Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Steroid Biochem Mol Biol ; 212: 105926, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34091027

RESUMO

The main physiological function of 17ß-estradiol (E2) in vertebrates is to regulate sexual development and reproduction. In fish, especially hermaphroditic fish, estrogen is often used to aid reproduction, but it also can trigger an inflammatory response. However, the molecular mechanism for this E2-induced inflammatory reaction is not clear. In this study, we found that the ERß-CXCL19/CXCR4-NFκB cascade regulated the E2-induced inflammatory response in the orange-spotted grouper (Epinephelus coioides). Strikingly, E2 treatment resulted in significantly high expression of inflammatory cytokines and induced phosphorylation and degradation of IκBα and translocation of NFκB subunit p65 to the nucleus in grouper spleen cells. However, the E2-induced inflammatory response could be prevented by the broad estrogen receptor (ER) ligand ICI 182,780. Moreover, the luciferase assay showed that E2 induced the inflammatory response by activating the promotor of chemokine CXCL19 through ERß1 and ERß2. Knockdown of CXCL19 blocked the E2-induced inflammatory response and NFκB nucleus translocation. Additionally, knockdown of chemokines CXCR4a and CXCR4b together, but not alone, blocked the E2-induced inflammatory response. The immunofluorescence assay and co-immunoprecipitation analysis showed that CXCL19 mediated the E2-induced inflammatory response by activating CXCR4a or CXCR4b. Taken together, these results showed that the ERß-CXCL19/CXCR4-NFκB pathway mediated the E2-induced inflammatory response in grouper. These findings are valuable for future comparative immunological studies and provide a theoretical basis for mitigating the adverse reactions that occur when using E2 to help fish reproduce.


Assuntos
Quimiocinas CXC/imunologia , Estradiol/farmacologia , Receptor beta de Estrogênio/imunologia , Estrogênios/farmacologia , Proteínas de Peixes/imunologia , Inflamação/induzido quimicamente , NF-kappa B/imunologia , Receptores CXCR4/imunologia , Animais , Quimiocinas CXC/genética , Citocinas/imunologia , Receptor beta de Estrogênio/genética , Proteínas de Peixes/genética , Células HEK293 , Humanos , Inflamação/imunologia , NF-kappa B/metabolismo , Perciformes , Receptores CXCR4/genética , Transdução de Sinais/efeitos dos fármacos , Baço/imunologia
2.
Anat Rec (Hoboken) ; 304(6): 1185-1193, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33856123

RESUMO

Estrogen is an important hormone for health in both genders. It is indispensable to glucose homeostasis, immune robustness, bone health, cardiovascular health, and neural functions. The main way that estrogen acts in the cells is through estrogen receptors (ERs). The presence of specific estrogen receptors is required for estrogen to have its characteristic ubiquitous action in almost all tissues. Estrogen receptor alpha (ERα) and estrogen receptor beta (ERß) are the major isoforms of estrogen that are highly specific in humans and enable selective hormonal actions in different tissues. This article reviews some of the observed estrogen actions and effects in different tissues and cells through these specific receptors. This ubiquitous, almost ordinary hormone may reveal itself as a significant factor that helped us to better understand the complexity of the human immune system response against respiratory infections, including the COVID-19, and especially in the current state of this painful pandemic.


Assuntos
COVID-19/imunologia , Receptor alfa de Estrogênio/imunologia , Receptor beta de Estrogênio/imunologia , Sistema Imunitário/imunologia , Sistema Respiratório/imunologia , SARS-CoV-2/imunologia , Animais , COVID-19/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Humanos , Sistema Imunitário/metabolismo , Mucosa Nasal/imunologia , Mucosa Nasal/metabolismo , Sistema Respiratório/metabolismo , SARS-CoV-2/metabolismo
3.
Int J Mol Sci ; 23(1)2021 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-35008514

RESUMO

High ERß/HER oncogenic signaling defines lung tumors with an aggressive biology. We previously showed that combining the anti-estrogen fulvestrant with the pan-HER inhibitor dacomitinib reduced ER/HER crosstalk and produced synergistic anti-tumor effects in immunocompromised lung cancer models, including KRAS mutant adenocarcinoma. How this combination affects the tumor microenvironment (TME) is not known. We evaluated the effects of fulvestrant and dacomitinib on murine bone marrow-derived macrophages (BMDMs) and CD8+ T cells, and tested the efficacy of the combination in vivo, using the KRAS mutant syngeneic lung adenocarcinoma model, FVBW-17. While this combination synergistically inhibited proliferation of FVBW-17 cells, it had unwanted effects on immune cells, by reducing CD8+ T cell activity and phagocytosis in BMDMs and inducing PD-1. The effects were largely attributed to dacomitinib, which caused downregulation of Src family kinases and Syk in immune cells. In a subcutaneous flank model, the combination induced an inflamed TME with increased myeloid cells and CD8+ T cells and enhanced PD-1 expression in the splenic compartment. Concomitant administration of anti-PD-1 antibody with fulvestrant and dacomitinib was more efficacious than fulvestrant plus dacomitinib alone. Administering anti-PD-1 sequentially after fulvestrant plus dacomitinib was synergistic, with a two-fold greater tumor inhibitory effect compared to concomitant therapy, in both the flank model and in a lung metastasis model. Sequential triple therapy has potential for treating lung cancer that shows limited response to current therapies, such as KRAS mutant lung adenocarcinoma.


Assuntos
Receptor beta de Estrogênio/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptor ErbB-2/genética , Microambiente Tumoral/genética , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Carcinogênese/genética , Carcinogênese/imunologia , Linhagem Celular Tumoral , Receptor beta de Estrogênio/imunologia , Feminino , Humanos , Imunoterapia/métodos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Oncogenes/genética , Oncogenes/imunologia , Receptor de Morte Celular Programada 1/genética , Proteínas Proto-Oncogênicas p21(ras)/imunologia , Quinazolinonas/farmacologia , Receptor ErbB-2/imunologia , Microambiente Tumoral/imunologia
4.
J Neuroinflammation ; 17(1): 317, 2020 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-33097048

RESUMO

The peri-menopause or menopausal transition-the time period that surrounds the final years of a woman's reproductive life-is associated with profound reproductive and hormonal changes in a woman's body and exponentially increases a woman's risk of cerebral ischemia and Alzheimer's disease. Although our understanding of the exact timeline or definition of peri-menopause is limited, it is clear that there are two stages to the peri-menopause. These are the early menopausal transition, where menstrual cycles are mostly regular, with relatively few interruptions, and the late transition, where amenorrhea becomes more prolonged and lasts for at least 60 days, up to the final menstrual period. Emerging evidence is showing that peri-menopause is pro-inflammatory and disrupts estrogen-regulated neurological systems. Estrogen is a master regulator that functions through a network of estrogen receptors subtypes alpha (ER-α) and beta (ER-ß). Estrogen receptor-beta has been shown to regulate a key component of the innate immune response known as the inflammasome, and it also is involved in regulation of neuronal mitochondrial function. This review will present an overview of the menopausal transition as an inflammatory event, with associated systemic and central nervous system inflammation, plus regulation of the innate immune response by ER-ß-mediated mechanisms.


Assuntos
Estrogênios/metabolismo , Imunidade Inata/fisiologia , Menopausa/metabolismo , Ciclo Menstrual/metabolismo , Doenças Neurodegenerativas/metabolismo , Perimenopausa/metabolismo , Receptor beta de Estrogênio/imunologia , Receptor beta de Estrogênio/metabolismo , Estrogênios/imunologia , Feminino , Humanos , Menopausa/imunologia , Ciclo Menstrual/imunologia , Doenças Neurodegenerativas/imunologia , Perimenopausa/imunologia
5.
Toxins (Basel) ; 12(3)2020 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-32235729

RESUMO

Nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) is commonly expressed in prostate cancer (PCa) cells and is associated with increased proliferation, metastases and androgen independence. Zearalenone (ZEA) is one of the most common mycotoxins contaminating food, which might mimic estrogens and bind to estrogen receptors (ERs). The ratio of androgens to estrogens in men decreases physiologically with age, and is believed to participate in prostate carcinogenesis. In this study, we evaluated the role of NFκB and ERß in the induction of oxidative stress in human PCa cells by ZEA. As observed, ZEA at a dose of 30 µM induces oxidative stress in PCa cells associated with DNA damage and G2/M cell cycle arrest. We also observed that the inhibition of ERß and NFΚB via specific inhibitors (PHTPP and BAY 117082) significantly increased ZEA-induced oxidative stress, although the mechanism seems to be different for androgen-dependent and androgen-independent cells. Based on our findings, it is possible that the activation of ERß and NFΚB in PCa might protect cancer cells from ZEA-induced oxidative stress. We therefore shed new light on the mechanism of ZEA toxicity in human cells.


Assuntos
Receptor beta de Estrogênio/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Zearalenona/toxicidade , Técnicas de Cultura de Células , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Receptor beta de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/imunologia , Humanos , Masculino , NF-kappa B/antagonistas & inibidores , NF-kappa B/imunologia , Estresse Oxidativo/imunologia , Células PC-3 , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo
6.
Immunopharmacol Immunotoxicol ; 42(1): 28-36, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31876196

RESUMO

Background: Epidemiological studies have shown that environmental factors accelerate the progress of primary Sjögren's syndrome (pSS). Bisphenol A (BPA), a classic endocrine disrupting chemical, affects the immune system. However, the impact of BPA on pSS has not yet been reported. The present study aimed to evaluate the potential relationship between BPA, estrogen receptor (ER), and pSS.Methods: We studied the impact of BPA on monocyte-derived dendritic cells (moDCs) from pSS patients and age-matched healthy controls (HCs). Morphological effects were observed under inverted microscope. Surface markers were analyzed by flow cytometry. ER and cytokine profiles were assessed using real-time polymerase chain reaction. The ability of moDCs to stimulate CD4+ T cells activation was assessed by mixed lymphocyte reaction (MLR).Results: moDCs from both pSS patients and HCs expressed ERα as well as ERß. After BPA-exposure, expression of ERα increased significantly in pSS patients, while that of ERß remained unchanged. moDCs from BPA-exposed pSS patients showed irregular morphology and reduction in cell aggregation. BPA increased HLA-DR on moDCs of pSS patients via ERα, and promoted the secretion of IL6 and IL12. When co-cultured with BPA-treated moDCs, cytokines (IFN-γ, IL4, IL17, IL10) and transcription factors (T-bet, Gata3, RoR-γt, Foxp3) of CD4+ T cells showed imbalance of Th1/Th2/Th17/Treg polarization, with Th1 and Th17 dominating.Conclusions: BPA altered the function of moDCs through ERα, including antigen capture, secretion of inflammatory factors, and ability to stimulate T cells, as well as accelerated the progression and further deterioration of pSS.


Assuntos
Compostos Benzidrílicos/toxicidade , Células Dendríticas/imunologia , Monócitos/imunologia , Fenóis/toxicidade , Síndrome de Sjogren/imunologia , Idoso , Citocinas/imunologia , Células Dendríticas/patologia , Receptor alfa de Estrogênio/imunologia , Receptor beta de Estrogênio/imunologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/patologia , Síndrome de Sjogren/patologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/patologia , Fatores de Transcrição/imunologia
7.
J Steroid Biochem Mol Biol ; 191: 105312, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30995525

RESUMO

ERbeta (ERß) celebrated its 20th birthday in 2016 and although the overwhelming data in the literature indicate a role for this receptor in the control of epithelial proliferation, neurodegeneration and immune function, no ERß agonists have yet made it to the clinics. This is the situation, despite the fact that very good safe ERß agonists have been synthesized and at least one has been donated to the NIH for distribution to researchers, who want to study its possible clinical use. Clinical trials are ongoing for the use of ERß agonists in prostate cancer and schizophrenia but even today reviewers of our grants still make comments like "The grant is excellent except that the focus of the grant is ERß". There are multiple reasons for the non-acceptance of the value of ERß and in this paper we will discuss issues raised by labs which do not support a role for ERß in physiology or pathology.


Assuntos
Receptor beta de Estrogênio/metabolismo , Animais , Anticorpos/imunologia , Descoberta de Drogas , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/imunologia , Expressão Gênica , Humanos , Imunidade , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo
8.
Brain Res ; 1712: 93-100, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30731078

RESUMO

The ovarian hormone 17ß-estradiol is known to regulate the release, expression and immunoreactivity of arginine-vasopressin (AVP) in the supraoptic and paraventricular hypothalamic nuclei of rodents. Previous studies have shown that estrogen receptor α is involved in the effects of chronic estradiol administration on arginine-vasopressin immunoreactivity in the female rat hypothalamus. In this study we have examined the effect of an acute administration of estradiol or specific agonists for estrogen receptors α, ß and G protein-coupled estrogen receptor 1 on the immunoreactivity of arginine-vasopressin in the hypothalamus of adult ovariectomized female rats. Acute estradiol administration resulted in a significant decrease in the number of arginine-vasopressin immunoreactive neurons in the supraoptic and paraventricular nuclei after 24 h. The effects of the specific estrogen receptors agonists suggest that the action of estradiol on arginine-vasopressin immunoreactivity is mediated in the supraoptic nucleus by G protein-coupled estrogen receptor 1 and in the paraventricular nucleus by both estrogen receptor ß and G protein-coupled estrogen receptor 1. Thus, in contrast to previous studies on the effect of chronic estrogenic treatments, the present findings suggest that estrogen receptor ß and G protein-coupled estrogen receptor 1 mediate the acute effects of estradiol on arginine-vasopressin immunoreactivity in the hypothalamus of ovariectomized rats.


Assuntos
Arginina Vasopressina/metabolismo , Receptor beta de Estrogênio/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Núcleo Supraóptico/metabolismo , Animais , Arginina Vasopressina/imunologia , Estradiol/farmacologia , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/imunologia , Feminino , Hipotálamo/imunologia , Hipotálamo/metabolismo , Neurônios/imunologia , Neurônios/metabolismo , Ovariectomia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/imunologia , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/imunologia , Núcleo Supraóptico/efeitos dos fármacos , Núcleo Supraóptico/imunologia
9.
Autoimmunity ; 51(4): 157-165, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-30022688

RESUMO

Oestrogen receptor beta (ERß) and epidermal growth factor receptor (EGFR) pathway can synergistically promote the proliferation, invasion, and metastasis of non-small-cell lung cancer (NSCLC) cells. ERß has five subtypes, and the selective splicing of exon 8 in ERß5 transcription translational phase makes its biological function different from other subtypes. The following study investigates whether ERß5 interacts with EGFR pathway in lung cancer. Briefly, we found that the overexpression of ERß5 and EGFR is associated with poor prognosis and decreased overall survival in NSCLC patients. Furthermore, the effects of ERß5 and EGFR on cell biological behaviour were investigated in vitro. These results indicated that the combination of ERß5 and EGF induces cell proliferation and invasion, while the combination of ERß5 and Gefitinib (EGFR inhibitors, Gef) induces cell apoptosis and promotes cell mitosis in A549 cell line. In addition, the combination of ERß5 and EGF increases the expression of ERß5, EGFR, and p-ERK1/2 in lung cancer cells. To sum up, the obtained results suggest that ERß5 and EGFR synergistically promote the progression of lung cancer by activating MEK/ERK signalling pathway, which provides a theoretical basis for more accurate combined targeted therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Receptor beta de Estrogênio/imunologia , Neoplasias Pulmonares/imunologia , Proteínas de Neoplasias/imunologia , Células A549 , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/imunologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Intervalo Livre de Doença , Receptores ErbB/agonistas , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Receptores ErbB/metabolismo , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Feminino , Gefitinibe/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Pessoa de Meia-Idade , Mitose/efeitos dos fármacos , Mitose/imunologia , Terapia de Alvo Molecular , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Taxa de Sobrevida
10.
Clin Immunol ; 190: 41-52, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29481981

RESUMO

Estrogens play important roles in autoimmune thyroiditis, but it remains unknown which estrogen receptor (ER) subtype mediates the stimulatory effects. Herein we treated ovariectomized mice with ERα or ERß selective agonist followed by thyroglobulin-immunization to induce experimental autoimmune thyroiditis (EAT), and observed the aggravation of EAT after diarylpropionitrile (DPN, ERß selective agonist) administration. The mRNA levels of interleukin(IL)-17A, IL-21 and RORγt and percentages of T helper (Th) 17 cells were up-regulated in the splenocytes of DPN-treated mice. Activated ERß was found directly binding to IL-17A and IL-21 gene promoters, and also indirectly promoting IL-21 and RORγt gene transcription through interaction with NF-κB. The expressions of co-stimulatory molecules were increased on antigen-presenting cells (APCs) after DPN administration. It suggests that ERß is the predominant ER subtype responsible for EAT development, and its activation may enhance Th17-type responses through genomic pathways and alteration of APCs' activities.


Assuntos
Receptor beta de Estrogênio/imunologia , Expressão Gênica/imunologia , Células Th17/imunologia , Tireoidite Autoimune/imunologia , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Interleucina-17/genética , Interleucina-17/imunologia , Interleucinas/genética , Interleucinas/imunologia , Interleucinas/metabolismo , Camundongos Endogâmicos CBA , Nitrilas/farmacologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Ovariectomia , Propionatos/farmacologia , Células Th17/efeitos dos fármacos , Células Th17/metabolismo , Tireoidite Autoimune/genética , Tireoidite Autoimune/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
11.
J Gastroenterol Hepatol ; 33(3): 747-755, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28884481

RESUMO

BACKGROUND AND AIM: The aim of this study is to explore the roles of estrogen receptor (ER) subtypes and corresponding agonists/antagonists on the development of cirrhosis and activation and proliferation of hepatic stellate cells (HSCs). METHODS: Carbon tetrachloride (CCl4 )-induced cirrhotic ovariectomized rats were administered non-selective ER agonist (ß-estradiol, E2), ER selective agonists (ERα agonist, propylpyrazoletriol; ERß agonist, diarylpropionitrile [DPN]; and G-protein-coupled ER [GPER] agonist, G1), or E2 + ER selective antagonists (ERα antagonist, MPP; ERß antagonist, PHTPP; and GPER antagonist, G15) for 12 weeks. The expression of the three ER subtypes in livers and HSCs and the effects of the drugs on hepatic fibrosis, isolated HSCs, and uteri were evaluated. RESULTS: Selective ER agonists/antagonists had various effects on CCl4 -induced cirrhosis. The cirrhotic rats in the CCl4  + E2, CCl4  + DPN, CCl4  + E2 + MPP, and CCl4  + E2 + G15 groups presented reduced fibrosis scores, compared with those in the CCl4 group. The cirrhotic rats in the E2 + PHTPP group presented increased fibrosis scores that similar to those in the CCl4 group. The ovariectomized rats had enlarged uteri with increased uterus indexes after E2 administration; however, the proliferative effects of E2 were partially blocked by MPP or G15, but not PHTPP. In the in vitro study, DPN attenuated the transformation of quiescent HSCs to activated phenotype, suppressed collagen I, and α-smooth muscle actin expression. DPN also suppressed platelet-derived growth factor-induced proliferation in cultured HSCs, which was reversed by PHTPP. CONCLUSIONS: The antifibrogenic effects of estrogen were mediated by ERß but not ERα or GPER. The ERß selective agonist exerted a fibrosuppressive effect by inhibiting the activation and proliferation of HSCs, but did not induce uterine hyperplasia.


Assuntos
Proliferação de Células/efeitos dos fármacos , Receptor beta de Estrogênio/agonistas , Células Estreladas do Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/patologia , Nitrilas/uso terapêutico , Fenóis/uso terapêutico , Propionatos/uso terapêutico , Pirazóis/uso terapêutico , Animais , Células Cultivadas , Depressão Química , Estradiol/farmacologia , Estradiol/uso terapêutico , Receptor beta de Estrogênio/imunologia , Feminino , Fibrose , Nitrilas/farmacologia , Fenóis/farmacologia , Propionatos/farmacologia , Pirazóis/farmacologia , Ratos Sprague-Dawley
12.
Mol Cell Endocrinol ; 440: 138-150, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27889472

RESUMO

Estrogen Receptor-ß (ERß) has been implicated in many cancers. In prostate and breast cancer its function is controversial, but genetic studies implicate a role in cancer progression. Much of the confusion around ERß stems from antibodies that are inadequately validated, yet have become standard tools for deciphering its role. Using an ERß-inducible cell system we assessed commonly utilized ERß antibodies and show that one of the most commonly used antibodies, NCL-ER-BETA, is non-specific for ERß. Other antibodies have limited ERß specificity or are only specific in one experimental modality. ERß is commonly studied in MCF-7 (breast) and LNCaP (prostate) cancer cell lines, but we found no ERß expression in either, using validated antibodies and independent mass spectrometry-based approaches. Our findings question conclusions made about ERß using the NCL-ER-BETA antibody, or LNCaP and MCF-7 cell lines. We describe robust reagents, which detect ERß across multiple experimental approaches and in clinical samples.


Assuntos
Anticorpos Antineoplásicos/farmacologia , Receptor beta de Estrogênio/imunologia , Mama/efeitos dos fármacos , Mama/metabolismo , Linhagem Celular Tumoral , Doxiciclina/farmacologia , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Indicadores e Reagentes , Masculino , Peptídeos , Próstata/efeitos dos fármacos , Próstata/metabolismo , Reprodutibilidade dos Testes
13.
Endocrinology ; 157(7): 2844-52, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27105387

RESUMO

Estrogens regulate normal sexual and reproductive development in females. Their actions are mediated mainly by estrogen receptor (ER)α and ERß. Understanding the function of ERs necessitates knowing their cellular location and protein partners, which, in turn, requires reliable and specific antibodies. Several antibodies are available for ERα; however, discrepancies in immunoreactivity have been reported for ERß. Here, we have developed antisera for mouse ERß (mERß) using a specific C-terminal 18-amino acid peptide conjugated to mariculture keyhole limpet hemocyanin. Sprague Dawley rats were immunized, and the resulting antisera were characterized by Western blot analysis of nuclear extracts from tissues of wild-type (WT) mice, and mice genetically modified to lack either ERα (CERαKO) or ERß (CERßKO). An approximately 56-kDa protein was detected in the hypothalamus, uterus, ovary, mammary gland, testes, and epididymis of WT mice, consistent with the predicted molecular size of ERß. In addition, the same protein band was identified in in vitro synthesized mERß protein and in the mammary glands of CERαKO mice. The approximately 56-kDa protein was not observed in in vitro synthesized mERα protein or in any tissue examined in the CERßKO mice. Immunohistochemistry using the antisera revealed ERß staining in the granulosa cells of WT ovaries and in the mediobasal hypothalamus, paraventricular nucleus, and cerebral cortex in the WT adult mouse brain. These data suggest that the novel rat anti-mERß sera are specific to ERß to allow investigators to explore to cellular and physiological role of ERß in the brain and other mouse tissues.


Assuntos
Receptor beta de Estrogênio/imunologia , Soros Imunes , Animais , Epididimo/metabolismo , Feminino , Hipotálamo/metabolismo , Masculino , Glândulas Mamárias Animais/metabolismo , Camundongos , Ovário/metabolismo , Ratos , Ratos Sprague-Dawley , Testículo/metabolismo , Útero/metabolismo
14.
LGBT Health ; 2(1): 81-3, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26790022

RESUMO

Estrogen exposure has been linked to a risk of testicular germ cell cancers, although predominantly in animal studies. In the human testis, estrogen receptor beta (ER-ß) mediates the physiological action of estrogen. We describe a case of a 30-year-old transgender woman (male-to-female [MtF]) with gender dysphoria who developed a mature testicular teratoma with positive estrogen receptor beta expression while receiving cross-sex hormonal therapy with estradiol and progesterone. Although the presence of hormone receptors does not always imply that hormones have played a role in the development of the tumor, a contribution of exposure to exogenous estrogen in this case cannot be ruled out.


Assuntos
Receptor beta de Estrogênio/imunologia , Teratoma/cirurgia , Neoplasias Testiculares/cirurgia , Pessoas Transgênero , Adulto , Estradiol/uso terapêutico , Estrogênios/metabolismo , Feminino , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/etiologia , Progesterona/uso terapêutico , Neoplasias Testiculares/etiologia , Testículo/metabolismo , Testículo/cirurgia
15.
J Invest Dermatol ; 134(9): 2447-2457, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24769859

RESUMO

Efficient local monocyte/macrophage recruitment is critical for tissue repair. Recruited macrophages are polarized toward classical (proinflammatory) or alternative (prohealing) activation in response to cytokines, with tight temporal regulation crucial for efficient wound repair. Estrogen acts as a potent anti-inflammatory regulator of cutaneous healing. However, an understanding of estrogen/estrogen receptor (ER) contribution to macrophage polarization and subsequent local effects on wound healing is lacking. Here we identify, to our knowledge previously unreported, a role whereby estrogen receptor α (ERα) signaling preferentially polarizes macrophages from a range of sources to an alternative phenotype. Cell-specific ER ablation studies confirm an in vivo role for inflammatory cell ERα, but not ERß, in poor healing associated with an altered cytokine profile and fewer alternatively activated macrophages. Furthermore, we reveal intrinsic changes in ERα-deficient macrophages, which are unable to respond to alternative activation signals in vitro. Collectively, our data reveal that inflammatory cell-expressed ERα promotes alternative macrophage polarization, which is beneficial for timely healing. Given the diverse physiological roles of ERs, these findings will likely be of relevance to many pathologies involving excessive inflammation.


Assuntos
Receptor alfa de Estrogênio/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos/imunologia , Transdução de Sinais/imunologia , Cicatrização/imunologia , Animais , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/imunologia , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Estrogênios/farmacologia , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/metabolismo , Ovariectomia , Transdução de Sinais/efeitos dos fármacos
16.
PLoS One ; 9(2): e88540, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551115

RESUMO

The innate immune system including microglia has a major contribution to maintenance of the physiological functions of the hippocampus by permanent monitoring of the neural milieu and elimination of tissue-damaging threats. The hippocampus is vulnerable to age-related changes ranging from gene expression to network connectivity. The risk of hippocampal deterioration increases with the decline of gonadal hormone supply. To explore the impact of hormone milieu on the function of the innate immune system in middle-aged female rats, we compared mRNA expression in the hippocampus after gonadal hormone withdrawal, with or without subsequent estrogen replacement using estradiol and isotype-selective estrogen receptor (ER) agonists. Targeted profiling assessed the status of the innate immune system (macrophage-associated receptors, complement, inhibitory neuronal ligands), local estradiol synthesis (P450 aromatase) and estrogen reception (ER). Results established upregulation of macrophage-associated (Cd45, Iba1, Cd68, Cd11b, Cd18, Fcgr1a, Fcgr2b) and complement (C3, factor B, properdin) genes in response to ovariectomy. Ovariectomy upregulated Cd22 and downregulated semaphorin3A (Sema3a) expression, indicating altered neuronal regulation of microglia. Ovariectomy also led to downregulation of aromatase and upregulation of ERα gene. Of note, analogous changes were observed in the hippocampus of postmenopausal women. In ovariectomized rats, estradiol replacement attenuated Iba1, Cd11b, Fcgr1a, C3, increased mannose receptor Mrc1, Cd163 and reversed Sema3a expression. In contrast, reduced expression of aromatase was not reversed by estradiol. While the effects of ERα agonist closely resembled those of estradiol, ERß agonist was also capable of attenuating the expression of several macrophage-associated and complement genes. These data together indicate that the innate immune system of the aging hippocampus is highly responsive to the gonadal hormone milieu. In ovariectomized female rats, estradiol replacement exerts potent immunomodulatory effects including attenuation of microglia sensitization, initiation of M2-like activation and modulation of complement expression by targeting hippocampal neurons and glial cells through ERα and ERß.


Assuntos
Envelhecimento/metabolismo , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Hipocampo/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Ovariectomia , Envelhecimento/genética , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Aromatase/genética , Aromatase/imunologia , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/imunologia , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/imunologia , Terapia de Reposição de Estrogênios , Feminino , Expressão Gênica , Hipocampo/citologia , Hipocampo/imunologia , Humanos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Microglia/citologia , Microglia/efeitos dos fármacos , Pessoa de Meia-Idade , Nitrilas/farmacologia , Pós-Menopausa , Propionatos/farmacologia , Ratos , Receptores de IgG/genética , Receptores de IgG/imunologia , Semaforina-3A/genética , Semaforina-3A/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia
17.
Antibiot Khimioter ; 57(5-6): 11-7, 2012.
Artigo em Russo | MEDLINE | ID: mdl-23156039

RESUMO

Comparability of the level and intensity of estrogen receptors beta (ERbeta) expression in non-small cell lung cancer tissue of 32 patients was analyzed by flow cytometry using various antibodies--to the total fraction of ERbeta (clone 14C8) as well to the full-length ERbeta1 isoform (clone EMRO2). The differences in the ER expression indexes detected by anti-ERbeta or anti-ERbeta1 antibodies were revealed in some patients, but it had no influence on average indexes of the ERbeta expression in the patient groups investigated. It was confirmed by the findings on more frequent and more intensive expression of ERbeta in the non-small cell lung cancer tissue of female patients vs. the males irrespective of antibody type - anti-ER/ or anti-ERbeta1. Therefore, in comparative analysis of ERbeta expression in the groups of the patients with different clinicomorphologic characteristics of the disease it is possible to use both the antibodies. For individual disease prognosis in the routine clinical practice it is recommended to use the antibodies to the total fraction of ERbeta, since there are individual differences between the ERbeta expression indexes revealing by various types of antibodies.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Receptor beta de Estrogênio/metabolismo , Neoplasias Pulmonares/metabolismo , Anticorpos Monoclonais Murinos/imunologia , Biomarcadores Tumorais/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Receptor beta de Estrogênio/imunologia , Feminino , Citometria de Fluxo , Humanos , Neoplasias Pulmonares/imunologia , Células MCF-7 , Masculino , Prognóstico , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Fatores Sexuais
18.
J Biol Chem ; 287(48): 40140-9, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22908233

RESUMO

BACKGROUND: Hepatocarcinoma cancer (HCC) occurs more often in men than in women, and little is known about its underlying molecular mechanisms. RESULTS: We identify that 17ß-estradiol (E2) could suppress tumor growth via regulating the polarization of macrophages. CONCLUSION: Estrogen functions as a suppressor for macrophage alternative activation. SIGNIFICANCE: These studies introduce a novel mechanism for suppressing male-predominant HCC. Hepatocarcinoma cancer (HCC), one of the most malignant cancers, occurs significantly more often in men than in women; however, little is known about its underlying molecular mechanisms. Here we identified that 17ß-estradiol (E2) could suppress tumor growth via regulating the polarization of macrophages. We showed that E2 re-administration reduced tumor growth in orthotopic and ectopic mice HCC models. E2 functioned as a suppressor for macrophage alternative activation and tumor progression by keeping estrogen receptor ß (ERß) away from interacting with ATP5J (also known as ATPase-coupling factor 6), a part of ATPase, thus inhibiting the JAK1-STAT6 signaling pathway. These studies introduce a novel mechanism for suppressing male-predominant HCC.


Assuntos
Carcinoma Hepatocelular/imunologia , Regulação para Baixo , Estrogênios/imunologia , Neoplasias Hepáticas/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/fisiopatologia , Linhagem Celular , Proliferação de Células , Células Cultivadas , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/imunologia , Feminino , Humanos , Janus Quinase 1/genética , Janus Quinase 1/imunologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/fisiopatologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/imunologia , Transdução de Sinais
19.
Mol Cell Endocrinol ; 358(1): 27-35, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22370157

RESUMO

Multiple phosphorylation sites on the human estrogen receptor (hER)α were identified and shown to influence mammary carcinogenesis. In contrast, functional phosphorylation sites of hERß have yet to be experimentally identified and validated. Here, using mass spectrometry, we uncovered three serines (S75, S87, and S105) in the N-terminus of hERß as targets of ERK1/2 and p38 kinases. We raised a specific antibody against phosphorylated S105 (pS105) and demonstrated that this site was endogenously phosphorylated in MDA-MB-231 and BT-474 cells. A phospho-mimetic mutant generated from hERß1 was found to exhibit higher transactivation activity than hERß1. Ectopic expression of this mutant inhibited cell migration and invasion, but did not affect cell growth and cell-cycle progression in these cell models. In breast cancer specimens, pS105-hERß immunoreactivity was detected with a higher prevalence and intensity than that of hERß1. These results underscore the functional importance of the first experimentally identified hERß-phosphorylation site in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Receptor beta de Estrogênio/química , Receptor beta de Estrogênio/metabolismo , Anticorpos/imunologia , Neoplasias da Mama/genética , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Receptor beta de Estrogênio/imunologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Fosforilação , Processamento de Proteína Pós-Traducional , Ativação Transcricional/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
J Cell Biochem ; 113(2): 711-23, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22095450

RESUMO

The role of estrogen receptor alpha (ERα) in breast cancer has been studied extensively, and its protein expression is prognostic and a primary determinant of endocrine sensitivity. However, much less is known about the role of ERß and its relevance remains unclear due to the publication of conflicting reports. Here, we provide evidence that much of this controversy may be explained by variability in antibody sensitivity and specificity and describe the development, characterization, and potential applications of a novel monoclonal antibody targeting full-length human ERß and its splice variant forms. Specifically, we demonstrate that a number of commercially available ERß antibodies are insensitive for ERß and exhibit significant cross-reaction with ERα. However, our newly developed MC10 ERß antibody is shown to be highly specific and sensitive for detection of full-length ERß and its variant forms. Strong and variable staining patterns for endogenous levels of ERß protein were detected in normal human tissues and breast tumors using the MC10 antibody. Importantly, ERß was shown to be expressed in a limited cohort of both ERα positive and ERα negative breast tumors. Taken together, these data demonstrate that the use of poorly validated ERß antibodies is likely to explain much of the controversy in the field with regard to the biological relevance of ERß in breast cancer. The use of the MC10 antibody, in combination with highly specific antibodies targeting only full-length ERß, is likely to provide additional discriminatory features in breast cancers that may be useful in predicting response to therapy.


Assuntos
Anticorpos Monoclonais Murinos/biossíntese , Biomarcadores Tumorais/imunologia , Neoplasias da Mama/metabolismo , Receptor beta de Estrogênio/imunologia , Animais , Especificidade de Anticorpos , Biomarcadores Tumorais/metabolismo , Mama/metabolismo , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/patologia , Linhagem Celular , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Masculino , Camundongos , Especificidade de Órgãos , Próstata/metabolismo , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...