Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 582
Filtrar
1.
J Pharmacol Exp Ther ; 351(1): 172-80, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25077525

RESUMO

Inflammation has been implicated in tumor initiation, angiogenesis, and metastasis, and linked to the development of more aggressive, therapy-resistant estrogen receptor (ER)-positive breast cancer. Resolvin D2 (RvD2) is a potent anti-inflammatory lipid mediator. As RvD2 may be synthesized within breast tumors by both tumor cells and the surrounding stroma cells and is present in plasma at bioactive concentrations, we sought to characterize the impact of RvD2 on cell processes underlying breast tumor growth and spread. Trypan-blue exclusion, transfection with estrogen response element (ERE) reporter, real-time quantitative polymerase chain reaction, competitive radioligand binding assays, Western blotting, and immunofluorescence were the techniques used. Unexpectedly, whereas RvD2 (10-1000 nM) supported the proliferation of the ER-positive breast tumor (MCF-7) cells, it did not affect the ER-negative MDA-MB-231 cell number. The proliferative effect of RvD2 in MCF-7 cells was attenuated by the ER antagonist ICI 182,780 (7α-[9-[(4,4,5,5,5-pentafluoropentyl)sulfinyl]nonyl]estra-1,3,5(10)-triene-3,17ß-diol). Furthermore, RvD2 increased ERE transcriptional activity in a number of ER-positive breast and ovarian tumor cell lines. This activation was also inhibited by ICI 182,780. RvD2 altered the expression of a subset of estrogen-responsive genes. Although binding experiments showed that RvD2 did not directly compete with [(3)H]17ß-estradiol for ER binding, prior exposure of MCF-7 cells to RvD2 resulted in a significant reduction in the apparent cytosolic ER density. Confocal immunocytochemistry and Western blotting studies showed that RvD2 promoted nuclear localization of ERα. These observations indicate that RvD2 displays significant but indirect estrogenic properties and has the potential to play a role in estrogen-dependent breast cancer progression.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Receptores de Estrogênio/metabolismo , Ácidos Docosa-Hexaenoicos/farmacocinética , Estradiol/análogos & derivados , Estradiol/farmacologia , Fulvestranto , Humanos , Células MCF-7 , Ligação Proteica , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores
2.
Environ Health ; 13(1): 41, 2014 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-24886603

RESUMO

BACKGROUND: Xenobiotic chemicals with estrogenic activity (EA), such as bisphenol A (BPA), have been reported to have potential adverse health effects in mammals, including humans, especially in fetal and infant stages. Concerns about safety have caused many manufacturers to use alternatives to polycarbonate (PC) resins to make hard and clear, reusable, plastic products that do not leach BPA. However, no study has focused on whether such BPA-free PC-replacement products, chosen for their perceived higher safety, especially for babies, also release other chemicals that have EA. METHODS: We used two, well-established, mammalian cell-based, assays (MCF-7 and BG1Luc) to assess the EA of chemicals that leached into over 1000 saline or ethanol extracts of 50 unstressed or stressed (autoclaving, microwaving, and UV radiation) BPA-free PC-replacement products. An EA antagonist, ICI 182,780, was used to confirm that agonist activity in leachates was due to chemicals that activated the mammalian estrogen receptor. RESULTS: Many unstressed and stressed, PC-replacement-products made from acrylic, polystyrene, polyethersulfone, and Tritan™ resins leached chemicals with EA, including products made for use by babies. Exposure to various forms of UV radiation often increased the leaching of chemicals with EA. In contrast, some BPA-free PC-replacement products made from glycol-modified polyethylene terephthalate or cyclic olefin polymer or co-polymer resins did not release chemicals with detectable EA under any conditions tested. CONCLUSIONS: This hazard assessment survey showed that many BPA-free PC- replacement products still leached chemicals having significant levels of EA, as did BPA-containing PC counterparts they were meant to replace. That is, BPA-free did not mean EA-free. However, this study also showed that some PC-replacement products did not leach chemicals having significant levels of EA. That is, EA-free PC-replacement products could be made in commercial quantities at prices that compete with PC-replacement products that were not BPA-free. Since plastic products often have advantages (price, weight, shatter-resistance, etc.) compared to other materials such as steel or glass, it is not necessary to forgo those advantages to avoid release into foodstuffs or the environment of chemicals having EA that may have potential adverse effects on our health or the health of future generations.


Assuntos
Estrogênios/análise , Plásticos/química , Compostos Benzidrílicos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Estrogênios/química , Estrogênios/farmacologia , Etanol/química , Temperatura Alta , Humanos , Luciferases/metabolismo , Células MCF-7 , Micro-Ondas , Fenóis , Plásticos/efeitos da radiação , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/metabolismo , Cloreto de Sódio/química , Raios Ultravioleta
4.
Nat Med ; 20(4): 419-24, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24658075

RESUMO

In response to microbial infection, expression of the defensin-like peptide hepcidin (encoded by Hamp) is induced in hepatocytes to decrease iron release from macrophages. To elucidate the mechanism by which Salmonella enterica var. Typhimurium (S. typhimurium), an intramacrophage bacterium, alters host iron metabolism for its own survival, we examined the role of nuclear receptor family members belonging to the NR3B subfamily in mouse hepatocytes. Here, we report that estrogen-related receptor γ (ERRγ, encoded by Esrrg) modulates the intramacrophage proliferation of S. typhimurium by altering host iron homeostasis, and we demonstrate an antimicrobial effect of an ERRγ inverse agonist. Hepatic ERRγ expression was induced by S. typhimurium-stimulated interleukin-6 signaling, resulting in an induction of hepcidin and eventual hypoferremia in mice. Conversely, ablation of ERRγ mRNA expression in liver attenuated the S. typhimurium-mediated induction of hepcidin and normalized the hypoferremia caused by S. typhimurium infection. An inverse agonist of ERRγ ameliorated S. typhimurium-mediated hypoferremia through reduction of ERRγ-mediated hepcidin mRNA expression and exerted a potent antimicrobial effect on the S. typhimurium infection, thereby improving host survival. Taken together, these findings suggest an alternative approach to control multidrug-resistant intracellular bacteria by modulating host iron homeostasis.


Assuntos
Hepatócitos/metabolismo , Hepcidinas/metabolismo , Interleucina-6/imunologia , Ferro/metabolismo , Macrófagos/metabolismo , Receptores de Estrogênio/metabolismo , Infecções por Salmonella/metabolismo , Salmonella typhimurium , Transdução de Sinais/imunologia , Animais , Proteínas de Transporte de Cátions/metabolismo , Hepatócitos/imunologia , Hepcidinas/imunologia , Homeostase , Fígado/metabolismo , Macrófagos/imunologia , Camundongos , Receptores de Estrogênio/agonistas , Infecções por Salmonella/imunologia , Transdução de Sinais/fisiologia
5.
J Cancer Res Clin Oncol ; 140(5): 713-23, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24553912

RESUMO

BACKGROUND: The orphan, membrane-bound estrogen receptor (GPER) is expressed at high levels in a large fraction of breast cancer patients and its expression is favorable for patients' survival. METHODS: We investigated the role of GPER as a potential tumor suppressor in triple-negative breast cancer cells MDA-MB-231 and MDA-MB-468 using cell cycle analysis and apoptosis assay. The constitutive activity of GPER was investigated. RESULTS: GPER-specific activation with G-1 agonist inhibited breast cancer cell growth in concentration-dependent manner via induction of the cell cycle arrest in G2/M phase, enhanced phosphorylation of histone H3 and caspase-3-mediated apoptosis. Analysis of the methylation status of the GPER promoter in the triple-negative breast cancer cells and in tissues derived from breast cancer patients revealed that GPER amount is regulated by epigenetic mechanisms and GPER expression is inactivated by promoter methylation. Furthermore, GPER expression was induced by stress factors, such as radiation, and GPER amount inversely correlated with the p53 expression level. CONCLUSIONS: Overall, our results establish the protective role in breast cancer tumorigenesis, and the cell surface expression of GPER makes it an excellent potential therapeutic target for triple-negative breast cancer.


Assuntos
Carcinogênese/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/genética , Neoplasias de Mama Triplo Negativas/genética , Apoptose/efeitos dos fármacos , Caspase 3/genética , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclopentanos/administração & dosagem , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Quinolinas/administração & dosagem , RNA Interferente Pequeno , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/patologia
6.
J Cancer Res Clin Oncol ; 140(4): 663-71, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24515910

RESUMO

PURPOSE: The orphan, membrane-bound estrogen receptor (GPER) is expressed at high levels in a large fraction of breast cancer patients, and its expression is favorable for patients' survival. We investigated the role of GPER as a potential tumor suppressor in MCF-7 and SK-BR-3 breast cancer cells. METHODS: The effect of GPER agonist G-1 in cell culture was used to determine whether GPER inhibit cell growth. The methylation status of GPER promoter was investigated by methylation-specific PCR. RESULTS: GPER-specific agonist G-1 inhibited breast cancer cell proliferation in concentration-dependent manner via induction of the cell cycle arrest in M-phase, enhanced phosphorylation of histone 3 and cell apoptosis. Analysis of the methylation status of the GPER promoter in MCF-7 and SK-BR-3 cells revealed that GPER expression is regulated by epigenetic mechanisms and GPER expression is inactivated by promoter methylation. Overall, our results are consistent with our recent findings in triple-negative breast cancer cells, and the cell surface expression of GPER makes it an excellent potential therapeutic target for non-triple-negative breast cancer.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Ciclopentanos/farmacologia , Metilação de DNA/efeitos dos fármacos , Feminino , Humanos , Técnicas Imunoenzimáticas , Regiões Promotoras Genéticas/genética , Quinolinas/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/genética , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
7.
Bioorg Med Chem Lett ; 24(5): 1403-6, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24507928

RESUMO

A novel biological activity of psoralidin as an agonist for both estrogen receptor (ER)α and ERß agonist has been demonstrated in our study. Psoralidin has been characterized as a full ER agonist, which activates the classical ER-signaling pathway in both ER-positive human breast and endometrial cell lines as well as non-human cultured cells transiently expressing either ERα or ERß. The estrogenic activity was determined using the relative expression levels of either reporter or the endogenous genes dependent on the agonist-bound ER to the estrogen response element (ERE). Psoralidin at 10 µM was able to induce the maximum reporter gene expression corresponding to that of E2-treated cells and such activation of the ERE-reporter gene by psoralidin was completely abolished by the cotreatment of a pure ER antagonist, implying that the biological activities of psoralidin are mediated by ER. Psoralidin was also able to induce the endogenous estrogen-responsive gene, pS2, in human breast cancer cells MCF-7. It was observed that activation of the classical ER-signaling pathway by psoralidin is mediated via induction of ER conformation by psoralidin and direct binding of the psoralidin-ER complex to the EREs present in the promoter region of estrogen-responsive genes, as shown by chromatin immunoprecipitation assay results. Finally, molecular docking of psoralidin to the ligand binding pocket of the ERα showed that psoralidin is able to mimic the binding interactions of E2, and thus, it could act as an ER agonist in the cellular environment.


Assuntos
Benzofuranos/química , Cumarínicos/química , Psoralea/química , Receptores de Estrogênio/metabolismo , Benzofuranos/isolamento & purificação , Benzofuranos/farmacologia , Sítios de Ligação , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cumarínicos/isolamento & purificação , Cumarínicos/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Psoralea/metabolismo , Receptores de Estrogênio/agonistas , Transdução de Sinais/efeitos dos fármacos
8.
Toxicol Lett ; 226(1): 6-13, 2014 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-24487124

RESUMO

The estrogen receptor (ER) and aryl hydrocarbon receptor (AhR) are ligand-activated transcription factors involved in estrogen or xenobiotic exposure, whereas the 90-kDa heat shock protein (HSP90), which is a ubiquitously expressed molecular chaperone, is involved in the signal transduction process. Although the interactions between these pathways have been under investigation, the mechanisms are unclear and the potential role of HSP90 in these interactions has not been reported. The results of goldfish primary hepatocytes showed that exposure to PCB77 and 17ß-estradiol (E2) alone induced significant protein expression of cytochrome P450 1A (CYP1A) and vitellogenin (VTG), respectively. On the other hand, the combined exposure to PCB77 and E2 led to the reduction of CYP1A and VTG compared to the single treatments. Although the AhRs and ERs were naturally induced during the co-treatment, the total amount of HSP90 binding to the receptors was not changed. Furthermore, while the HSP90 chaperon activity was blocked by the specific inhibitor (geldanamycin), reciprocal inhibition between AhR and ER pathways was not observed. These findings indicate a potential role of HSP90 where competition between AhR and ER for binding to HSP90 can occur and cause reciprocal inhibition.


Assuntos
Proteínas de Peixes/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Hepatócitos/metabolismo , Receptor Cross-Talk , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Animais , Ligação Competitiva , Cloreto de Cádmio/toxicidade , Células Cultivadas , Citocromo P-450 CYP1A1/biossíntese , Relação Dose-Resposta a Droga , Indução Enzimática , Estradiol/toxicidade , Proteínas de Peixes/agonistas , Carpa Dourada , Ligantes , Masculino , Bifenilos Policlorados/toxicidade , Receptor Cross-Talk/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Estrogênio/agonistas , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima , Vitelogeninas/metabolismo , Poluentes Químicos da Água/toxicidade
9.
Endocr Relat Cancer ; 21(2): 355-69, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24481325

RESUMO

Cancer-associated fibroblasts (CAFs) are crucial co-mediators of breast cancer progression. Estrogen is the predominant driving force in the cyclic regulation of the mammary extracellular matrix, thus potentially affecting the tumor-associated stroma. Recently, a third estrogen receptor, estrogen (G-protein-coupled) receptor (GPER), has been reported to be expressed in breast CAFs. In this study, GPER was detected by immunohistochemical analysis in stromal fibroblasts of 41.8% (59/141) of the primary breast cancer samples. GPER expression in CAFs isolated from primary breast cancer tissues was confirmed by immunostaining and RT-PCR analyses. Tamoxifen (TAM) in addition to 17ß-estradiol (E2) and the GPER agonist G1 activated GPER, resulting in transient increases in cell index, intracellular calcium, and ERK1/2 phosphorylation. Furthermore, TAM, E2, and G1 promoted CAF proliferation and cell-cycle progression, both of which were blocked by GPER interference, the selective GPER antagonist G15, the epidermal growth factor receptor (EGFR) inhibitor AG1478, and the ERK1/2 inhibitor U0126. Importantly, TAM as well as G1 increased E2 production in breast CAFs via GPER/EGFR/ERK signaling when the substrate of E2, testosterone, was added to the medium. GPER-induced aromatase upregulation was probably responsible for this phenomenon, as TAM- and G1-induced CYP19A1 gene expression was reduced by GPER knockdown and G15, AG1478, and U0126 administration. Accordingly, GPER-mediated CAF-dependent estrogenic effects on the tumor-associated stroma are conceivable, and CAF is likely to contribute to breast cancer progression, especially TAM resistance, via a positive feedback loop involving GPER/EGFR/ERK signaling and E2 production.


Assuntos
Neoplasias da Mama/metabolismo , Proliferação de Células , Fibroblastos/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ciclopentanos/farmacologia , Estradiol/farmacologia , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Quinolinas/farmacologia , Receptores de Estrogênio/agonistas , Receptores Acoplados a Proteínas G/agonistas , Tamoxifeno/farmacologia
10.
Vitam Horm ; 94: 229-51, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24388193

RESUMO

Bisphenol-A (BPA) is one of the highest-volume chemicals produced worldwide and the widespread exposure of individuals to BPA is suspected to affect a variety of physiological functions, including reproduction, development, and metabolism. Its estrogenic activity has been well documented in the last 15 years. In addition to estrogen receptors, BPA has been also shown to bind to and activate the estrogen-related receptor γ and pregnane X receptor and inhibit the androgen receptor. Halogenated BPAs were also shown to activate the peroxisome proliferator-activated receptor γ and inhibit thyroid hormone receptors. In this chapter, we review recent studies shedding light on the structural and molecular mechanisms by which BPA and its halogenated derivatives interfere with nuclear hormone receptor signaling. These data provide guidelines for the development of safer substitutes devoid of hormonal activity and may help environmental risk assessment.


Assuntos
Antagonistas de Androgênios/toxicidade , Compostos Benzidrílicos/toxicidade , Poluentes Ambientais/toxicidade , Estrogênios não Esteroides/toxicidade , PPAR gama/agonistas , Fenóis/toxicidade , Receptores de Esteroides/agonistas , Receptores dos Hormônios Tireóideos/antagonistas & inibidores , Antagonistas de Androgênios/química , Animais , Compostos Benzidrílicos/química , Disruptores Endócrinos/química , Disruptores Endócrinos/toxicidade , Poluentes Ambientais/química , Estrogênios não Esteroides/química , Halogenação , Humanos , PPAR gama/metabolismo , Fenóis/química , Receptor de Pregnano X , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/metabolismo , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo
11.
Toxicol Sci ; 137(2): 335-49, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24213142

RESUMO

Endocrine-disrupting chemicals with estrogenic activity (EA) or anti-EA (AEA) have been extensively reported to possibly have many adverse health effects. We have developed robotized assays using MCF-7:WS8 cell proliferation (or suppression) to detect EA (or AEA) of 78 test substances supplied by the Interagency Coordinating Committee on the Validation of Alternative Methods and the National Toxicology Program's Interagency Center for the Evaluation of Alternative Toxicological Methods for validation studies. We also assayed ICI 182,780, a strong estrogen antagonist. Chemicals to be assayed were initially examined for solubility and volatility to determine optimal assay conditions. For both EA and AEA determinations, a Range-Finder assay was conducted to determine the concentration range for testing, followed by a Comprehensive assay. Test substances with potentially positive results from an EA Comprehensive assay were subjected to an EA Confirmation assay that evaluated the ability of ICI 182,780 to reverse chemically induced MCF-7 cell proliferation. The AEA assays examined the ability of chemicals to decrease MCF-7 cell proliferation induced by nonsaturating concentrations of 17ß-estradiol (E2), relative to ICI or raloxifene, also a strong estrogen antagonist. To be classified as having AEA, a saturating concentration of E2 had to significantly reverse the decrease in cell proliferation produced by the test substance in nonsaturating E2. We conclude that our robotized MCF-7 EA and AEA assays have accuracy, sensitivity, and specificity values at least equivalent to validated test methods accepted by the U.S. Environmental Protection Agency and the Organisation for Economic Co-operation and Development.


Assuntos
Bioensaio/métodos , Proliferação de Células/efeitos dos fármacos , Disruptores Endócrinos/farmacologia , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Estrogênios/agonistas , Alternativas aos Testes com Animais , Bioensaio/instrumentação , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Disruptores Endócrinos/química , Estradiol/química , Estradiol/farmacologia , Antagonistas de Estrogênios/química , Fulvestranto , Humanos , Células MCF-7 , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Robótica
12.
Environ Int ; 63: 216-23, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24317228

RESUMO

Polybrominated diphenyl ethers are compounds widely used as flame-retardants, which are of increasing environmental concern due to their persistence, and potential adverse effects. This study had two objectives. First, we assessed if BDE-209 in sediment was bioavailable and bioaccumulated into zebrafish embryos. Secondly, we assessed the potential impact on human and environmental health of bioavailable BDE-209 using human in vitro cell assays and zebrafish embryos. Zebrafish were exposed from 4h to 8days post-fertilization to sediments spiked with 12.5mg/kg of BDE-209. Zebrafish larvae accumulated ten fold more BDE-209 than controls in unspiked sediment after 8days. BDE-209 impacted expression of neurological pathways and altered behavior of larvae, although BDE-209 had no visible affect on thyroid function or motoneuron and neuromast development. Zebrafish data and in silico predictions suggested that BDE-209 would also interact with key human transcription factors and receptors. We therefore tested these predictions using mammalian in vitro assays. BDE-209 activated human aryl hydrocarbon receptor, peroxisome proliferator activating receptors, CF/b-cat, activator protein 1, Oct-MLP, and the estrogen receptor-related alpha (ERRα) receptor in cell-based assays. BDE-209 also inhibited human acetylcholinesterase activity. The observation that BDE-209 can be bioaccumulated from contaminated sediment highlights the need to consider this as a potential environmental exposure route. Once accumulated, our data also show that BDE-209 has the potential to cause impacts on both human and environmental health.


Assuntos
Retardadores de Chama/toxicidade , Sedimentos Geológicos , Éteres Difenil Halogenados/toxicidade , Poluentes Químicos da Água/toxicidade , Poluição Química da Água , Peixe-Zebra/embriologia , Acetilcolinesterase/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/agonistas , Inibidores da Colinesterase/toxicidade , Simulação por Computador , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Estrogênio/agonistas , Medição de Risco , Receptor ERRalfa Relacionado ao Estrogênio
13.
Neurotox Res ; 25(3): 271-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23943137

RESUMO

A low dose of the organophosphorus insecticide dimethoate (DMT) produces oxidation of lipids and proteins and impairs mitochondrial function in the brain of male rats, together with a reduction of gonadal hormones in plasma. Here, we have assessed whether DMT affected the expression of inflammatory molecules, the production of reactive oxygen species (ROS), and the expression of steroidogenic proteins and estrogen receptors in cortical astrocyte-enriched cultures obtained separately from male and female CD1 mice pups. Furthermore, we have analyzed whether estradiol may counteract the effects of DMT. A dose of DMT (2 µg/mL) did not affect cell viability, increased interleukin (IL) 6, IL1ß, tumor necrosis factor (TNF)α, interferon-γ-inducible protein 10 (IP10), ERß, steroidogenic acute regulatory protein, and aromatase mRNA levels and ERα protein levels in male but not in female cultures. Estradiol decreased the mRNA levels of IL6, IP10, TNFα, and IL1ß in male but not in female cultures treated with DMT. The effect of estradiol was prevented by the ER antagonist ICI 182,780, fully imitated by an ERß agonist and partially imitated by an ERα agonist. Furthermore, DMT increased the production of ROS in male astrocytes while estradiol reduced ROS production to control levels. These findings indicate that a sublethal dose of DMT alters astrocyte function. The selective action of estradiol on male astrocytes and the sexually dimorphic action of DMT suggest that the pesticide may have different neurological outcomes in males and females.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Dimetoato/toxicidade , Estradiol/farmacologia , Estrogênios/farmacologia , Inseticidas/toxicidade , Animais , Astrócitos/imunologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/imunologia , Córtex Cerebral/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo , Fatores Sexuais
14.
Neuroscience ; 256: 322-33, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24157932

RESUMO

Protection of motoneurons is an important goal in the treatment of spinal cord injury (SCI). We tested whether neuroprotective microRNAs (miRs) like miR-206, miR-17, miR-21, miR-7-1, and miR-106a could enhance efficacy of estrogen receptor (ER) agonists such as 1,3,5-tris (4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT, ERα agonist), Way200070 (WAY, ERß agonist), and estrogen (EST, ERα and ERß agonist) in preventing apoptosis in the calcium ionophore (CI)-insulted ventral spinal cord 4.1 (VSC4.1) motoneurons. We determined that 200 nM CI induced 70% cell death. Treatment with 50 nM PPT, 100 nM WAY, and 150 nM EST induced overexpression of ERα, ERß, and both receptors, respectively, at mRNA and protein levels. Treatment with ER agonists significantly upregulated miR-206, miR-17, and miR-7-1 in the CI-insulted VSC4.1 motoneurons. Transfection with miR-206, miR-17, or miR-7-1 mimic potentiated WAY or EST to inhibit apoptosis in the CI-insulted VSC4.1 motoneurons. Overexpression of miR-7-1 maximally increased efficacy of WAY and EST for down regulation of pro-apoptotic Bax and upregulation of anti-apoptotic Bcl-2. A search using microRNA database (miRDB) indicated that miR-7-1 could inhibit the expression of L-type Ca(2+) channel protein alpha 1C (CPα1C). miR-7-1 overexpression and WAY or EST treatment down regulated CPα1C but upregulated p-Akt to trigger cell survival signaling. The same therapeutic strategy increased expression of the Ca(2+)/calmodulin-dependent protein kinase II beta (CaMKIIß) and the phosphorylated cAMP response element binding protein (p-CREB) so as to promote Bcl-2 transcription. Whole cell membrane potential and mitochondrial membrane potential studies indicated that miR-7-1 highly potentiated EST to preserve functionality in the CI-insulted VSC4.1 motoneurons. In conclusion, our data indicated that miR-7-1 most significantly potentiated efficacy of EST for functional neuroprotection and this therapeutic strategy could be used in the future to attenuate apoptosis of motoneurons in SCI.


Assuntos
MicroRNAs/farmacologia , Neurônios Motores/efeitos dos fármacos , Neuroprostanos/farmacologia , Receptores de Estrogênio/agonistas , Medula Espinal/citologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Cloretos/farmacologia , Relação Dose-Resposta a Droga , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Embrião de Mamíferos , Estrogênios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ginsenosídeos/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Oxazóis/farmacologia , Fenóis/farmacologia , Ratos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Sapogeninas/farmacologia
15.
J Steroid Biochem Mol Biol ; 142: 142-54, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24373794

RESUMO

Selective estrogen receptor modulators (SERMs) are synthetic non-steroidal agents which have varying estrogen agonist and antagonist activities in different tissues, most likely due to the receptor conformation changes associated with that SERM's binding and the subsequent effect on transcription. Clinical trials aim to differentiate amongst SERMs on selected target tissues for use in postmenopausal women including effects on breast, bone, cardiovascular venous thrombosis risk, endometrium, vagina, vasomotor symptoms, and brain. This paper describes differences in clinical effects on selected target tissues of SERMs that are approved, discontinued or in development. FDA approved SERMs include tamoxifen and toremifene used for prevention and treatment of breast cancer, raloxifene approved for prevention and treatment of osteoporosis and prevention of invasive breast cancer, and ospemifene approved for treatment of dyspareunia from menopausal vaginal atrophy. The FDA approved first tissue selective estrogen complex (TSEC) a pairing of conjugated equine estrogens with the SERM, bazedoxifene. This pairing reduces the risk of endometrial hyperplasia that can occur with the estrogenic component of the TSEC without the need for a progestogen in women with a uterus. It also allows for the estrogenic benefits on relief of hot flashes and prevention of bone loss without stimulating the breast or the endometrium. In clinical practice, the tissue-selective actions of SERMs, alone or paired with estrogens, allow for individualization in meeting the treatment needs of postmenopausal women by providing targeted tissue effects. This article is part of a Special Issue entitled 'Menopause'.


Assuntos
Pós-Menopausa , Receptores de Estrogênio/agonistas , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Ensaios Clínicos como Assunto , Dispareunia/tratamento farmacológico , Estrogênios Conjugados (USP)/uso terapêutico , Feminino , Fogachos/tratamento farmacológico , Humanos , Indóis/uso terapêutico , Osteoporose/tratamento farmacológico , Osteoporose Pós-Menopausa/tratamento farmacológico , Osteoporose Pós-Menopausa/prevenção & controle , Pós-Menopausa/efeitos dos fármacos , Pirrolidinas/uso terapêutico , Cloridrato de Raloxifeno/uso terapêutico , Receptores de Estrogênio/antagonistas & inibidores , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Tamoxifeno/análogos & derivados , Tamoxifeno/uso terapêutico , Tetra-Hidronaftalenos/uso terapêutico , Toremifeno/uso terapêutico
16.
J Chem Inf Model ; 53(12): 3244-61, 2013 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-24279462

RESUMO

There are thousands of environmental chemicals subject to regulatory decisions for endocrine disrupting potential. The ToxCast and Tox21 programs have tested ∼8200 chemicals in a broad screening panel of in vitro high-throughput screening (HTS) assays for estrogen receptor (ER) agonist and antagonist activity. The present work uses this large data set to develop in silico quantitative structure-activity relationship (QSAR) models using machine learning (ML) methods and a novel approach to manage the imbalanced data distribution. Training compounds from the ToxCast project were categorized as active or inactive (binding or nonbinding) classes based on a composite ER Interaction Score derived from a collection of 13 ER in vitro assays. A total of 1537 chemicals from ToxCast were used to derive and optimize the binary classification models while 5073 additional chemicals from the Tox21 project, evaluated in 2 of the 13 in vitro assays, were used to externally validate the model performance. In order to handle the imbalanced distribution of active and inactive chemicals, we developed a cluster-selection strategy to minimize information loss and increase predictive performance and compared this strategy to three currently popular techniques: cost-sensitive learning, oversampling of the minority class, and undersampling of the majority class. QSAR classification models were built to relate the molecular structures of chemicals to their ER activities using linear discriminant analysis (LDA), classification and regression trees (CART), and support vector machines (SVM) with 51 molecular descriptors from QikProp and 4328 bits of structural fingerprints as explanatory variables. A random forest (RF) feature selection method was employed to extract the structural features most relevant to the ER activity. The best model was obtained using SVM in combination with a subset of descriptors identified from a large set via the RF algorithm, which recognized the active and inactive compounds at the accuracies of 76.1% and 82.8% with a total accuracy of 81.6% on the internal test set and 70.8% on the external test set. These results demonstrate that a combination of high-quality experimental data and ML methods can lead to robust models that achieve excellent predictive accuracy, which are potentially useful for facilitating the virtual screening of chemicals for environmental risk assessment.


Assuntos
Algoritmos , Inteligência Artificial , Disruptores Endócrinos/classificação , Receptores de Estrogênio/metabolismo , Poluentes Químicos da Água/classificação , Análise Discriminante , Disruptores Endócrinos/farmacologia , Monitoramento Ambiental , Ensaios de Triagem em Larga Escala , Humanos , Relação Quantitativa Estrutura-Atividade , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Medição de Risco , Poluentes Químicos da Água/farmacologia
17.
Cell Death Dis ; 4: e869, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24136233

RESUMO

The G-protein-coupled estrogen receptor 1 (GPER) has recently been reported to mediate the non-genomic action of estrogen in different types of cells and tissues. G-1 (1-[4-(6-bromobenzo[1,3] dioxol-5yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone) was developed as a potent and selective agonist for GPER. G-1 has been shown to induce the expression of genes and activate pathways that facilitate cancer cell proliferation by activating GPER. Here we demonstrate that G-1 has an anticancer potential with a mechanism similar to vinca alkaloids, the commonly used chemotherapy drugs. We found that G-1 blocks tubulin polymerization and thereby interrupts microtubule assembly in ovarian cancer cells leading to the arrest of cell cycle in the prophase of mitosis and the suppression of ovarian cancer cell proliferation. G-1 treatment also induces apoptosis of ovarian cancer cells. The ability of G-1 to target microtubules to suppress ovarian cancer cell proliferation makes it a promising candidate drug for treatment of ovarian cancer.


Assuntos
Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Polimerização/efeitos dos fármacos , Receptores de Estrogênio/agonistas , Receptores Acoplados a Proteínas G/agonistas , Tubulina (Proteína)/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclopentanos/farmacologia , Feminino , Humanos , Prófase/efeitos dos fármacos , Quinolinas/farmacologia , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Sus scrofa
18.
Biochem Pharmacol ; 86(12): 1627-42, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24099797

RESUMO

Cardiovascular disease (CVD) is less common in premenopausal women than men of the same age or postmenopausal women, suggesting vascular benefits of estrogen. Estrogen activates estrogen receptors ERα, ERß and GPR30 in endothelium and vascular smooth muscle (VSM), which trigger downstream signaling pathways and lead to genomic and non-genomic vascular effects such as vasodilation, decreased VSM contraction and growth and reduced vascular remodeling. However, randomized clinical trials (RCTs), such as the Women's Health Initiative (WHI) and Heart and Estrogen/progestin Replacement Study (HERS), have shown little vascular benefits and even adverse events with menopausal hormone therapy (MHT), likely due to factors related to the MHT used, ER profile, and RCT design. Some MHT forms, dose, combinations or route of administration may have inadequate vascular effects. Age-related changes in ER amount, distribution, integrity and post-ER signaling could alter the vascular response to MHT. The subject's age, preexisting CVD, and hormone environment could also reduce the effects of MHT. Further evaluation of natural and synthetic estrogens, phytoestrogens, and selective estrogen-receptor modulators (SERMs), and the design of appropriate MHT combinations, dose, route and 'timing' could improve the effectiveness of conventional MHT and provide alternative therapies in the peri-menopausal period. Targeting ER using specific ER agonists, localized MHT delivery, and activation of specific post-ER signaling pathways could counter age-related changes in ER. Examination of the hormone environment and conditions associated with hormone imbalance such as polycystic ovary syndrome may reveal the causes of abnormal hormone-receptor interactions. Consideration of these factors in new RCTs such as the Kronos Early Estrogen Prevention Study (KEEPS) could enhance the vascular benefits of estrogen in postmenopausal CVD.


Assuntos
Vasos Sanguíneos/fisiopatologia , Terapia de Reposição de Estrogênios , Estrogênios/fisiologia , Receptores de Estrogênio/fisiologia , Doenças Vasculares/fisiopatologia , Relação Dose-Resposta a Droga , Terapia de Reposição de Estrogênios/efeitos adversos , Feminino , Humanos , Pessoa de Meia-Idade , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores
19.
J Immunol ; 191(9): 4628-39, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24062489

RESUMO

Neutrophils are major participants in innate host responses. It is well known that estrogens have an immune-modulatory role, and some evidence exists that neutrophil physiology can be altered by these molecules. Traditionally, estrogens act via classical nuclear estrogen receptors, but the identification of a G protein-coupled estrogen receptor (GPER), a membrane estrogen receptor that binds estradiol and other estrogens, has opened up the possibility of exploring additional estrogen-mediated effects. However, information on the importance of GPER for immunity, especially, in neutrophils is scant. In this study, we report that gilthead seabream (Sparus aurata L.) acidophilic granulocytes, which are the functional equivalent of mammalian neutrophils, express GPER at both mRNA and protein levels. By using a GPER selective agonist, G1, it was found that GPER activation in vitro slightly reduced the respiratory burst of acidophilic granulocytes and drastically altered the expression profile of several genes encoding major pro- and anti-inflammatory mediators. In addition, GPER signaling in vivo modulated adaptive immunity. Finally, a cAMP analog mimicked the effects of G1 in the induction of the gene coding for PG-endoperoxide synthase 2 and in the induction of CREB phosphorylation, whereas pharmacological inhibition of protein kinase A superinduced PG-endoperoxide synthase 2. Taken together, our results demonstrate for the first time, to our knowledge, that estrogens are able to modulate vertebrate granulocyte functions through a GPER/cAMP/protein kinase A/CREB signaling pathway and could establish therapeutic targets for several immune disorders in which estrogens play a prominent role.


Assuntos
Estrogênios/metabolismo , Granulócitos/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células Cultivadas , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Granulócitos/imunologia , Ativação de Neutrófilo , Neutrófilos/imunologia , Fosforilação , Prostaglandina-Endoperóxido Sintases/genética , RNA Mensageiro/biossíntese , Receptores de Estrogênio/agonistas , Receptores Acoplados a Proteínas G/agonistas , Dourada , Transdução de Sinais
20.
Gynecol Endocrinol ; 29(11): 993-6, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24000936

RESUMO

OBJECTIVE: To assess the advantages and disadvantages of using letrozole for controlled ovarian stimulation (COH) in young patients with estrogen receptor-positive (ER+) breast cancer, wishing to cryopreserve oocytes. DESIGN: Retrospective cohort analysis. SETTING: Sixteen Italian units for reproductive medicine and in vitro fertilization. METHODS: Data of 50 ER+ breast cancer patients undergoing COH to cryopreserve oocytes before gonadotoxic chemotherapy with a letrozole plus gonadotropins (Le+Gn) protocol were compared with those of 25 young women with ER- breast cancer, submitted to COH using a protocol with gonadotropins alone (Gn-only). RESULTS: The Le+Gn protocol implied a significantly lower total Gn consumption and allowed to maintain significantly lower circulating E2 levels at all checkpoints throughout stimulation (peak E2 value 446 ± 357 versus 1553 ± 908 pg/ml, respectively; p = 0.001). On the other side, the Le+Gn protocol allowed a significantly lower yield of oocytes available for cryostorage (6.6 ± 3.5 versus 8 ± 5, respectively; p = 0.038). CONCLUSIONS: In breast cancer patients, the association of letrozole to Gn significantly reduces the number of oocytes available for cryostorage in comparison with the use of Gn alone. On the other side, it is associated with significantly lower E2 levels during the whole stimulation cycle, a safety issue that has been traditionally considered advantageous in case of ER+ cancers.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Nitrilas/uso terapêutico , Indução da Ovulação , Receptores de Estrogênio/metabolismo , Triazóis/uso terapêutico , Adulto , Antineoplásicos/efeitos adversos , Inibidores da Aromatase/efeitos adversos , Neoplasias da Mama/sangue , Neoplasias da Mama/metabolismo , Estudos de Coortes , Criopreservação , Estradiol/sangue , Feminino , Preservação da Fertilidade/efeitos adversos , Gonadotropinas/uso terapêutico , Humanos , Itália , Letrozol , Proteínas de Neoplasias/agonistas , Nitrilas/efeitos adversos , Recuperação de Oócitos , Oócitos , Oogênese/efeitos dos fármacos , Receptores de Estrogênio/agonistas , Estudos Retrospectivos , Triazóis/efeitos adversos , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...