Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
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
2.
Nat Commun ; 11(1): 6175, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33268821

RESUMO

To elucidate the effects of neoadjuvant chemotherapy (NAC), we conduct whole transcriptome profiling coupled with histopathology analyses of a longitudinal breast cancer cohort of 146 patients including 110 pairs of serial tumor biopsies collected before treatment, after the first cycle of treatment and at the time of surgery. Here, we show that cytotoxic chemotherapies induce dynamic changes in the tumor immune microenvironment that vary by subtype and pathologic response. Just one cycle of treatment induces an immune stimulatory microenvironment harboring more tumor infiltrating lymphocytes (TILs) and up-regulation of inflammatory signatures predictive of response to anti-PD1 therapies while residual tumors are immune suppressed at end-of-treatment compared to the baseline. Increases in TILs and CD8+ T cell proportions in response to NAC are independently associated with pathologic complete response. Further, on-treatment immune response is more predictive of treatment outcome than immune features in paired baseline samples although these are strongly correlated.


Assuntos
Antígeno B7-H1/genética , Neoplasias da Mama/tratamento farmacológico , Carcinoma Ductal de Mama/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Terapia Neoadjuvante/métodos , Antraciclinas/uso terapêutico , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Neoplasias da Mama/mortalidade , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/imunologia , Carcinoma Ductal de Mama/mortalidade , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/imunologia , Ciclofosfamida/uso terapêutico , Intervalo Livre de Doença , Docetaxel/uso terapêutico , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/imunologia , Feminino , Perfilação da Expressão Gênica , Humanos , Imunidade Inata , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Estudos Longitudinais , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Neoplasia Residual , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Trastuzumab/uso terapêutico , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
3.
Front Immunol ; 11: 582214, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33240270

RESUMO

The dramatic female sex bias observed in human lupus is thought to be due, at least in part, to estrogens. Using mouse models, we have shown that estrogens, acting through estrogen receptor alpha (ERα) promote lupus development and contribute significantly to the female sex bias observed in this disease. C57Bl/6 (B6) mice carrying the lupus susceptibility locus Sle1 locus exhibit immune cell hyperactivation and loss of tolerance, and the action of Sle1 displays a strong female sex bias. Previously, we showed that disruption of ERα completely eliminates the female sex bias in the effects of Sle1. Here we report that ERα signaling selectively modulates the action of Sle1b, one of the three subloci that together constitute Sle1. We observed that disruption of ERα signaling attenuated T cell hyperactivation, formation of spontaneous germinal centers, loss of tolerance, and the development of anti-chromatin autoantibodies in B6.Sle1b female mice, but had no impact on these phenotypes in B6.Sle1b male mice. In fact, disruption of ERα completely abolished the female sex bias that is seen in each of these phenotypes in B6.Sle1b mice. Strikingly, Sle1b-induced B cell hyperactivation, a female sex-specific manifestation of Sle1b, was completely abrogated by disruption of ERα in B6.Sle1b females. Altogether, these results demonstrate that ERα signaling is responsible for the female sex bias in the actions of Sle1b, and is absolutely required for the female-specific B cell hyperactivation phenotype associated with this lupus susceptibility locus. By contrast, we found that ERα signaling had no impact on Sle1a, the other Sle1 sublocus that exerts effects that show a female sex bias.


Assuntos
Receptor alfa de Estrogênio/imunologia , Tolerância Imunológica/imunologia , Nefrite Lúpica/imunologia , Ativação Linfocitária/imunologia , Transdução de Sinais/imunologia , Animais , Linfócitos B/imunologia , Cromatina/genética , Cromatina/imunologia , Receptor alfa de Estrogênio/genética , Feminino , Predisposição Genética para Doença/genética , Centro Germinativo/imunologia , Tolerância Imunológica/genética , Nefrite Lúpica/genética , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Transdução de Sinais/genética
4.
Immunogenetics ; 72(8): 413-422, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33063129

RESUMO

It is well known that the estrogen receptor alpha gene (ESR1) affects the reproductive traits of pigs; however, the immune role of ESR1 gene has not yet been resolved. Here, we characterized the pleiotropic aspects of ESR1 gene in immunity using the pig model. Tissue expression profile showed that the ESR1 gene had a broad ectopic expression in multiple reproductive and immune-related tissues/organs, which provided the tissue-level spatial fundamental of ESR1 gene that might function as a pleiotropic immune regulator. Using the peripheral blood cell model, a coupling transcriptome analytical strategy was proposed and verified that there existed strong positive or negative correlations of ESR1 gene with hundreds of differentially expressed genes that were involved in the immune regulation, indicating that the ESR1 gene might affect or be affected by, directly or indirectly, dozens of immune-related genes in the peripheral blood cells. Furthermore, the results of genetic association analysis showed that the SmaI-polymorphism of ESR1 gene had significant or highly significant associations with multiple immune traits, including platelet (PLT), hematocrit (HCT), the number of CD4-CD8-CD3- cells, plateletcrit (PCT), mean corpuscular volume (MCV), and mean corpuscular hemoglobin concentration (MCHC). Multiple evidences supported the immune pleiotropic roles of ESR1 gene in pigs. The study advances our understanding of the cross-species immune pleiotropic landscape of ESR1 gene and also provides a potential pleiotropic molecular marker for disease-resistant breeding in pigs.


Assuntos
Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/imunologia , Regulação da Expressão Gênica , Estudos de Associação Genética/veterinária , Polimorfismo Genético , Suínos/genética , Suínos/imunologia , Animais , Feminino , Genótipo , Masculino , Fenótipo , Transcriptoma
5.
J Nutr Biochem ; 83: 108438, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32563803

RESUMO

Soy isoflavones (SIFs) are selective estrogen receptor modulators (SERMs) that have anti-inflammatory activities. Our previous study found that estrogen receptor α (ERα) directly regulates the NLRP3 transcription and NLRP3 inflammasome assembly. Therefore, we hypothesized that SIFs alleviate colitis via an ERα-dependent mechanism by targeting the NLRP3 inflammasome. The influence of SIFs on colitis and the potential mechanisms were thoroughly determined in this study. The results suggested that SIFs ameliorated dextran sodium sulfate (DSS)-induced body weight loss, reduced disease activity index and promoted the recovery of colon pathological damage in mice. Moreover, expression of the NLRP3 inflammasome was significantly inhibited, and the release of IL-1ß and IL-18 was suppressed by SIFs. Furthermore, ERα blockade ameliorated DSS-induced inflammatory responses in the intestine, and SIFs markedly suppressed the expression of ERα in a dose-dependent manner. Our study demonstrated that the protective therapeutic action of SIFs on DSS-induced colitis depended on inhibition of ERα and subsequent NLRP3 inflammasome activation, and SIFs are promising therapeutic agents for the treatment of colitis.


Assuntos
Anti-Inflamatórios/administração & dosagem , Colite/tratamento farmacológico , Receptor alfa de Estrogênio/imunologia , Inflamassomos/efeitos dos fármacos , Isoflavonas/administração & dosagem , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Extratos Vegetais/administração & dosagem , Animais , Colite/genética , Colite/imunologia , Receptor alfa de Estrogênio/genética , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Glycine max/química
6.
Bioorg Med Chem Lett ; 30(4): 126907, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31902710

RESUMO

Chimeric molecules which effect intracellular degradation of target proteins via E3 ligase-mediated ubiquitination (e.g., PROTACs) are currently of high interest in medicinal chemistry. However, these entities are relatively large compounds that often possess molecular characteristics which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. Accordingly, we explored whether conjugation of chimeric degraders to monoclonal antibodies using technologies originally developed for cytotoxic payloads might provide alternate delivery options for these novel agents. In this report we describe the construction of several degrader-antibody conjugates comprised of two distinct ERα-targeting degrader entities and three independent ADC linker modalities. We subsequently demonstrate the antigen-dependent delivery to MCF7-neo/HER2 cells of the degrader payloads that are incorporated into these conjugates. We also provide evidence for efficient intracellular degrader release from one of the employed linkers. In addition, preliminary data are described which suggest that reasonably favorable in vivo stability properties are associated with the linkers utilized to construct the degrader conjugates.


Assuntos
Anticorpos Monoclonais/imunologia , Portadores de Fármacos/química , Receptor alfa de Estrogênio/imunologia , Anticorpos Monoclonais/química , Antineoplásicos/química , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Desenho de Fármacos , Receptor alfa de Estrogênio/metabolismo , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoconjugados/farmacologia , Células MCF-7 , Proteólise/efeitos dos fármacos , Receptor ErbB-2/metabolismo
7.
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
8.
Cells ; 8(7)2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31331091

RESUMO

Tamoxifen resistance is a major hurdle in the treatment of estrogen receptor (ER)-positive breast cancer. The mechanisms of tamoxifen resistance are not fully understood although several underlying molecular events have been suggested. Recently, we identified autoantibodies reacting with membrane-associated ERα (anti-ERα Abs) in sera of breast cancer patients, able to promote tumor growth. Here, we investigated whether anti-ERα Abs purified from sera of ER-positive breast cancer patients could contribute to tamoxifen resistance. Anti-ERα Abs inhibited tamoxifen-mediated effects on cell cycle and proliferation in MCF-7 cells. Moreover, anti-ERα Abs hampered the tamoxifen-mediated reduction of tumor growth in SCID mice xenografted with breast tumor. Notably, simvastatin-mediated disaggregation of lipid rafts, where membrane-associated ERα is embedded, restored tamoxifen sensitivity, preventing anti-ERα Abs effects. In conclusion, detection of serum anti-ERα Abs may help predict tamoxifen resistance and concur to appropriately inform therapeutic decisions concerning hormone therapy in ER-positive breast cancer patients.


Assuntos
Antineoplásicos Hormonais/imunologia , Autoanticorpos/sangue , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/imunologia , Receptor alfa de Estrogênio/imunologia , Tamoxifeno/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos Hormonais/uso terapêutico , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Ensaios Antitumorais Modelo de Xenoenxerto
9.
PLoS One ; 14(4): e0215340, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30970003

RESUMO

Estrogen Receptor alpha (ERα) plays a major role in most breast cancers, and it is the target of endocrine therapies used in the clinic as standard of care for women with breast cancer expressing this receptor. The two methods ChIP-seq (chromatin immunoprecipitation coupled with deep sequencing) and RIME (Rapid Immunoprecipitation of Endogenous Proteins) have greatly improved our understanding of ERα function during breast cancer progression and in response to anti-estrogens. A critical component of both ChIP-seq and RIME protocols is the antibody that is used against the bait protein. To date, most of the ChIP-seq and RIME experiments for the study of ERα have been performed using the sc-543 antibody from Santa Cruz Biotechnology. However, this antibody has been discontinued, thereby severely impacting the study of ERα in normal physiology as well as diseases such as breast cancer and ovarian cancer. Here, we compare the sc-543 antibody with other commercially available antibodies, and we show that 06-935 (EMD Millipore) and ab3575 (Abcam) antibodies can successfully replace the sc-543 antibody for ChIP-seq and RIME experiments.


Assuntos
Anticorpos , Sequenciamento de Cromatina por Imunoprecipitação/métodos , Receptor alfa de Estrogênio/imunologia , Imunoprecipitação/métodos , Especificidade de Anticorpos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Células MCF-7
10.
Menopause ; 25(11): 1201-1207, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30358714

RESUMO

OBJECTIVE: The objective of this study was to determine whether estrogen could be formed locally in the coronary arteries. DESIGN: Coronary arteries were examined from monkeys (Macaca fascicularis, one male and one female) and human subjects (one premenopausal woman, one postmenopausal woman, and one man) by immunocytochemistry, using purified antisera against human placental estrogen synthetase (aromatase) and ER α. The arteries were graded for the amount of atherosclerosis. RESULTS: There was clear immunopositivity for both aromatase and estrogen receptors in all arteries studied. Although all endothelial cells (CD31 positive) stained for both antigens, the staining in macrophages, fibroblasts, and smooth muscle cells was irregular. CONCLUSION: The present results provide the first evidence for the local formation of estrogen in the coronary arteries. In addition to complementing the evidence of a cardioprotective effect of estrogen on the coronary circulation, our results highlight the potential importance of local regulation of estrogen formation and the role of available precursor androgens in maintaining the cardiovascular system.


Assuntos
Aromatase/metabolismo , Aterosclerose/metabolismo , Vasos Coronários/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/biossíntese , Adulto , Idoso de 80 Anos ou mais , Animais , Aromatase/imunologia , Aterosclerose/patologia , Autopsia , Vasos Coronários/imunologia , Vasos Coronários/patologia , Dieta Aterogênica , Estradiol/biossíntese , Receptor alfa de Estrogênio/imunologia , Feminino , Humanos , Imuno-Histoquímica , Macaca fascicularis , Masculino , Microscopia , Pessoa de Meia-Idade , Projetos Piloto , Pós-Menopausa/fisiologia , Pré-Menopausa/fisiologia
11.
Breast Cancer Res ; 20(1): 50, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29898754

RESUMO

BACKGROUND: Obesity and type II diabetes are linked to increased breast cancer risk in postmenopausal women. Patients treated with the antidiabetic drug metformin for diabetes or metabolic syndrome have reduced breast cancer risk, a greater pathologic complete response to neoadjuvant therapy, and improved breast cancer survival. We hypothesized that metformin may be especially effective when targeted to the menopausal transition, as this is a lifecycle window when weight gain and metabolic syndrome increase, and is also when the risk for obesity-related breast cancer increases. METHODS: Here, we used an 1-methyl-1-nitrosourea (MNU)-induced mammary tumor rat model of estrogen receptor (ER)-positive postmenopausal breast cancer to evaluate the long-term effects of metformin administration on metabolic and tumor endpoints. In this model, ovariectomy (OVX) induces rapid weight gain, and an impaired whole-body response to excess calories contributes to increased tumor glucose uptake and increased tumor proliferation. Metformin treatment was initiated in tumor-bearing animals immediately prior to OVX and maintained for the duration of the study. RESULTS: Metformin decreased the size of existing mammary tumors and inhibited new tumor formation without changing body weight or adiposity. Decreased lipid accumulation in the livers of metformin-treated animals supports the ability of metformin to improve overall metabolic health. We also found a decrease in the number of aromatase-positive, CD68-positive macrophages within the tumor microenvironment, suggesting that metformin targets the immune microenvironment in addition to improving whole-body metabolism. CONCLUSIONS: These findings suggest that peri-menopause/menopause represents a unique window of time during which metformin may be highly effective in women with established, or at high risk for developing, breast cancer.


Assuntos
Aromatase/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias Mamárias Animais/tratamento farmacológico , Metformina/administração & dosagem , Animais , Mama/efeitos dos fármacos , Mama/imunologia , Mama/patologia , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Progressão da Doença , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Mamárias Animais/induzido quimicamente , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Metilnitrosoureia/toxicidade , Ovariectomia , Pós-Menopausa/efeitos dos fármacos , Pós-Menopausa/genética , Pós-Menopausa/imunologia , Ratos , Células Estromais/efeitos dos fármacos , Células Estromais/enzimologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
12.
Mol Cancer Res ; 16(10): 1454-1457, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29934327

RESUMO

Inhibitors of the cyclin-dependent kinases 4 and 6 (CDK4/6) were originally designed to block proliferation and cell cycle progression of cancer cells in which the activity of these kinases is dysregulated. CDK4/6 inhibitors have already been FDA approved for the treatment of estrogen receptor (ER)-positive breast cancer and are being tested in numerous other cancer types. However, several recent studies have identified novel effects of CDK4/6 inhibitors on tumor growth, most notably an indirect effect resulting from the activation of immune surveillance. This Perspective discusses these recent observations, including the effects that CDK4/6 inhibitors may have on immune cells themselves. It is likely that CDK4/6 inhibitors will have a broader impact than their expected induction of cell cycle arrest in the treatment of human cancers. Mol Cancer Res; 16(10); 1454-7. ©2018 AACR.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Quinase 4 Dependente de Ciclina/imunologia , Quinase 6 Dependente de Ciclina/imunologia , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Neoplasias da Mama/imunologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Receptor alfa de Estrogênio/imunologia , Feminino , Humanos , Células MCF-7 , Camundongos , Inibidores de Proteínas Quinases/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Front Immunol ; 9: 1033, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29881378

RESUMO

Estradiol-based therapies predispose women to vaginal infections. Moreover, it has long been known that neutrophils are absent from the vaginal lumen during the ovulatory phase (high estradiol). However, the mechanisms that regulate neutrophil influx to the vagina remain unknown. We investigated the neutrophil transepithelial migration (TEM) into the vaginal lumen. We revealed that estradiol reduces the CD44 and CD47 epithelial expression in the vaginal ectocervix and fornix, which retain neutrophils at the apical epithelium through the estradiol receptor-alpha. In contrast, luteal progesterone increases epithelial expression of CD44 and CD47 to promote neutrophil migration into the vaginal lumen and Candida albicans destruction. Distinctive to vaginal mucosa, neutrophil infiltration is contingent to sex hormones to prevent sperm from neutrophil attack; although it may compromise immunity during ovulation. Thus, sex hormones orchestrate tolerance and immunity in the vaginal lumen by regulating neutrophil TEM.


Assuntos
Candidíase Vulvovaginal/imunologia , Receptor alfa de Estrogênio/genética , Infiltração de Neutrófilos , Neutrófilos/imunologia , Migração Transendotelial e Transepitelial , Vagina/imunologia , Animais , Antígeno CD47/genética , Antígeno CD47/imunologia , Candida albicans , Células Cultivadas , Colo do Útero/imunologia , Colo do Útero/microbiologia , Estradiol/farmacologia , Receptor alfa de Estrogênio/imunologia , Feminino , Hormônios Esteroides Gonadais/farmacologia , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Progesterona/farmacologia , Vagina/microbiologia
14.
Nat Commun ; 9(1): 1725, 2018 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-29713003

RESUMO

Breast cancer (BC) in the Asia Pacific regions is enriched in younger patients and rapidly rising in incidence yet its molecular bases remain poorly characterized. Here we analyze the whole exomes and transcriptomes of 187 primary tumors from a Korean BC cohort (SMC) enriched in pre-menopausal patients and perform systematic comparison with a primarily Caucasian and post-menopausal BC cohort (TCGA). SMC harbors higher proportions of HER2+ and Luminal B subtypes, lower proportion of Luminal A with decreased ESR1 expression compared to TCGA. We also observe increased mutation prevalence affecting BRCA1, BRCA2, and TP53 in SMC with an enrichment of a mutation signature linked to homologous recombination repair deficiency in TNBC. Finally, virtual microdissection and multivariate analyses reveal that Korean BC status is independently associated with increased TIL and decreased TGF-ß signaling expression signatures, suggesting that younger Asian BCs harbor more immune-active microenvironment than western BCs.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Carcinoma Ductal/genética , Carcinoma Lobular/genética , Transcriptoma , Adulto , Povo Asiático , Proteína BRCA1/genética , Proteína BRCA1/imunologia , Proteína BRCA2/genética , Proteína BRCA2/imunologia , Biomarcadores Tumorais/imunologia , Neoplasias da Mama/etnologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Carcinoma Ductal/etnologia , Carcinoma Ductal/imunologia , Carcinoma Ductal/patologia , Carcinoma Lobular/etnologia , Carcinoma Lobular/imunologia , Carcinoma Lobular/patologia , Estudos de Coortes , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/imunologia , Feminino , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Pós-Menopausa , Pré-Menopausa , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Microambiente Tumoral/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia , População Branca , Sequenciamento do Exoma
15.
Nucleic Acids Res ; 46(12): e75, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29672735

RESUMO

A key challenge in quantitative ChIP combined with high-throughput sequencing (ChIP-seq) is the normalization of data in the presence of genome-wide changes in occupancy. Analysis-based normalization methods were developed for transcriptomic data and these are dependent on the underlying assumption that total transcription does not change between conditions. For genome-wide changes in transcription factor (TF) binding, these assumptions do not hold true. The challenges in normalization are confounded by experimental variability during sample preparation, processing and recovery. We present a novel normalization strategy utilizing an internal standard of unchanged peaks for reference. Our method can be readily applied to monitor genome-wide changes by ChIP-seq that are otherwise lost or misrepresented through analytical normalization. We compare our approach to normalization by total read depth and two alternative methods that utilize external experimental controls to study TF binding. We successfully resolve the key challenges in quantitative ChIP-seq analysis and demonstrate its application by monitoring the loss of Estrogen Receptor-alpha (ER) binding upon fulvestrant treatment, ER binding in response to estrodiol, ER mediated change in H4K12 acetylation and profiling ER binding in patient-derived xenographs. This is supported by an adaptable pipeline to normalize and quantify differential TF binding genome-wide and generate metrics for differential binding at individual sites.


Assuntos
Imunoprecipitação da Cromatina/normas , Sequenciamento de Nucleotídeos em Larga Escala/normas , Análise de Sequência de DNA/normas , Animais , Anticorpos , Fator de Ligação a CCCTC/imunologia , Drosophila melanogaster/genética , Receptor alfa de Estrogênio/imunologia , Receptor alfa de Estrogênio/metabolismo , Histonas/imunologia , Histonas/metabolismo , Humanos , Células MCF-7 , Camundongos , Padrões de Referência
16.
Sci Signal ; 11(526)2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29666308

RESUMO

It has long been appreciated that most autoimmune disorders are characterized by increased prevalence in females, suggesting a potential role for sex hormones in the etiology of autoimmunity. To study how estrogen receptor α (ERα) contributes to autoimmune diseases, we generated mice in which ERα was deleted specifically in T lymphocytes. We found that ERα deletion in T cells reduced their pathogenic potential in a mouse model of colitis and correlated with transcriptomic changes that affected T cell activation. ERα deletion in T cells contributed to multiple aspects of T cell function, including reducing T cell activation and proliferation and increasing the expression of Foxp3, which encodes a critical transcription factor for the differentiation and function of regulatory T cells. Thus, these data demonstrate that ERα in T cells plays an important role in inflammation and suggest that ERα-targeted immunotherapies could be used to treat autoimmune disorders.


Assuntos
Autoimunidade/imunologia , Proliferação de Células , Receptor alfa de Estrogênio/imunologia , Inflamação/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Animais , Autoimunidade/genética , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Colite/genética , Colite/imunologia , Colite/metabolismo , Modelos Animais de Doenças , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Inflamação/genética , Inflamação/metabolismo , Ativação Linfocitária/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/metabolismo
17.
Int J Mol Sci ; 19(2)2018 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-29385743

RESUMO

The detection of human anti-estrogen receptor α antibodies (ERαABs) inducing estrogenic responses in MCF-7 mammary tumor cells suggests their implication in breast cancer emergence and/or evolution. A recent report revealing a correlation between the titer of such antibodies in sera from patients suffering from this disease and the percentage of proliferative cells in samples taken from their tumors supports this concept. Complementary evidence of the ability of ERαABs to interact with an epitope localized within the estradiol-binding core of ERα also argues in its favor. This epitope is indeed inserted in a regulatory platform implicated in ERα-initiated signal transduction pathways and transcriptions. According to some experimental observations, two auto-immune reactions may already be advocated to explain the emergence of ERαABs: one involving probably the idiotypic network to produce antibodies acting as estrogenic secretions and the other based on antibodies able to abrogate the action of a natural ERα inhibitor or to prevent the competitive inhibitory potency of released receptor degradation products able to entrap circulating estrogens and co-activators. All of this information, the aspect of which is mainly fundamental, may open new ways in the current tendency to combine immunological and endocrine approaches for the management of breast cancer.


Assuntos
Anticorpos Antineoplásicos/imunologia , Autoanticorpos/imunologia , Neoplasias da Mama/imunologia , Receptor alfa de Estrogênio/imunologia , Estrogênios/imunologia , Proteínas de Neoplasias/imunologia , Transdução de Sinais/imunologia , Animais , Neoplasias da Mama/patologia , Epitopos/imunologia , Feminino , Humanos
18.
Clin Immunol ; 183: 132-141, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28822833

RESUMO

Systemic lupus erythematosus (SLE) is a chronic and potentially severe autoimmune disease that disproportionately affects women. Despite a known role for hormonal factors impacting autoimmunity and disease pathogenesis, the specific mechanisms of action remain poorly understood. Our laboratory previously backcrossed "estrogen receptor alpha knockout (ERαKO)" mice onto the NZM2410 lupus prone background to generate NZM/ERαKO mice. This original ERαKO mouse, developed in the mid-1990s and utilized in hundreds of published studies, is not in fact ERα null. They express an N-terminally truncated ERα, and are considered a functional KO. They have physiologic deficiencies including infertility due to disruption of a critical activation domain (AF-1) at the N terminus of ERα, required for most classic estrogen (E2) actions. We demonstrated that female NZM/ERαKO mice had significantly less renal disease and significantly prolonged survival compared to WT littermates despite similar serum levels of autoantibodies and glomerular immune complex deposition. Herein, we present results of experiments using a lupus prone true ERα-/- mice (deletional KO mice on the NZM2410 background), surprisingly finding that these animals were not protected if they were ovariectomized, suggesting that another hormonal component confers protection, possibly testosterone, rather than the absence of the full-length ERα.


Assuntos
Anticorpos Antinucleares/imunologia , Receptor alfa de Estrogênio/genética , Lúpus Eritematoso Sistêmico/genética , Animais , Formação de Anticorpos , Autoanticorpos/imunologia , DNA/imunologia , Estradiol/sangue , Receptor alfa de Estrogênio/imunologia , Feminino , Predisposição Genética para Doença , Rim/patologia , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Camundongos Knockout , Ovariectomia , Testosterona/sangue
19.
Biochem Biophys Res Commun ; 490(3): 780-785, 2017 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-28645612

RESUMO

Inflammation has been recently acknowledged as a key participant in the physiopathology of oncogenesis and tumor progression. The inflammatory cytokine IL-1ß has been reported to induce the expression of markers associated with malignancy in breast cancerous cells through Epithelial-Mesenchymal Transition (EMT). Aggressive breast cancer tumors classified as Triple Negative do not respond to hormonal treatment because they lack three crucial receptors, one of which is the estrogen receptor alpha (ERα). Expression of ERα is then considered a good prognostic marker for tamoxifen treatment of this type of cancer, as the binding of this drug to the receptor blocks the transcriptional activity of the latter. Although it has been suggested that inflammatory cytokines in the tumor microenvironment could regulate ERα expression, the mechanism(s) involved in this process have not yet been established. We show here that, in a cell model of breast cancer cells (6D cells), in which the inflammatory cytokine IL-1ß induces EMT by activation of the IL-1ß/IL-1RI/ß-catenin pathway, the up regulation of TWIST1 leads to methylation of the ESR1 gene promoter. This epigenetic modification produced significant decrease of the ERα receptor levels and increased resistance to tamoxifen. The direct participation of IL-1ß in these processes was validated by blockage of the cytokine-induced signaling pathway by wortmannin inactivation of the effectors PI3K/AKT. These results support our previous reports that have suggested direct participation of the inflammatory cytokine IL-1ß in the transition to malignancy of breast cancer cells.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Metilação de DNA , Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/genética , Interleucina-1beta/imunologia , Tamoxifeno/farmacologia , Mama/efeitos dos fármacos , Mama/imunologia , Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Metilação de DNA/efeitos dos fármacos , Receptor alfa de Estrogênio/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/imunologia
20.
Cancer Immunol Immunother ; 66(5): 615-625, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28229217

RESUMO

Tumors evade immune recognition and destruction in many ways including the creation of an immune-suppressive tumor microenvironment (TME). Dendritic cells (DC) that infiltrate the TME are tolerogenic, and suppress effector T cells and anti-tumor activity. Previous reports demonstrated that a key regulator of tolerance in DC is the transcription factor FOXO3. Gender disparity has been studied in cancer in relation to incidence, aggressiveness, and prognosis. Few studies have touched on the importance in relation to impact on the immune system. In the current study, we show that there are significant differences in tumor growth between males and females. Additionally, frequencies and the function of FOXO3 expressed by DC subsets that infiltrate tumors vary between genders. Our results show for the first time that DC FOXO3 expression and function is altered in females. In vitro results indicate that these differences may be the result of exposure to estrogen. These differences may be critical considerations for the enhancement of immunotherapy for cancer.


Assuntos
Células Dendríticas/imunologia , Receptor alfa de Estrogênio/imunologia , Proteína Forkhead Box O3/metabolismo , Imunoterapia Adotiva/métodos , Melanoma Experimental/imunologia , Receptores Androgênicos/imunologia , Animais , Feminino , Proteína Forkhead Box O3/genética , Masculino , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA