Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 410
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
IUBMB Life ; 74(10): 1012-1028, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36054915

RESUMO

Steviol glycosides, the active sweet components of stevia plant, have been recently found to possess a number of therapeutic properties, including some recorded anticancer ones against various cancer cell types (breast, ovarian, cervical, pancreatic, and colon cancer). Our aim was to investigate this anticancer potential on the two most commonly used breast cancer cell lines which differ in the phenotype and estrogen receptor (ER) status: the low metastatic, ERα+ MCF-7 and the highly metastatic, ERα-/ERß+ MDA-MB-231. Specifically, glycosides' effect was studied on cancer cells': (a) viability, (b) functionality (proliferation, migration, and adhesion), and (c) gene expression (mRNA level) of crucial molecules implicated in cancer's pathophysiology. Results showed that steviol glycosides induced cell death in both cell lines, in the first 24 hr, which was in line with the antiapoptotic BCL2 decrease. However, cells that managed to survive showcased diametrically opposite behavior. The low metastatic ERα+ MCF-7 cells acquired an aggressive phenotype, depicted by the upregulation of all receptors and co-receptors (ESR, PGR, AR, GPER1, EGFR, IGF1R, CD44, SDC2, and SDC4), as well as VIM and MMP14. On the contrary, the highly metastatic ERα-/ERß+ MDA-MB-231 cells became less aggressive as pointed out by the respective downregulation of EGFR, IGF1R, CD44, and SDC2. Changes observed in gene expression were compatible with altered cell functions. Glycosides increased MCF-7 cells migration and adhesion, but reduced MDA-MB-231 cells migratory and metastatic potential. In conclusion, the above data clearly demonstrate that steviol glycosides have different effects on breast cancer cells according to their ER status, suggesting that steviol glycosides might be examined for their potential anticancer activity against breast cancer, especially triple negative breast cancer (TNBC).


Assuntos
Receptor alfa de Estrogênio , Neoplasias , Diterpenos do Tipo Caurano , Receptores ErbB , Receptor beta de Estrogênio/fisiologia , Glucosídeos , Glicosídeos/farmacologia , Metaloproteinase 14 da Matriz , Proteínas Proto-Oncogênicas c-bcl-2 , RNA Mensageiro , Receptores de Estrogênio
2.
Inflamm Res ; 71(2): 255-266, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35064304

RESUMO

BACKGROUND: The role of estrogen receptor ß (ERß) in the pathogenesis and development of breast cancer (BC) is controversial, and it is currently considered to play contradictory roles in different phenotypes. ERß2 is thought to promote the BC process, but its role in triple-negative breast cancer (TNBC) has not been reported. METHODS: In this study, we collected tumor tissues from 15 patients with TNBC and obtained a variety of TNBC cell lines as research objects. The plasmid vectors and RNA interference techniques were used to change the level of target genes in cells, quantitative PCR and Western Blots were used to detect gene expression levels, CCK-8 and EdU assay were used to detect cell growth, and Transwell was used to detect cell migration and invasion. Dual-luciferase gene reports and RNA immunoprecipitation (RIP) were used to verify gene targeting relationships. RESULTS: ERß2 was up-regulated in TNBC tissues and promoted the growth, migration, and invasion of TNBC cells. ERß2 regulated hsa_circ_0000732 expression by binding to SCARF1 promoter. Knockdown of hsa_circ_0000732 inhibited TNBC cell proliferation, migration, and invasion by upregulating miR-1184. CONCLUSION: Our present study found that ERß2 is upregulated in some TNBC cells and promotes TNBC cell growth, migration and invasion by regulating hsa_circ_0000732 targeting miR-1184. The special role of ERß2 in TNBC may be the breakthrough of a targeted treatment strategy for TNBC.


Assuntos
Receptor beta de Estrogênio/fisiologia , MicroRNAs/fisiologia , RNA Circular/fisiologia , Neoplasias de Mama Triplo Negativas/etiologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Invasividade Neoplásica , Regiões Promotoras Genéticas , Receptores Depuradores Classe F/genética , Neoplasias de Mama Triplo Negativas/patologia , Regulação para Cima
3.
Mol Biol Rep ; 49(2): 1223-1232, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34792729

RESUMO

BACKGROUND: Female breast cancer has become the most commonly diagnosed cancer worldwide. As a tumor suppressor, estrogen receptor ß (ERß) can be potentially targeted for breast cancer therapy. METHODS AND RESULTS: TAD1822-7 was evaluated for ERß-mediated autophagy and cell death using cell proliferation assay, Annexin V/PI staining, immunofluorescence, western blotting, ERß siRNA, ERß plasmid transfection and hypoxia cell models. TAD1822-7 upregulated ERß causing cell death and induced mitochondrial dysfunction and autophagy companied with mitochondrial located ERß. Enhanced levels of microtubule associated protein1 light chain 3 (LC3)-II and p62/SQSTM1 (p62) indicated that TAD1822-7 blocked the late-stage autolysosome formation, leading to cell death. Mechanistically, TAD1822-7-induced cell death was mediated by phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathways. Moreover, TAD1822-7 modulated hypoxia inducible factor (HIF) functions and autophagy via the inhibition of HIF-1ß in the context of hypoxia-induced autophagy. ERß overexpression and ERß agonist showed similar effects, whereas ERß siRNA abrogated TAD1822-7-induced cell death, the inhibition of PI3K/AKT pathway and autophagy. The involvement of PI3K/AKT pathway and autophagy was also demonstrated in TAD1822-7-treated hypoxic breast cancer cells. CONCLUSIONS: These findings provide new insight into the mechanism underlying the inhibitory effects of TAD1822-7 via ERß-mediated pathways in breast cancer cells.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Receptor beta de Estrogênio/metabolismo , Morfolinas/farmacologia , Compostos de Fenilureia/farmacologia , Alcaloides , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Compostos de Bifenilo , Neoplasias da Mama/metabolismo , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptor beta de Estrogênio/fisiologia , Feminino , Humanos , Morfolinas/metabolismo , Compostos de Fenilureia/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ureia
4.
Biol Pharm Bull ; 44(11): 1594-1597, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34719637

RESUMO

Estrogen is essential for the growth and development of mammary glands and its signaling is associated with breast cancer growth. Estrogen can exert physiological actions via estrogen receptors α/ß (ERα/ß). There is experimental evidence suggesting that in ERα/ß-positive breast cancer, ERα promotes tumor cell proliferation and ERß inhibits ERα-mediated transcriptional activity, resulting in abrogation of cell growth. Therefore, ERß is attracting attention as a potential tumor suppressor, and as a biomarker and therapeutic target in the ERα/ß-positive breast cancer. Based on this information, we have hypothesized that some endocrine-disrupting chemicals (EDCs) that can perturb the balance between ERα and ERß expression levels in breast cancer cells might have effects on the breast cancer proliferation (i.e., down-regulation of the α-type of ER). We have recently reported that 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), an active metabolite of bisphenol A, in ERα/ß-positive human breast cancer significantly down-regulates ERα expression, yet stimulates cell proliferation through the activation of ERß-mediated transcription. These results support our hypothesis by demonstrating that exposure to MBP altered the functional role of ERß in breast cancer cells from suppressor to promoter. In contrast, some EDCs, such as Δ9-tetrahydrocannabinol and bisphenol AF, can exhibit anti-estrogenic effects through up-regulation of ERß expression without affecting the ERα expression levels. However, there is no consensus on the correlation between ERß expression levels and clinical prognosis, which might be due to differences in exposed chemicals. Therefore, elucidating the exposure effects of EDCs can reveal the reason for inconsistent functional role of ERß in ERα/ß-positive breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Disruptores Endócrinos/toxicidade , Receptor beta de Estrogênio/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Neoplasias da Mama/induzido quimicamente , Antagonistas de Estrogênios/uso terapêutico , Receptor beta de Estrogênio/metabolismo , Receptor beta de Estrogênio/fisiologia , Feminino , Humanos
5.
Horm Behav ; 125: 104827, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32735801

RESUMO

Estrogens play a key role in the sexual differentiation of the brain and behavior. While early estrogen actions exert masculinizing effects on the brain of male rodents, a diametrically opposite effect is observed in birds where estrogens demasculinize the brain of females. Yet, the two vertebrate classes express similar sex differences in the brain and behavior. Although ERα is thought to play a major role in these processes in rodents, the role of ERß is still controversial. In birds, the identity of the estrogen receptor(s) underlying the demasculinization of the female brain remains unclear. The aim of the present study was thus to determine in Japanese quail the effects of specific agonists of ERα (propylpyrazole triol, PPT) and ERß (diarylpropionitrile, DPN) administered at the beginning of the sensitive period (embryonic day 7, E7) on the sexual differentiation of male sexual behavior and on the density of vasotocin-immunoreactive (VT-ir) fibers, a known marker of the organizational action of estrogens on the quail brain. We demonstrate that estradiol benzoate and the ERß agonist (DPN) demasculinize male sexual behavior and decrease the density of VT-ir fibers in the medial preoptic nucleus and the bed nucleus of the stria terminalis, while PPT has no effect on these measures. These results clearly indicate that ERß, but not ERα, is involved in the estrogen-induced sexual differentiation of brain and sexual behavior in quail.


Assuntos
Encéfalo/anatomia & histologia , Coturnix/fisiologia , Receptor beta de Estrogênio/fisiologia , Comportamento Sexual Animal , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Coturnix/metabolismo , Estradiol/análogos & derivados , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/agonistas , Estrogênios/farmacologia , Feminino , Masculino , Nitrilas/farmacologia , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Propionatos/farmacologia , Caracteres Sexuais , Diferenciação Sexual/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Vasotocina/farmacologia
6.
Proc Natl Acad Sci U S A ; 114(48): E10428-E10437, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29133394

RESUMO

The ability to propagate mature cells and tissue from pluripotent stem cells offers enormous promise for treating many diseases, including neurodegenerative diseases. Before such cells can be used successfully in neurodegenerative diseases without causing unwanted cell growth and migration, genes regulating growth and migration of neural stem cells need to be well characterized. Estrogen receptor beta (ERß) is essential for migration of neurons and glial cells in the developing mouse brain. To examine whether ERß influences differentiation of mouse embryonic stem cells (mESC) into neural lineages, we compared control and ERß knockout (BERKO) mESCs at defined stages of neural development and examined the effects of an ERß-selective ligand (LY3201) with a combination of global and targeted gene-expression profiling and the expression of key pluripotency markers. We found that ERß was induced in embryoid bodies (EBs) and neural precursor cells (NPCs) during development. Proliferation was higher in BERKO NPCs and was inhibited by LY3201. Neurogenesis was reduced in BERKO ES cells, and oligodendrogliogenesis was enhanced. BERKO EBs expressed higher levels of key ectodermal and neural progenitor markers and lower levels of markers for mesoderm and endoderm lineages. ERß-regulated factors are involved in cell adhesion, axon guidance, and signaling of Notch and GABA receptor pathways, as well as factors important for the differentiation of neuronal precursors into dopaminergic neurons (Engrailed 1) and for the oligodendrocyte fate acquisition (Olig2). Our data suggest that ERß is an important component for differentiation into midbrain neurons as well as for preventing precocious oligodendrogliogenesis.


Assuntos
Diferenciação Celular/fisiologia , Receptor beta de Estrogênio/fisiologia , Mesencéfalo/fisiologia , Células-Tronco Embrionárias Murinas/fisiologia , Células-Tronco Neurais/fisiologia , Regeneração/fisiologia , Animais , Benzopiranos/farmacologia , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Neurônios Dopaminérgicos/fisiologia , Receptor beta de Estrogênio/agonistas , Feminino , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Mesencéfalo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Oligodendroglia/fisiologia , Transdução de Sinais/fisiologia
7.
Int J Mol Sci ; 21(21)2020 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-33153202

RESUMO

Endometriosis is defined as the presence of endometrial foci, localized beyond their primary site, i.e., the uterine cavity. The etiology of this disease is rather complex. Its development is supported by hormonal, immunological, and environmental factors. During recent years, particular attention has been focused on the genetic mechanisms that may be of particular significance for the increased incidence rates of endometriosis. According to most recent studies, ESR2 and CYP19A1 genes may account for the potential risk factors of infertility associated with endometriosis. The paper presents a thorough review of the latest reports and data concerning the genetic background of the risk for endometriosis development.


Assuntos
Aromatase/fisiologia , Endometriose/genética , Receptor beta de Estrogênio/fisiologia , Patrimônio Genético , Endometriose/epidemiologia , Endometriose/etiologia , Endométrio/metabolismo , Endométrio/patologia , Feminino , Predisposição Genética para Doença , Humanos , Infertilidade/genética , Fatores de Risco
8.
Am J Respir Cell Mol Biol ; 61(4): 469-480, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30958966

RESUMO

Evidence suggests that airway hyperresponsiveness (AHR) is a characteristic feature of asthma. Epidemiological studies have confirmed that the severity of asthma is greater in women, suggesting a critical role of female sex steroid hormones (especially estrogen). Very few in vivo studies have examined the role of sex steroid hormones in asthma, and the sequence of events that occur through differential activation of estrogen receptors (ERs) remains to be determined in asthmatic airways. Our recent in vitro findings indicated that ERß had increased expression in asthmatic airway smooth muscle (ASM), and that its activation by an ERß-specific agonist downregulated airway remodeling. In this study, we translated the in vitro findings to a murine asthma model and examined the differential role of ER activation in modulating lung mechanics. C57BL/6J male, female, and ovariectomized mice were exposed to mixed allergen (MA) and subcutaneously implanted with sustained-release pellets of placebo, an ERα agonist (4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol [PPT]), and/or an ERß agonist (WAY-200070). We then evaluated the effects of these treatments on airway mechanics, biochemical, molecular, and histological parameters. Mice exposed to MA showed a significant increase in airway resistance, elastance, and tissue damping, and a decrease in compliance; pronounced effects were observed in females. Compared with PPT, WAY treatment significantly reversed the MA-induced changes. The increased mRNA/protein expression of ERα, ERß, and remodeling genes observed in MA-treated mice was significantly reversed in WAY-treated mice. This novel study indicates that activation of ERß signaling downregulates AHR and airway remodeling, and is a promising target in the development of treatments for asthma.


Assuntos
Remodelação das Vias Aéreas/fisiologia , Asma/fisiopatologia , Hiper-Reatividade Brônquica/fisiopatologia , Receptor beta de Estrogênio/fisiologia , Estrogênios/fisiologia , Resistência das Vias Respiratórias , Alérgenos/toxicidade , Animais , Modelos Animais de Doenças , Implantes de Medicamento , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/agonistas , Feminino , Microdissecção e Captura a Laser , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Oxazóis/farmacologia , Fenóis/farmacologia , Pirazóis/farmacologia , Caracteres Sexuais
9.
J Pharmacol Sci ; 138(1): 1-8, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30236540

RESUMO

Androgen-independent prostate cancer (PCa) is a developed tumor derived from the local androgen dependent PCa, which often affects elderly men. Psoralea corylifolia L, a traditional Chinese medicine, has been widely used for PCa treatment as an important part of a common prescription, while the mechanism remains unclear. Our study was aimed to investigate the tumor-inhibitory effect of its main component bakuchiol in androgen-independent PCa cell line PC-3 cells. Bakuchiol significantly suppressed PC-3 cell proliferation and migration; the expressions of PCNA and MMP-9 were consistently down regulated as well. Meanwhile, both the constitutive and LPS-induced NF-κB activation were significantly inhibited by bakuchiol. The inhibitory effects of bakuchiol on cell proliferation, migration and invasion were recovered when LPS were added together with bakuchiol. SiRNA against androgen receptor (AR) or estrogen receptor ß (ERß) were transfected and the regulation of bakuchiol-suppressed proliferation, invasion, NF-κB signaling and MMP-9 secretion in response to LPS were blocked. Taken together, our data demonstrated that bakuchiol inactivated NF-κB signaling via AR and ERß, which contributes to inhibition of PC-3 cell proliferation and migration, indicating that bakuchiol is one of the key component from P. corylifolia L for PCa treatment and has a potential as anti-prostate cancer drug candidates.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Receptor beta de Estrogênio/fisiologia , NF-kappa B/metabolismo , Fenóis/farmacologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores Androgênicos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo/efeitos dos fármacos , Receptor beta de Estrogênio/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Humanos , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Fenóis/isolamento & purificação , Fenóis/uso terapêutico , Fitoterapia , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Psoralea/química , RNA Interferente Pequeno , Receptores Androgênicos/genética
10.
Adv Exp Med Biol ; 1071: 115-127, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30357742

RESUMO

We tested the hypothesis that ERß is involved in respiratory control in female mice. We used young adult (5-6 months-old) and aged (17-18 months-old) ERßKO or wild-type controls (WT) female mice to assess arterial blood pressure (via a tail-cuff sensor) and indices of respiratory pattern (sighs and apneas - recorded by whole body plethysmography at rest). We also measured respiratory parameters at rest and in response to brief (<10 min) exposure to hypoxia (12% O2) or hypercapnia (5% CO2). Because ERß is localized in mitochondria, and because estradiol and ERß agonist increase mitochondrial O2 consumption, we assessed the mitochondrial respiration (with a high-resolution oxygraph system) and the in vitro activity of the complex I of the electron transfer chain in samples of brain cortex in aged wild-type and ERßKO female mice. Compared to young WT mice, young ERßKO mice had elevated arterial blood pressure, but similar ventilatory responses to hypoxia and hypercapnia. In old ERßKO female mice compared to old WT mice, the arterial blood pressure was lower, the frequency of sighs was higher and the frequency of apneas was lower, and the hypoxic and hypercapnic ventilatory responses were reduced. In old ERßKO mice mitochondrial respiration and complex I activities in the brain cortex were lower than in WT mice. We conclude that ERß has age-specific effects on vascular and respiratory functions in female mice.


Assuntos
Fatores Etários , Pressão Arterial , Receptor beta de Estrogênio/fisiologia , Mitocôndrias/fisiologia , Animais , Feminino , Hipercapnia/fisiopatologia , Hipóxia/fisiopatologia , Camundongos
11.
J Biol Chem ; 291(28): 14747-60, 2016 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-27226548

RESUMO

Current pharmacotherapies for symptomatic benign prostatic hyperplasia (BPH), an androgen receptor-driven, inflammatory disorder affecting elderly men, include 5α-reductase (5AR) inhibitors (i.e. dutasteride and finasteride) to block the conversion of testosterone to the more potent androgen receptor ligand dihydrotestosterone. Because dihydrotestosterone is the precursor for estrogen receptor ß (ERß) ligands, 5AR inhibitors could potentially limit ERß activation, which maintains prostate tissue homeostasis. We have uncovered signaling pathways in BPH-derived prostate epithelial cells (BPH-1) that are impacted by 5AR inhibition. The induction of apoptosis and repression of the cell adhesion protein E-cadherin by the 5AR inhibitor dutasteride requires both ERß and TGFß. Dutasteride also induces cyclooxygenase type 2 (COX-2), which functions in a negative feedback loop in TGFß and ERß signaling pathways as evidenced by the potentiation of apoptosis induced by dutasteride or finasteride upon pharmacological inhibition or shRNA-mediated ablation of COX-2. Concurrently, COX-2 positively impacts ERß action through its effect on the expression of a number of steroidogenic enzymes in the ERß ligand metabolic pathway. Therefore, effective combination pharmacotherapies, which have included non-steroidal anti-inflammatory drugs, must take into account biochemical pathways affected by 5AR inhibition and opposing effects of COX-2 on the tissue-protective action of ERß.


Assuntos
Inibidores de 5-alfa Redutase/farmacologia , Ciclo-Oxigenase 2/metabolismo , Dutasterida/farmacologia , Receptor beta de Estrogênio/fisiologia , Próstata/metabolismo , Células Cultivadas , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Humanos , Masculino , Prostaglandinas/biossíntese , Próstata/citologia , Próstata/enzimologia
12.
Biochem Biophys Res Commun ; 493(1): 100-107, 2017 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-28919414

RESUMO

Recently, several studies have indicated that circular RNAs (circRNAs) play significant roles in various disease; however, little is known about the chronology of estrogen receptor beta (ERß) deficiency and altered circRNA expression, or their relationship with osteogenesis. Herein, we show through western-blot and quantitative real-time PCR assays, that when ERß is silenced, the expression of osteogenesis-related proteins and mRNAs were down-regulated. We then performed RNA-Seq to analyze differential circRNA expression between the control and ERß knockdown group. This analysis revealed that, 146 circRNAs were differentially expressed by fold-change≥2.0, p ≤ 0.05, and, among this group, 68 circRNAs were down-regulated, while 78 were up-regulated. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and PANTHER pathway analyses were performed to predict the function of these differentially expressed circRNAs. Finally, co-expressed targets gene, and circRNA-microRNA network were constructed for predicted miRNA sponges. This research suggested that ERß may through 2:27713879|27755789/2:240822115|240867796-miR-328-5p-mRNA axis to regulate osteogenic differentiation.


Assuntos
Receptor beta de Estrogênio/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Osteogênese/fisiologia , RNA/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Técnicas de Silenciamento de Genes , RNA Circular , Ratos
13.
Am J Physiol Regul Integr Comp Physiol ; 311(1): R14-23, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27122368

RESUMO

Estradiol (E2) decreases both water and saline intakes by female rats. The ERα and ERß subtypes are expressed in areas of the brain that control fluid intake; however, the role that these receptors play in E2's antidipsogenic and antinatriorexigenic effects have not been examined. Accordingly, we tested the hypothesis that activation of ERα and ERß decreases water and saline intakes by female rats. We found a divergence in E2's inhibitory effect on intake: activation of ERα decreased water intake, whereas activation of ERß decreased saline intake. E2 decreases expression of the angiotensin II type 1 receptor (AT1R), a receptor with known relevance to water and salt intakes, in multiple areas of the brain where ERα and ERß are differentially expressed. Therefore, we tested for agonist-induced changes in AT1R mRNA expression by RT-PCR and protein expression by analyzing receptor binding to test the hypothesis that the divergent effects of these ER subtypes are mediated by region-specific changes in AT1R expression. Although we found no changes in AT1R mRNA or binding in areas of the brain known to control fluid intake associated with agonist treatment, the experimental results replicate and extend previous findings that body weight changes mediate alterations in AT1R expression in distinct brain regions. Together, the results reveal selective effects of ER subtypes on ingestive behaviors, advancing our understanding of E2's inhibitory role in the controls of fluid intake by female rats.


Assuntos
Peso Corporal/fisiologia , Ingestão de Líquidos/fisiologia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Receptor Tipo 1 de Angiotensina/biossíntese , Receptor Tipo 1 de Angiotensina/genética , Angiotensina II/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Química Encefálica/genética , Ingestão de Líquidos/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/efeitos dos fármacos , Estrogênios/farmacologia , Feminino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Long-Evans , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos
14.
Clin Sci (Lond) ; 130(5): 365-76, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26608078

RESUMO

ERß (oestrogen receptor ß) activation has been shown to be cardioprotective, but the cell types and mechanisms involved are not understood. To investigate whether ERß restricted to cardiomyocytes contributes to the observed cardioprotection, we tested the effects of cardiomyocyte-specific ERß-OE (ERß overexpression) on survival, cardiac remodelling and function after MI (myocardial infarction) and studied the molecular pathways potentially involved. Female and male mice with cardiomyocyte-specific ERß-OE and WT (wild-type) littermates were subjected to chronic anterior coronary artery ligation or sham surgery. Two weeks after MI, ERß-OE mice showed improved survival (100% and 83% compared with 76% and 58% in WT females and males respectively). ERß-OE was associated with attenuated LV (left ventricular) dilatation, smaller increase in heart weight, less lung congestion at similar MI size, and improved systolic and diastolic function in both sexes. We identified two potential pathways for ERß-mediated myocardial protection. First, male and female ERß-OE mice had a lower reduction of SERCA2a (sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase 2a) expression after MI, suggesting less reduction in diastolic Ca(2+)-reuptake into the sarcoplasmic reticulum post-MI. Secondly, male ERß-OE revealed attenuated cardiac fibrosis in the remote LV tissue and expression of fibrosis markers collagen I and III, periostin and miR-21. Cardiomyocyte-specific ERß-OE improved survival associated with reduced maladaptive remodelling, improved cardiac function and less heart failure development after MI in both sexes. These effects seem to be related, at least in part, to a better maintenance of Ca(2+) cycling in both sexes and a lower induction of cardiac fibrosis in males after MI.


Assuntos
Receptor beta de Estrogênio/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Biomarcadores/metabolismo , Diástole/fisiologia , Receptor beta de Estrogênio/fisiologia , Feminino , Fibrose , Masculino , Camundongos Transgênicos , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Fatores Sexuais , Sístole/fisiologia , Ultrassonografia , Remodelação Ventricular/fisiologia
15.
Horm Behav ; 86: 1-7, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27594441

RESUMO

The present study was designed to assess the participation of estrogen receptors alpha (ERα) and beta (ERß) in the short-term facilitation of lordosis behavior in ovariectomized (ovx), estradiol (E2) primed rats. In experiment 1, dose response curves for PPT and DPN (ERα and ERß agonists, respectively) facilitation of lordosis behavior (lordosis quotient and lordosis score) were established by infusing these agonists into the right lateral ventricle (icv) in female rats injected 40h previously with 5µg of E2 benzoate. PPT doses of 0.08 and 0.4ng produced high lordosis quotients starting at 30min and continuing at 120 and 240min post-injection. DPN induced high levels of lordosis behavior at all times tested. However, the intensity of lordosis induced by both agonists was weak. In experiment 2, we tested the involvement of each ER in facilitation of lordosis by icv infusion of MPP (ERα-selective antagonist) or PHTPP (ERß-selective antagonist) prior to infusion of 2ng of free E2. Icv infusion of either MPP or PHTPP 30min before free E2 significantly depressed E2 facilitation of lordosis. The results suggest that both forms of ER are involved in the short-latency facilitation of lordosis behavior in E2-primed rats.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Postura/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/fisiologia , Feminino , Masculino , Ovariectomia , Ratos , Ratos Sprague-Dawley , Comportamento Sexual Animal/efeitos dos fármacos
16.
Climacteric ; 19(3): 247-8, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27021900

RESUMO

Many studies have investigated potential links between postmenopausal hormone therapy (HT) and the risk for colorectal cancer. Most of these studies showed some reduction in risk, but association with the exact grade of the tumor or with cancer mortality is less consistent. Activation of estrogen receptor beta (ERß) related cellular pathways is probably the pathophysiological basis for this HT effect, since ERßs exert antiproliferative and pro-apoptotic signaling, inhibit inflammatory signaling and modulate the tumor microenvironment. To note, the impact on health may not be substantial as the absolute numbers point at the order of only a few saved cases per 10 000 women per year of hormone use.


Assuntos
Neoplasias Colorretais/epidemiologia , Neoplasias Colorretais/prevenção & controle , Terapia de Reposição de Estrogênios , Pós-Menopausa , Adulto , Idoso , Idoso de 80 Anos ou mais , Receptor beta de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/fisiologia , Feminino , Humanos , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto , Fatores de Risco
17.
Gen Comp Endocrinol ; 233: 100-108, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27222348

RESUMO

Estrogen receptors ESR1, ESR2 and GPER are present on mature ejaculated horse spermatozoa, suggesting these cells as putative targets for estrogens. Indeed, spermatozoa are exposed to high level of estrogens during the transit in the male and female genital tracts but their roles are not investigated. So, we evaluated in vitro the role of 17ß-estradiol during post-testicular maturations: regulation of motility, capacitation and acrosome reaction. Moreover according to the pseudo-seasonal breeder status of the stallion, we analyzed the putative seasonal variations in the presence of ESRs in spermatozoa. We showed that ESRs are more present on stallion sperm during the breeding season. We showed that capacitation and acrosome reaction are independent of estradiol action in horse. Estradiol can weakly modulate the motility and this effect is strictly associated with GPER and not with ESR1 and ESR2. The subcellular localization of GPER in the neck on stallion sperm is coherent with this effect. It seems that estrogens are not major regulators of sperm maturations associated to mare genital tract, so they could act during the epididymal maturations.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Cavalos/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Capacitação Espermática , Maturação do Esperma , Reação Acrossômica/efeitos dos fármacos , Animais , Epididimo/efeitos dos fármacos , Epididimo/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Cavalos/genética , Masculino , Receptores de Estrogênio/metabolismo , Receptores de Estrogênio/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Capacitação Espermática/efeitos dos fármacos , Maturação do Esperma/efeitos dos fármacos , Maturação do Esperma/fisiologia , Motilidade dos Espermatozoides/efeitos dos fármacos , Transporte Espermático/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo , Espermatozoides/fisiologia , Distribuição Tecidual
18.
Arch Gynecol Obstet ; 293(6): 1161-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26861465

RESUMO

INTRODUCTION: Ovarian cancer remains the leading cause of mortality due to gynecological tumors. Estrogen receptors (ERs) seem to participate in tumor progression even in the absence of estrogens. Twenty years after the cloning of the second estrogen receptor, a wide spectrum of studies have shown its implication in both physiologic and pathologic pathways. ERß, being the predominant type of ER in normal ovary tissue, has not only been linked with pathogenesis of ovarian cancer, but also with response to treatment. Unlike ERα, which is primarily linked with cell growth, ERß presence is prominent in signaling pathways, cell cycle regulation and apoptosis. METHODS: Literature review of relevant published material (from PubMed, Scopus, and Cochrane databases) was conducted. RESULTS: Polymorphisms in the respective ESR2 gene, epigenetic modifications and isoforms of the receptor have been extensively studied to assess potential correlations with responsiveness to treatment and tumor behavior. Studies on the exact roles of ERß and its genetic variations in altering effectiveness and toxicity of ovarian cancer treatment regimens are lacking. CONCLUSION: Clinical utilization of ERß actions in the management of ovarian cancer is discussed in an up-to-date review.


Assuntos
Receptor beta de Estrogênio , Neoplasias Ovarianas , Antineoplásicos/uso terapêutico , Apoptose , Ciclo Celular/fisiologia , Proliferação de Células , Epigênese Genética , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/fisiologia , Feminino , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Polimorfismo Genético , Isoformas de Proteínas , Transdução de Sinais/fisiologia
19.
Genet Mol Res ; 15(3)2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27706665

RESUMO

The role of estrogen in inducing chemoresistance is not yet fully understood. The objective of this study was to observe the relationship between estrogen levels and cellular response to chemotherapeutic drugs in non-small cell lung cancer (NSCLC) and to reveal the potential mechanisms involved. Cell viability was analyzed after pre-treating NSCLC cells with different levels of estrogen (E2), followed by treatment with an anti-tumor drug for 48 h. The roles of various estrogen receptors (ERs) were examined in vitro by blocking the activity of each ER individually. The ER pathway was further confirmed in NSCLC tissues. It was found that 10-1000 nM E2 resulted in a decreased cellular response to DDP in H1650 cells compared to the use of cisplatin alone (P < 0.05). However, this result was not demonstrated in H1299 cells, which lack p53. Both ERa and ERb were associated with E2-induced cisplatin chemoresistance, though they had opposite functions. p53 expression did not correlate with the expression of ERa or ERb individually. However, a statistically significant correlation between p53 expression and ERa to ERb mRNA ratio was observed (P < 0.001, R = -0.676). These findings suggest that E2-induced DDP chemoresistance depends on the balance between ERa and ERb expression and the p53 pathway.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Cisplatino/farmacologia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Neoplasias Pulmonares/metabolismo , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Estradiol/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
20.
IUBMB Life ; 67(11): 861-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26488768

RESUMO

Estrogen exerts its physiological and pathological functions through two estrogen receptors (ERs), ERα and ERß, which act as transcription factors. Coregulators, including coactivators and corepressors, have been shown to be crucial for regulation of ER transcriptional activity. Although many coregulators have been identified to regulate activities of ERs, novel coregulators are still needed to be investigated. Here, we show that human methyltransferase-like 17 (METTL17), whose function is unknown, physically interacts with ERα and ERß, and functionally acts as a coactivator for ERs. METTL17 interacts with ER in vitro and in yeast and mammalian cells. Activation function-1 (AF1) and AF2 domains of ERs are responsible for the interaction between METTL17 and ERs. Knockdown of METTL17 reduces transcriptional activities of ERα and ERß in breast cancer cells, whereas METTL17 overexpression increases ERα and ERß transcriptional activities. Inhibition of METTL17 expression decreases mRNA and protein levels of ER target genes, including PR, cathepsin D, and pS2. Moreover, METTL17 knockdown reduces breast cancer cell growth. These results indicate that METTL17 is a novel coactivator of ERs and may play a role in breast tumorigenesis.


Assuntos
Neoplasias da Mama/enzimologia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Metiltransferases/fisiologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Estradiol/fisiologia , Receptor alfa de Estrogênio/química , Receptor beta de Estrogênio/química , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Metiltransferases/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA