1/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Neoplasias do Endométrio/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas/efeitos dos fármacos , Transdução de Sinais , Tamoxifeno/farmacologia
5.
Environ Health Perspect ; 116(12): 1648-55, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19079715

RESUMO

BACKGROUND: Atrazine, one of the most common pesticide contaminants, has been shown to up-regulate aromatase activity in certain estrogen-sensitive tumors without binding or activating the estrogen receptor (ER). Recent investigations have demonstrated that the orphan G-protein-coupled receptor 30 (GPR30), which is structurally unrelated to the ER, mediates rapid actions of 17beta-estradiol and environmental estrogens. OBJECTIVES: Given the ability of atrazine to exert estrogen-like activity in cancer cells, we evaluated the potential of atrazine to signal through GPR30 in stimulating biological responses in cancer cells. METHODS AND RESULTS: Atrazine did not transactivate the endogenous ERalpha in different cancer cell contexts or chimeric proteins encoding the ERalpha and ERbeta hormone-binding domain in gene reporter assays. Moreover, atrazine neither regulated the expression of ERalpha nor stimulated aromatase activity. Interestingly, atrazine induced extracellular signal-regulated kinase (ERK) phosphorylation and the expression of estrogen target genes. Using specific signaling inhibitors and gene silencing, we demonstrated that atrazine stimulated the proliferation of ovarian cancer cells through the GPR30-epidermal growth factor receptor transduction pathway and the involvement of ERalpha. CONCLUSIONS: Our results indicate a novel mechanism through which atrazine may exert relevant biological effects in cancer cells. On the basis of the present data, atrazine should be included among the environmental contaminants potentially able to signal via GPR30 in eliciting estrogenic action.


Assuntos
Atrazina/toxicidade , Proliferação de Células , Herbicidas/toxicidade , Neoplasias Ovarianas/patologia , Receptores de Estrogênio/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Aromatase/metabolismo , Sequência de Bases , Proliferação de Células/efeitos dos fármacos , Primers do DNA , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Neoplasias Ovarianas/fisiopatologia , Fosforilação , Células Tumorais Cultivadas
7.
J Exp Clin Cancer Res ; 37(1): 94, 2018 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-29716623

RESUMO

BACKGROUND: MicroRNA (miRNAs) are non-coding small RNA molecules that regulate gene expression by inhibiting the translation of target mRNAs. Among several dysregulated miRNAs in human cancer, the up-regulation of miR-221 has been associated with development of a variety of hematologic and solid malignancies. In this study, we investigated the involvement of miR-221 in breast cancer. METHODS: TaqMan microRNA assay was used to detect the miR-221 levels in normal cells and in MDA-MB 231 and SkBr3 breast cancer cells as well as in main players of the tumor microenvironment, namely cancer-associated fibroblasts (CAFs). miR-221 mimic sequence and locked nucleic acid (LNA)-i-miR-221 construct were used to induce or inhibit, respectively, the miR-221 expression in cells used. Quantitative PCR and western blotting analysis were performed to evaluate the levels of the miR-221 target gene A20 (TNFAIP3), as well as the member of the NF-kB complex namely c-Rel and the connective tissue growth factor (CTGF). Chromatin immunoprecipitation (ChIP) assay was performed to ascertain the recruitment of c-Rel to the CTFG promoter. Finally, the cell growth and migration in the presence of LNA-i-miR-221 or silencing c-Rel and CTGF by specific short hairpin were assessed by cell count, colony formation and boyden chambers assays. Statistical analysis was performed by ANOVA. RESULTS: We first demonstrated that LNA-i-miR-221 inhibits both endogenous and ectopic expression of miR-221 in our experimental models. Next, we found that the A20 down-regulation, as well as the up-regulation of c-Rel induced by miR-221 were no longer evident using LNA-i-miR-221. Moreover, we established that the miR-221 dependent recruitment of c-Rel to the NF-kB binding site located within the CTGF promoter region is prevented by using LNA-i-miR-221. Furthermore, we determined that the up-regulation of CTGF mRNA and protein levels by miR-221 is no longer evident using LNA-i-miR221 and silencing c-Rel. Finally, we assessed that cell growth and migration induced by miR-221 in MDA-MB 231 and SkBr3 breast cancer cells as well as in CAFs are abolished by LNAi-miR-221 and silencing c-Rel or CTGF. CONCLUSIONS: Overall, these data provide novel insights into the stimulatory action of miR-221 in breast cancer cells and CAFs, suggesting that its inhibition may be considered toward targeted therapeutic approaches in breast cancer patients.


Assuntos
Neoplasias da Mama/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibroblastos/metabolismo , MicroRNAs/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Transdução de Sinais , Transfecção
8.
Oncotarget ; 8(63): 106608-106624, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29290975

RESUMO

The cytochrome P450 1B1 (CYP1B1) is a heme-thiolate monooxygenase involved in both estrogen biosynthesis and metabolism. For instance, CYP1B1 catalyzes the hydroxylation of E2 leading to the production of 4-hydroxyestradiol that may act as a potent carcinogenic agent. In addition, CYP1B1 is overexpressed in different tumors including breast cancer. In this scenario, it is worth mentioning that CYP1B1 expression is triggered by estrogens through the estrogen receptor (ER)α in breast cancer cells. In the present study, we evaluated whether the G protein estrogen receptor namely GPER may provide an alternate route toward the expression and function of CYP1B1 in ER-negative breast cancer cells, in main players of the tumor microenvironment as cancer associated fibroblasts (CAFs) that were obtained from breast cancer patients, in CAFs derived from a cutaneous metastasis of an invasive mammary ductal carcinoma and in breast tumor xenografts. Our results show that GPER along with the EGFR/ERK/c-Fos transduction pathway can lead to CYP1B1 regulation through the involvement of a half-ERE sequence located within the CYP1B1 promoter region. As a biological counterpart, we found that both GPER and CYP1B1 mediate growth effects in vitro and in vivo. Altogether, our data suggest that estrogens in ER-negative cell contexts may engage the alternate GPER signaling toward CYP1B1 regulation. Estrogen-CYP1B1 landscape via GPER should be taken into account in setting novel pharmacological approaches targeting breast cancer development.

9.
Sci Rep ; 6: 24354, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-27072893

RESUMO

Cancer-associated fibroblasts (CAFs) contribute to the malignant aggressiveness through secreted factors like IL1ß, which may drive pro-tumorigenic inflammatory phenotypes mainly acting via the cognate receptor named IL1R1. Here, we demonstrate that signalling mediated by the G protein estrogen receptor (GPER) triggers IL1ß and IL1R1 expression in CAFs and breast cancer cells, respectively. Thereby, ligand-activation of GPER generates a feedforward loop coupling IL1ß induction by CAFs to IL1R1 expression by cancer cells, promoting the up-regulation of IL1ß/IL1R1 target genes such as PTGES, COX2, RAGE and ABCG2. This regulatory interaction between the two cell types induces migration and invasive features in breast cancer cells including fibroblastoid cytoarchitecture and F-actin reorganization. A better understanding of the mechanisms involved in the regulation of pro-inflammatory cytokines by GPER-integrated estrogen signals may be useful to target these stroma-cancer interactions.


Assuntos
Neoplasias da Mama/metabolismo , Interleucina-1beta/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Transdução de Sinais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Fibroblastos/metabolismo , Humanos
10.
Oncotarget ; 6(18): 16573-87, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26030000

RESUMO

MicroRNAs (miRNAs) are small non coding RNA molecules that play a crucial role in several pathophysiological conditions, including cancer. The stimulation of hormone-sensitive tumors by estrogens are mediated by estrogen receptor (ER)α and G protein estrogen receptor (GPER). Previous studies have reported that ERα regulates miRNA expression, while this ability of GPER remains to be elucidated. Here, we demonstrate that in SkBr3 breast cancer and HepG2 hepatocarcinoma cells, 17ß-estradiol (E2) and the selective GPER ligand G-1 induce miR144 expression through GPER and the involvement of the PI3K/ERK1/2/Elk1 transduction pathway. Moreover, we show that E2 and G-1 down-regulate through miR144 the onco-suppressor Runx1 and increase cell cycle progression. The capability of E2 and G-1 in triggering the induction of miR144 and the down-regulation of Runx1 was also confirmed in cancer-associated fibroblasts (CAFs) that are main components of the tumor microenvironment driving cancer progression. Further confirming these results, Runx1 protein levels were found decreased in tumor xenografts upon G-1 treatment. On the basis of our findings miR144 and Runx1 may be included among the oncotargets of GPER action. Moreover, the present data provide new insights regarding the ability of estrogens to trigger the GPER/miR144/Runx1 transduction pathway toward the stimulation of cancer progression.


Assuntos
Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , MicroRNAs/genética , Neoplasias/patologia , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Linhagem Celular Tumoral , Subunidade alfa 2 de Fator de Ligação ao Core/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Xenoenxertos , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , MicroRNAs/biossíntese , Transplante de Neoplasias , Neoplasias/genética , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/genética , Microambiente Tumoral/fisiologia , Proteínas Elk-1 do Domínio ets/metabolismo
11.
Environ Health Perspect ; 123(5): 493-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25616260

RESUMO

BACKGROUND: The pesticide atrazine does not bind to or activate the classical estrogen receptor (ER), but it up-regulates the aromatase activity in estrogen-sensitive tumor cells. The G protein estrogen receptor (GPR30/GPER) has been reported to be involved in certain biological responses to endogenous estrogens and environmental compounds exerting estrogen-like activity. OBJECTIVES: We aimed to evaluate the potential of atrazine to trigger GPER-mediated signaling in cancer cells and cancer-associated fibroblasts (CAFs). METHODS AND RESULTS: Using gene reporter assays in diverse types of cancer cells, we found that atrazine did not transactivate endogenous ERα or chimeric proteins that encode the ERα and ERß hormone binding domains. Conversely, atrazine was able to bind to GPER to induce ERK activation and the expression of estrogen target genes, which, interestingly, appeared to rely on both GPER and ERα expression. As a biological counterpart, atrazine stimulated the proliferation of ovarian cancer cells that depend on GPER and ERα, as evidenced by gene silencing experiments and the use of specific signaling inhibitors. Of note, through GPER, atrazine elicited ERK phosphorylation, gene expression, and migration in CAFs, thus extending its stimulatory role to these main players of the tumor microenvironment. CONCLUSIONS: Our results suggest a novel mechanism through which atrazine may exert relevant biological effects in cancer cells and CAFs. On the basis of our data, atrazine should be included among the environmental contaminants that may elicit estrogenic activity through GPER-mediated signaling.


Assuntos
Atrazina/farmacologia , Receptor alfa de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos
12.
Dis Model Mech ; 8(10): 1237-46, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26183213

RESUMO

Estrogens regulate numerous pathophysiological processes, mainly by binding to and activating estrogen receptor (ER)α and ERß. Increasing amounts of evidence have recently demonstrated that G-protein coupled receptor 30 (GPR30; also known as GPER) is also involved in diverse biological responses to estrogens both in normal and cancer cells. The classical ER and GPER share several features, including the ability to bind to identical compounds; nevertheless, some ligands exhibit opposed activity through these receptors. It is worth noting that, owing to the availability of selective agonists and antagonists of GPER for research, certain differential roles elicited by GPER compared with ER have been identified. Here, we provide evidence on the molecular mechanisms through which a calixpyrrole derivative acts as a GPER antagonist in different model systems, such as breast tumor cells and cancer-associated fibroblasts (CAFs) obtained from breast cancer patients. Our data might open new perspectives toward the development of a further class of selective GPER ligands in order to better dissect the role exerted by this receptor in different pathophysiological conditions. Moreover, calixpyrrole derivatives could be considered in future anticancer strategies targeting GPER in cancer cells.


Assuntos
Modelos Biológicos , Modelos Moleculares , Pirróis/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Bioensaio , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Fibroblastos/metabolismo , Humanos , Ligantes , Neoplasias/patologia , Pirróis/química , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Xenopus
13.
Anticancer Res ; 22(3): 1681-8, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12168854

RESUMO

The effects of low power millimetric wave (MMW) radiation on the growth of tumor and healthy cells were studied. A wide-band frequency range between 53.57-78.33 GHz with a radiation density power of 27 x 10(-17) watt/Hz were used. The radiating energy was low enough not to increase the temperature of the cellular samples (cold irradiation). One hour of radiation treatment given every other day to three tumoral human stable cell lines, produced a noticeable inhibition of the cellular growth. The analogous treatment given to two healthy cell lines gave a weak growth stimulation. A scanning electron microscopy study of MCF-7-and K562-irradiated cells revealed that MMW irradiation induced profound morphological changes of the membrane. Finally, we also provided a mechanistic indication, based on millimeter wave spectroscopy of the cells: water is the primary absorber of these electromagnetic waves. Our work provides interesting evidence that wide band low power MMW irradiation, in the appropriate frequency range, could be used in the future as a cold means to cause selective inhibition of tumor cell growth.


Assuntos
Neoplasias da Mama/radioterapia , Campos Eletromagnéticos , Neoplasias da Mama/patologia , Divisão Celular/efeitos da radiação , Humanos , Células K562/patologia , Células K562/efeitos da radiação , Células Tumorais Cultivadas
14.
Int J Biochem Cell Biol ; 46: 56-67, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24275097

RESUMO

The G protein-coupled receptor GPR30/GPER has been shown to mediate rapid effects of 17ß-estradiol (E2) in diverse types of cancer cells. Here, we provide evidence for a novel crosstalk between GPER and the Notch signaling pathway in breast cancer cells and cancer-associated fibroblasts (CAFs). We show that E2 and the GPER selective ligand G-1 induce both the γ-secretase-dependent activation of Notch-1 and the expression of the Notch target gene Hes-1. These inductions are prevented by knocking down GPER or by using a dominant-negative mutant of the Notch transcriptional co-activator Master-mind like-1 (DN-MAML-1), hence suggesting the involvement of GPER in the Notch-dependent transcription. By performing chromatin-immunoprecipitation experiments and luciferase assays, we also demonstrate that E2 and G-1 induce the recruitment of the intracellular domain of Notch-1 (N1ICD) to the Hes-1 promoter and the transactivation of a Hes-1-reporter gene, respectively. Functionally, the E2 and G-1-induced migration of breast cancer cells and CAFs is abolished in presence of the γ-secretase inhibitor GSI or DN-MAML-1, which both inhibit the Notch signaling pathway. In addition, we demonstrate that E2 and G-1 prevent the expression of VE-Cadherin, while both compounds induce the expression of Snail, a Notch target gene acting as a repressor of cadherins expression. Notably, both GSI and DN-MAML-1 abolish the up-regulation of Snail-1 by E2 and G-1, whereas the use of GSI rescues VE-Cadherin expression. Taken together, our results prove the involvement of the Notch signaling pathway in mediating the effects of estrogenic GPER signaling in breast cancer cells and CAFs.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Notch/metabolismo , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Ciclopentanos/farmacologia , Estradiol/farmacologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Quinolinas/farmacologia , Receptor Cross-Talk , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
15.
Cell Signal ; 26(7): 1466-75, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24662263

RESUMO

Nicotinic acid, also known as niacin, is the water soluble vitamin B3 used for decades for the treatment of dyslipidemic diseases. Its action is mainly mediated by the G protein-coupled receptor (GPR) 109A; however, certain regulatory effects on lipid levels occur in a GPR109A-independent manner. The amide form of nicotinic acid, named nicotinamide, acts as a vitamin although neither activates the GPR109A nor exhibits the pharmacological properties of nicotinic acid. In the present study, we demonstrate for the first time that nicotinic acid and nicotinamide bind to and activate the GPER-mediated signalling in breast cancer cells and cancer-associated fibroblasts (CAFs). In particular, we show that both molecules are able to promote the up-regulation of well established GPER target genes through the EGFR/ERK transduction pathway. As a biological counterpart, nicotinic acid and nicotinamide induce proliferative and migratory effects in breast cancer cells and CAFs in a GPER-dependent fashion. Moreover, nicotinic acid prevents the up-regulation of ICAM-1 triggered by the pro-inflammatory cytokine TNF-α and stimulates the formation of endothelial tubes through GPER in HUVECs. Together, our findings concerning the agonist activity for GPER displayed by both nicotinic acid and nicotinamide broaden the mechanisms involved in the biological action of these molecules and further support the potential of a ligand to induce different responses mediated in a promiscuous manner by distinct GPCRs.


Assuntos
Niacina/farmacologia , Niacinamida/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/metabolismo , Neoplasias da Mama , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Modelos Moleculares , Simulação de Acoplamento Molecular , Interferência de RNA , RNA Interferente Pequeno , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Ativação Transcricional , Fator de Necrose Tumoral alfa/metabolismo , Complexo Vitamínico B/farmacologia
16.
Endocr Relat Cancer ; 21(5): 739-53, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25012984

RESUMO

Elevated insulin levels have been associated with an increased cancer risk as well as with aggressive and metastatic cancer phenotypes characterized by a poor prognosis. Insulin stimulates the proliferation, migration, and invasiveness of cancer cells through diverse transduction pathways, including estrogen signaling. As G protein estrogen receptor 1 (GPER1) mediates rapid cell responses to estrogens, we evaluated the potential of insulin to regulate GPER1 expression and function in leiomyosarcoma cancer cells (SKUT-1) and breast cancer-associated fibroblasts (CAFs), which were used as a model system. We found that insulin transactivates the GPER1 promoter sequence and increases the mRNA and protein expression of GPER1 through the activation of the PRKCD/MAPK1/c-Fos/AP1 transduction pathway, as ascertained by means of specific pharmacological inhibitors and gene-silencing experiments. Moreover, cell migration triggered by insulin occurred through GPER1 and its main target gene CTGF, whereas the insulin-induced expression of GPER1 boosted cell-cycle progression and the glucose uptake stimulated by estrogens. Notably, a positive correlation between insulin serum levels and GPER1 expression was found in cancer fibroblasts obtained from breast cancer patients. Altogether, our data indicate that GPER1 may be included among the complex network of transduction signaling triggered by insulin that drives cells toward cancer progression.


Assuntos
Fibroblastos/metabolismo , Insulina/metabolismo , Neoplasias/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Ciclo Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Glucose/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo
17.
Cancer Res ; 74(15): 4053-64, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24894716

RESUMO

Biological responses to estrogens in normal and malignant tissues are mainly mediated by the estrogen receptors ERα and ERß, which function as ligand-activated transcription factors. In addition, the G protein-coupled receptor GPR30 (GPER) mediates estrogenic signaling in breast cancer cells and cancer-associated fibroblasts (CAF) that contribute to cancer progression. In this study, we evaluated the role elicited by GPER in the estrogen-regulated expression and function of vascular endothelial growth factor (VEGF) in ER-negative breast cancer cells and CAF. We demonstrated that 17ß-estradiol (E2) and the GPER-selective ligand G-1 triggered a GPER/EGFR/ERK/c-fos signaling pathway that leads to increased VEGF via upregulation of HIF1α. In further extending the mechanisms involved in E2-supported angiogenesis, we also showed that conditioned medium from CAF treated with E2 and G-1 promoted human endothelial tube formation in a GPER-dependent manner. In vivo, ligand-activated GPER was sufficient to enhance tumor growth and the expression of HIF1α, VEGF, and the endothelial marker CD34 in a mouse xenograft model of breast cancer. Our findings offer important new insights into the ability of estrogenic GPER signaling to trigger HIF1α-dependent VEGF expression that supports angiogenesis and progression in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Estrogênios/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Ciclopentanos/farmacologia , Estradiol/farmacologia , Feminino , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Camundongos , Camundongos Nus , Quinolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/biossíntese
18.
Plast Surg Int ; 2013: 406375, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23346394

RESUMO

Background. Most women with breast cancer today can be managed with breast conservation; however, some women still require mastectomy for treatment of their disease. Skin-sparing mastectomy (SSM) with immediate reconstruction has emerged as a favorable option for many of these patients. The authors combined the SSM technique with the preservation of a small part of the areola with immediate nipple together with with breast reconstruction. Methods. In an 8-year-period 155 female patients (age: 20-52 years old; mean age: 37.5 years) with extensive ductal intraepithelial neoplasia (DIN) or invasive breast cancer were treated with areola skin sparing mastectomy with immediate nipple and breast reconstruction. Patients were followed up prospectively by the breast surgeon, the plastic surgeon, and the oncologist for complications and recurrence. Results. After treatment, only 2 cases (1.29%) had a local recurrence. 8 out of 155 (5.5%) patients developed early complications (infections, seroma, haematoma), and 5 out of 155 patients (3.22%) developed delayed complications (implant rotation, aestethic deterioration) in the post operative time period. The final aesthetic outcome was judged as positive in 150 out of 155 patients (96.78%). Conclusion. In our experience, immediate nipple reconstruction after skin-sparing mastectomy is a technically feasible procedure which can give excellent cosmetic results.

19.
Mol Cell Endocrinol ; 376(1-2): 23-32, 2013 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-23748028

RESUMO

Cancer associated fibroblasts (CAFs) actively contribute to the growth and invasion of cancer cells. In recent years, the G protein estrogen receptor (GPER) has been largely involved in the estrogenic signals in diverse types of normal and tumor cells. In CAFs, GPER was localized into the nucleus, however the molecular mechanisms which regulate its nuclear shuttle remain to be clarified. In the present study, we demonstrate that in breast CAFs GPER translocates into the nucleus through an importin-dependent mechanism. Moreover, we show that a nuclear localization signal is involved in the nuclear import of GPER, in the up-regulation of its target genes c-fos and CTGF and in the migration of CAFs induced by estrogens. Our data provide novel insights into the nuclear localization and function of GPER in CAFs toward a better understanding of the estrogen action elicited through these key players of the tumor microenvironment.


Assuntos
Neoplasias da Mama/metabolismo , Núcleo Celular/metabolismo , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Sinais de Localização Nuclear/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Movimento Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Proliferação de Células , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Estrogênios/farmacologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Sinais de Localização Nuclear/química , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Microambiente Tumoral/genética
20.
Environ Health Perspect ; 120(8): 1177-82, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22552965

RESUMO

BACKGROUND: Bisphenol A (BPA) is the principal constituent of baby bottles, reusable water bottles, metal cans, and plastic food containers. BPA exerts estrogen-like activity by interacting with the classical estrogen receptors (ERα and ERß) and through the G protein-coupled receptor (GPR30/GPER). In this regard, recent studies have shown that GPER was involved in the proliferative effects induced by BPA in both normal and tumor cells. OBJECTIVES: We studied the transduction signaling pathways through which BPA influences cell proliferation and migration in human breast cancer cells and cancer-associated fibroblasts (CAFs). METHODS AND RESULTS: We used as a model system SKBR3 breast cancer cells and CAFs that lack the classical ERs. Specific pharmacological inhibitors and gene-silencing procedures were used to show that BPA induces the expression of the GPER target genes c-FOS, EGR-1, and CTGF through the GPER/EGFR/ERK transduction pathway in SKBR3 breast cancer cells and CAFs. Moreover, we observed that GPER is required for growth effects and migration stimulated by BPA in both cell types. CONCLUSIONS: Results indicate that GPER is involved in the biological action elicited by BPA in breast cancer cells and CAFs. Hence, GPER-mediated signaling should be included among the transduction mechanisms through which BPA may stimulate cancer progression.


Assuntos
Compostos Benzidrílicos/toxicidade , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Fenóis/toxicidade , Receptores de Estrogênio/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Western Blotting , Feminino , Fibroblastos/patologia , Inativação Gênica , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA