Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Curr Pharm Des ; 23(25): 3658-3666, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28641547

RESUMO

BACKGROUND: Insulin resistance is a pathological condition characterized by the failure of target cells to uptake and metabolize glucose in response to insulin. In particular, the elevated concentrations of glucose, insulin and free insulin growth factor-1, which result from insulin resistance, may generate a pro-inflammatory and protumorigenic state. These alterations may underlie the increased risk to develop various types of cancer as well as the worse cancer prognosis observed in obese and diabetic patients. MicroRNAs are short molecules of noncoding endogenous RNA, which are involved in several physio-pathological conditions like glucose homeostasis, lipid metabolism, insulin signaling and resistance. MicroRNAs play also a crucial role in tumorigenesis, acting as oncomirs or tumor suppressors depending on the cell context. METHODS: Here, we recapitulate the role of certain microRNAs that are associated with both insulin resistance and cancer, and discuss their potential to be considered as therapeutic targets. RESULTS: Several studies have highlighted the action of diverse microRNAs in the aforementioned disorders. For instance, three microRNA clusters namely miR-103/107, miR-221/222 and miR-29 have been found to be upregulated in insulin resistance and certain types of cancer. These microRNAs have been shown to target genes like PTEN, Dicer and caveolins that are largely involved in important processes relevant to both insulin resistance and cancer. CONCLUSION: Certain microRNAs may represent potential drug targets common to both insulin resistance and cancer. In particular, the inhibition of miR-103/107, miR-221/222 and miR-29 may be taken into account in novel pharmacological approaches aiming to treat these two disorders.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos/tendências , Resistência à Insulina/fisiologia , MicroRNAs/antagonistas & inibidores , MicroRNAs/fisiologia , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/metabolismo , Humanos , Neoplasias/metabolismo
3.
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.

4.
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
5.
Oncotarget ; 7(33): 52710-52728, 2016 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-27384677

RESUMO

Insulin-like growth factor-I (IGF-I)/IGF-I receptor (IGF-IR) system has been largely involved in the pathogenesis and development of various tumors. We have previously demonstrated that IGF-IR cooperates with the G-protein estrogen receptor (GPER) and the collagen receptor discoidin domain 1 (DDR1) that are implicated in cancer progression. Here, we provide novel evidence regarding the molecular mechanisms through which IGF-I/IGF-IR signaling triggers a functional cross-talk with GPER and DDR1 in both mesothelioma and lung cancer cells. In particular, we show that IGF-I activates the transduction network mediated by IGF-IR leading to the up-regulation of GPER and its main target genes CTGF and EGR1 as well as the induction of DDR1 target genes like MATN-2, FBN-1, NOTCH 1 and HES-1. Of note, certain DDR1-mediated effects upon IGF-I stimulation required both IGF-IR and GPER as determined knocking-down the expression of these receptors. The aforementioned findings were nicely recapitulated in important biological outcomes like IGF-I promoted chemotaxis and migration of both mesothelioma and lung cancer cells. Overall, our data suggest that IGF-I/IGF-IR system triggers stimulatory actions through both GPER and DDR1 in aggressive tumors as mesothelioma and lung tumors. Hence, this novel signaling pathway may represent a further target in setting innovative anticancer strategies.


Assuntos
Receptor com Domínio Discoidina 1/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Receptor Cross-Talk/efeitos dos fármacos , Receptor IGF Tipo 1/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células A549 , Linhagem Celular Tumoral , Receptor com Domínio Discoidina 1/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Mesotelioma/genética , Mesotelioma/metabolismo , Mesotelioma/patologia , Interferência de RNA , Receptor IGF Tipo 1/genética , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Imagem com Lapso de Tempo/métodos
6.
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
7.
AAPS J ; 18(2): 305-10, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26865461

RESUMO

G protein-coupled receptors (GPCRs) are cell surface proteins mainly involved in signal transmission; however, they play a role also in several pathophysiological conditions. Chemically heterogeneous molecules like peptides, hormones, lipids, and neurotransmitters activate second messengers and induce several biological responses by binding to these seven transmembrane receptors, which are coupled to heterotrimeric G proteins. Recently, additional molecular mechanisms have been involved in GPCR-mediated signaling, leading to an intricate network of transduction pathways. In this regard, it should be mentioned that diverse GPCR family members contribute to the adaptive cell responses to low oxygen tension, which is a distinguishing feature of several illnesses like neoplastic and cardiovascular diseases. For instance, the G protein estrogen receptor, namely G protein estrogen receptor (GPER)/GPR30, has been shown to contribute to relevant biological effects induced by hypoxia via the hypoxia-inducible factor (HIF)-1α in diverse cell contexts, including cancer. Likewise, GPER has been found to modulate the biological outcome of hypoxic/ischemic stress in both cardiovascular and central nervous systems. Here, we describe the role exerted by GPCR-mediated signaling in low oxygen conditions, discussing, in particular, the involvement of GPER by a hypoxic microenvironment.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Descoberta de Drogas/métodos , Humanos , Hipóxia/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
8.
Oncotarget ; 7(1): 94-111, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26646587

RESUMO

Aldosterone induces relevant effects binding to the mineralcorticoid receptor (MR), which acts as a ligand-gated transcription factor. Alternate mechanisms can mediate the action of aldosterone such as the activation of epidermal growth factor receptor (EGFR), MAPK/ERK, transcription factors and ion channels. The G-protein estrogen receptor (GPER) has been involved in the stimulatory effects of estrogenic signalling in breast cancer. GPER has been also shown to contribute to certain responses to aldosterone, however the role played by GPER and the molecular mechanisms implicated remain to be fully understood. Here, we evaluated the involvement of GPER in the stimulatory action exerted by aldosterone in breast cancer cells and breast tumor derived endothelial cells (B-TEC). Competition assays, gene expression and silencing studies, immunoblotting and immunofluorescence experiments, cell proliferation and migration were performed in order to provide novel insights into the role of GPER in the aldosterone-activated signalling. Our results demonstrate that aldosterone triggers the EGFR/ERK transduction pathway in a MR- and GPER-dependent manner. Aldosterone does not bind to GPER, it however induces the direct interaction between MR and GPER as well as between GPER and EGFR. Next, we ascertain that the up-regulation of the Na+/H+ exchanger-1 (NHE-1) induced by aldosterone involves MR and GPER. Biologically, both MR and GPER contribute to the proliferation and migration of breast and endothelial cancer cells mediated by NHE-1 upon aldosterone exposure. Our data further extend the current knowledge on the molecular mechanisms through which GPER may contribute to the stimulatory action elicited by aldosterone in breast cancer.


Assuntos
Aldosterona/farmacologia , Células Endoteliais/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Endoteliais/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Immunoblotting , Microscopia de Fluorescência , Ligação Proteica/efeitos dos fármacos , Interferência de RNA , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Imagem com Lapso de Tempo/métodos
9.
Oncotarget ; 6(33): 34158-77, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26415222

RESUMO

Copper promotes tumor angiogenesis, nevertheless the mechanisms involved remain to be fully understood. We have recently demonstrated that the G-protein estrogen receptor (GPER) cooperates with hypoxia inducible factor-1α (HIF-1α) toward the regulation of the pro-angiogenic factor VEGF. Here, we show that copper sulfate (CuSO4) induces the expression of HIF-1α as well as GPER and VEGF in breast and hepatic cancer cells through the activation of the EGFR/ERK/c-fos transduction pathway. Worthy, the copper chelating agent TEPA and the ROS scavenger NAC prevented the aforementioned stimulatory effects. We also ascertained that HIF-1α and GPER are required for the transcriptional activation of VEGF induced by CuSO4. In addition, in human endothelial cells, the conditioned medium from breast cancer cells treated with CuSO4 promoted cell migration and tube formation through HIF-1α and GPER. The present results provide novel insights into the molecular mechanisms involved by copper in triggering angiogenesis and tumor progression. Our data broaden the therapeutic potential of copper chelating agents against tumor angiogenesis and progression.


Assuntos
Sulfato de Cobre/toxicidade , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neovascularização Patológica/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Western Blotting , Linhagem Celular Tumoral , Imunofluorescência , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Transcriptoma
10.
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
11.
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
12.
Artigo em Inglês | MEDLINE | ID: mdl-25798130

RESUMO

The insulin/IGF system plays an important role in cancer progression. Accordingly, elevated levels of circulating insulin have been associated with an increased cancer risk as well as with aggressive and metastatic cancer phenotypes. Numerous studies have documented that estrogens cooperate with the insulin/IGF system in multiple pathophysiological conditions. The biological responses to estrogens are mainly mediated by the estrogen receptors (ER)α and ERß, which act as transcription factors; however, several studies have recently demonstrated that a member of the G protein-coupled receptors, named GPR30/G-protein estrogen receptor (GPER), is also involved in the estrogen signaling in normal and malignant cells as well as in cancer-associated fibroblasts (CAFs). In this regard, novel mechanisms linking the action of estrogens through GPER with the insulin/IGF system have been recently demonstrated. This review recapitulates the relevant aspects of this functional cross-talk between the insulin/IGF and the estrogenic GPER transduction pathways, which occurs in various cell types and may account for cancer progression.

13.
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
14.
J Steroid Biochem Mol Biol ; 137: 50-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23542661

RESUMO

G protein-coupled receptors (GPCRs) and growth factor receptors mediate multiple physio-pathological responses to a diverse array of extracellular stimuli. In this regard, it has been largely demonstrated that GPCRs and growth factor receptors generate a multifaceted signaling network, which triggers relevant biological effects in normal and cancer cells. For instance, some GPCRs transactivate the epidermal growth factor receptor (EGFR), which stimulates diverse transduction pathways leading to gene expression changes, cell migration, survival and proliferation. Moreover, it has been reported that a functional interaction between growth factor receptors and steroid hormones like estrogens is involved in the growth of many types of tumors as well as in the resistance to endocrine therapy. This review highlights recent findings on the cross-talk between a member of the GPCR family, the G protein-coupled estrogen receptor 1 (GPER, formerly known as GPR30) and two main growth factor receptors like EGFR and insulin-like growth factor-I receptor (IGF-IR). The biological implications of the functional interaction between these important mediators of cell responses particularly in cancer are discussed. This article is part of a Special Issue entitled 'CSR 2013'.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Receptor Cross-Talk , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Humanos , Ligantes , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores
15.
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
16.
Cell Signal ; 24(8): 1515-21, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22481093

RESUMO

There is increasing awareness that estrogens may affect cell functions through the integration with a network of signaling pathways. The IGF system is a phylogenetically highly conserved axis that includes the insulin receptor (IR) and the insulin-like growth factor I receptor (IGF-IR) pathways, which are of crucial importance in the regulation of metabolism and cell growth in relationship to nutrient availability. Numerous studies nowadays document that estrogens cooperate with IGF system at multiple levels both in physiology and in disease. Several studies have focused on this bidirectional cross-talk in central nervous system, in mammary gland development and in cancer. Notably, cancer cells show frequent deregulation of the IGF system with overexpression of IR and/or IGF-IR and their ligands as well as frequent upregulation of the classical estrogen receptor (ER)α and the novel ER named GPER. Recent studies have, therefore, unraveled further mechanisms of cross-talk involving membrane initiated estrogen actions and the IGF system in cancer, that converge in the stimulation of pro-tumoral effects. These studies offer hope for new strategies aimed at the treatment of estrogen related cancers in order to prevent an estrogen-independent and more aggressive tumor progression.


Assuntos
Estrogênios/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias/metabolismo , Transdução de Sinais , Animais , Humanos , Fator de Crescimento Insulin-Like I/genética , Neoplasias/patologia , Receptor IGF Tipo 1/metabolismo
17.
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
18.
Mol Cell Endocrinol ; 320(1-2): 162-70, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20138962

RESUMO

Estrogens are structurally related steroids that regulate important physiological processes. 17beta-estradiol (E2) is reversibly oxidized to estrone (E1) and both E2 and E1 can be irreversibly converted to estriol (E3), which also originates directly from androstenedione. The action of E2 has been traditionally explained by the binding to the estrogen receptor (ER) alpha and ER beta, however the G protein-coupled receptor (GPR) 30 has been recently involved in the rapid signaling triggered by estrogens. Although the role of E2 in the development of breast cancer has been largely documented, the contribution of E3 still remains to be completely evaluated. Here, we demonstrate for the first time that E3 acts as a GPR30 antagonist since it was able to inhibit the GPR30-mediated responses such as the rapid ERK activation, the up-regulation of target genes like c-fos and connective tissue growth factor, the proliferative effects observed in ER-negative SkBr3 cells.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Estriol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Sítios de Ligação , Ligação Competitiva/efeitos dos fármacos , Bioensaio , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Simulação por Computador , Fator de Crescimento do Tecido Conjuntivo/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Estrogênio
19.
Mol Endocrinol ; 23(11): 1815-26, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19749156

RESUMO

In the present study, we evaluated the regulation of G protein-coupled receptor (GPR)30 expression in estrogen receptor (ER)-positive endometrial, ovarian, and estrogen-sensitive, as well as tamoxifen-resistant breast cancer cells. We demonstrate that epidermal growth factor (EGF) and TGF alpha transactivate the GPR30 promoter and accordingly up-regulate GPR30 mRNA and protein levels only in endometrial and tamoxifen-resistant breast cancer cells. These effects exerted by EGF and TGF alpha were dependent on EGF receptor (EGFR) expression and activation and involved phosphorylation of the Tyr(1045) and Tyr(1173) EGFR sites. Using gene-silencing experiments and specific pharmacological inhibitors, we have ascertained that EGF and TGF alpha induce GPR30 expression through the EGFR/ERK transduction pathway, and the recruitment of c-fos to the activator protein-1 site located within GPR30 promoter sequence. Interestingly, we show that functional cross talk of GPR30 with both activated EGFR and ER alpha relies on a physical interaction among these receptors, further extending the potential of estrogen to trigger a complex stimulatory signaling network in hormone-sensitive tumors. Given that EGFR/HER2 overexpression is associated with tamoxifen resistance, our data may suggest that ligand-activated EGFR could contribute to the failure of tamoxifen therapy also by up-regulating GPR30, which in turn could facilitates the action of estrogen. In addition, important for resistance is the ability of tamoxifen to bind to and activate GPR30, the expression of which is up-regulated by EGFR activation. Our results emphasize the need for new endocrine agents able to block widespread actions of estrogen without exerting any stimulatory activity on transduction pathways shared by the steroid and growth factor-signaling networks.


Assuntos
Receptor alfa de Estrogênio/biossíntese , Regulação Neoplásica da Expressão Gênica , Receptores Acoplados a Proteínas G/biossíntese , Fator de Crescimento Transformador alfa/biossíntese , Regulação para Cima , Linhagem Celular Tumoral , Humanos , Modelos Biológicos , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Estrogênio , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , Tirosina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...