Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 644
Filter
1.
Endocrinology ; 163(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34791151

ABSTRACT

Cancer immunology is the most rapidly expanding field in cancer research, with the importance of immunity in cancer pathogenesis now well accepted including in the endocrine-related cancers. The immune system plays an essential role in the development of ductal and luminal epithelial differentiation in the mammary gland. Originally identified as evolutionarily conserved antipathogen cytokines, interferons (IFNs) have shown important immune-modulatory and antineoplastic properties when administered to patients with various types of cancer, including breast cancer. Recent studies have drawn attention to the role of tumor- and stromal-infiltrating lymphocytes in dictating therapy response and outcome of breast cancer patients, which, however, is highly dependent on the breast cancer subtype. The emerging role of tumor cell-inherent IFN signaling in the subtype-defined tumor microenvironment could influence therapy response with protumor activities in breast cancer. Here we review evidence with new insights into tumor cell-intrinsic and tumor microenvironment-derived IFN signaling, and the crosstalk of IFN signaling with key signaling pathways in estrogen receptor-positive (ER+) breast cancer. We also discuss clinical implications and opportunities exploiting IFN signaling to treat advanced ER+ breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/biosynthesis , Interferons/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Differentiation , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cytokines/metabolism , Dendritic Cells/cytology , Female , Fibroblasts/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/cytology , Mice , NF-kappa B/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism
2.
Int J Mol Sci ; 22(12)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207568

ABSTRACT

Ovarian cancer remains the leading cause of death due to gynecologic malignancy. Estrogen-related pathways genes, such as estrogen receptors (ESR1 and ESR2) and their coregulators, proline-, glutamic acid-, and leucine-rich protein 1 (PELP1), and proto-oncogene tyrosine-protein kinase c-Src (SRC) are involved in ovarian cancer induction and development, still they require in-depth study. In our study, tissue samples were obtained from 52 females of Caucasian descent (control group without cancerous evidence (n = 27), including noncancerous benign changes (n = 15), and the ovarian carcinoma (n = 25)). Using quantitative analyses, we investigated ESRs, PELP1, and SRC mRNA expression association with ovarian tumorigenesis. Proteins' presence and their location were determined by Western blot and immunohistochemistry. Results showed that PELP1 and SRC expression levels were found to differ in tissues of different sample types. The expression patterns were complex and differed in the case of ovarian cancer patients compared to controls. The most robust protein immunoreactivity was observed for PELP1 and the weakest for ESR1. The expression patterns of analyzed genes represent a potentially interesting target in ovarian cancer biology, especially PELP1. This study suggests that specific estrogen-mediated functions in the ovary and ovary-derived cancer might result from different local interactions of estrogen with their receptors and coregulators.


Subject(s)
CSK Tyrosine-Protein Kinase/biosynthesis , Co-Repressor Proteins/biosynthesis , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Ovarian Neoplasms/metabolism , Transcription Factors/biosynthesis , Adult , Aged , CSK Tyrosine-Protein Kinase/genetics , Co-Repressor Proteins/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Humans , Middle Aged , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proto-Oncogene Mas , Transcription Factors/genetics
3.
J Cancer Res Clin Oncol ; 147(11): 3211-3224, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34244855

ABSTRACT

PURPOSE: Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors are widely used for the treatment of advanced estrogen receptor (ER)-positive breast cancer. To develop a treatment strategy for cancers resistant to CDK4/6 inhibitors, here, we established palbociclib-resistant sublines and analyzed their resistance mechanisms. METHODS: Palbociclib-resistant sublines were established from T47D and MCF7 cells. Sensitivity to other drugs was assessed via the WST assay. Altered expression/phosphorylation of proteins related to signal transduction and cell cycle regulation was examined using western blotting. Copy number alterations and mutations in the retinoblastoma (RB1) gene were also analyzed. RESULTS: Although an increase in CDK6 and decrease in retinoblastoma protein (Rb) expression/phosphorylation were commonly observed in the resistant sublines, changes in other cell cycle-related proteins were heterogeneous. Upon extended exposure to palbociclib, the expression/phosphorylation of these proteins became altered, and the long-term removal of palbociclib did not restore the Rb expression/phosphorylation patterns. Consistently a copy number decrease, as well as RB1 mutations were detected. Moreover, although the resistant sublines exhibited cross-resistance to abemaciclib, their response to dinaciclib was the same as that of wild-type cells. Of note, the cell line exhibiting increased mTOR phosphorylation also showed a higher sensitivity to everolimus. However, the sensitivity to chemotherapeutic agents was unchanged in palbociclib-resistant sublines. CONCLUSION: ER-positive breast cancer cells use multiple molecular mechanisms to survive in the presence of palbociclib, suggesting that targeting activated proteins may be an effective strategy to overcome resistance. Additionally, palbociclib monotherapy induces mutations and copy number alterations in the RB1 gene.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Piperazines/pharmacology , Pyridines/pharmacology , Receptors, Estrogen/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Cycle/physiology , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Drug Resistance, Neoplasm , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/metabolism , Humans , MCF-7 Cells , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/biosynthesis , Signal Transduction
4.
Anticancer Res ; 41(3): 1377-1386, 2021 03.
Article in English | MEDLINE | ID: mdl-33788729

ABSTRACT

BACKGROUND/AIM: The purpose of this study was to examine the expression of estrogen receptor α (ERα) and ß (ERß), androgen receptor (AR), SIRT1, SIRT2 and SIRT3 in prostate cancer (PCa). MATERIALS AND METHODS: From October 2010 to January 2015, 70 patients who had undergone radical prostatectomy following a PCa diagnosis were enrolled in our study. Normal prostate tissue (NPT) and prostate cancer tissues (PCAT) were separated, and the expression of each receptor in each tissue was analyzed with immunochemical staining. Univariate and multivariate analyses were performed to identify factors affecting the development of PCa. RESULTS: ERß and AR were highly expressed in PCAT compared with NPT (p<0.05). SIRT2 was highly expressed in NPT and PCAT (p<0.05). Univariate and multivariate analyses showed that AR and SIRT2 affect PCa development. CONCLUSION: AR is a risk factor for PC, and SIRT2 is associated with a lower incidence of PCa.


Subject(s)
Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Prostatic Neoplasms/metabolism , Receptors, Androgen/biosynthesis , Sirtuin 1/biosynthesis , Sirtuin 2/biosynthesis , Sirtuin 3/biosynthesis , Aged , Humans , Immunohistochemistry/methods , Male , Middle Aged , Multivariate Analysis , Prostatectomy/methods , Prostatic Neoplasms/surgery
5.
Cell Oncol (Dordr) ; 44(4): 951-957, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33723801

ABSTRACT

PURPOSE: Anti-estrogen therapy may be used as a palliative treatment option in high-grade serous ovarian carcinomas (HGSC). However, clinical implementation is limited as the use of estrogen receptor (ER) protein expression by immunohistochemistry remains insufficient in predicting therapy response. To determine the accuracy of ER protein expression as a marker for ER signaling pathway activity, we aimed to correlate ER protein expression to functional ER signaling pathway activity in HGSC. METHODS: Immunohistochemical ER protein expression was visually scored using total percentages of stained tumor cells and histoscores. Subsequently, mRNA was extracted, and RT-qPCR analysis was performed. Functional ER pathway activity was assessed by a computational Bayesian model inferring ER signaling pathway activity from mRNA levels of ER-specific target genes. RESULTS: Our analysis of 29 HGSCs shows that neither total percentage of ER protein expression, nor ER histoscores are significantly correlated to ER signaling pathway activity (respectively, p = 0.473 and p = 0.606). Classification of HGSC into three groups based on ER histoscores 0-100 (n = 6), 101-200 (n = 15) and 201-300 (n = 8) resulted in comparable mean ER signaling pathway activity among the groups (p = 0.356). Several samples in the higher ER histoscore groups had low ER signaling pathway activity, indicating that nuclear ER protein expression is not sufficient to describe transcriptional ER activation. CONCLUSION: Positive immunohistochemical ER staining is not always indicative of an active ER signaling pathway and is, therefore, a poor predictor of anti-estrogen response. Further research is needed to prove the predictive value of ER signaling pathway activity regarding anti-estrogen sensitivity in HGSC patients.


Subject(s)
Biomarkers, Tumor/genetics , Cystadenocarcinoma, Serous/genetics , Estrogen Receptor alpha/genetics , Ovarian Neoplasms/genetics , Signal Transduction/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Estrogen Receptor alpha/biosynthesis , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction
6.
J Ethnopharmacol ; 269: 113696, 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33358854

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Si-Wu-Tang (SWT), a prestigious herbal formula from China, has been extensively used for centuries for female-related diseases. It has been documented that SWT has a significant inhibitory effect on non-triple-negative breast cancer (non-TNBC) cells. However, there has been limited comprehensive analysis of the targeted effects of the anticancer components of SWT and its exact biological mechanism. AIM OF THE STUDY: This study aims to uncover the mechanism by which SWT treats non-TNBC by applying a network pharmacological method combined with experimental validation. MATERIALS AND METHODS: First, SWT compounds were collected from the Traditional Chinese Medicines Systems Pharmacology database (TCMSP) and The Encyclopedia of Traditional Chinese Medicine (ETCM), and then the targets related to SWT were obtained from the TCMSP and SwissTarget databases. Second, a target data set of non-TNBC proteins was established by using the Online Mendelian Inheritance in Man (OMIM), GeneCards and Gene Expression Omnibus (GEO) databases. Third, based on the overlap of targets between SWT and non-TNBC, a protein-protein interaction (PPI) network was built to analyse the interactions among these targets, which focused on screening for hub targets by topology. On these hub genes, we conducted a meta-analysis and survival analysis to screen the best match targets, ESR1, PPARG, CAT, and PTGS2, which had a strong correlation with the ingredients of SWT in our verification by molecular docking. In vitro experiments further proved the reliability of the network pharmacology findings. Finally, FunRich software and the ClusterProfiler package were utilized for the enrichment analysis of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) data. RESULTS: A total of 141 active ingredients and 116 targets of SWT were selected. GO enrichment analysis showed that the biological processes through which SWT acted against non-TNBC (FDR<0.01) mainly involved modulating energy metabolism and apoptosis. According to RT-qPCR and Western blotting, the mRNA and protein expression of ESR1, PPARG and PTGS2 were upregulated (P < 0.01), and the mRNA and protein levels of CAT were downregulated (P < 0.01), suggesting a multi-gene regulatory molecular mechanism of SWT against non-triple-negative breast cancer. CONCLUSIONS: This research explored the multi-gene pharmacological mechanism of action of SWT against non-TNBC through network pharmacology and in vitro experiments. The findings provide new ideas for research on the mechanism of action of Chinese medicine against breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Apoptosis/drug effects , Catalase/biosynthesis , Catalase/genetics , Cyclooxygenase 2/genetics , Databases, Chemical , Databases, Genetic , Energy Metabolism/drug effects , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Medicine, Chinese Traditional/methods , Molecular Docking Simulation , PPAR gamma/biosynthesis , PPAR gamma/genetics , Protein Binding , Protein Interaction Maps
7.
Mol Cell Biochem ; 476(2): 525-533, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33064289

ABSTRACT

Our previous study showed remarkable differences in the effect of R-sulforaphane (R-SFN) on the expression of CYPs 19, 1A1, 1A2, and 1B1 in ER(+) MCF7, ER( -) MDA-MB-231, and non-tumorigenic immortalized MCF10A (8). This study aimed to evaluate the effect of R-SFN on phase II enzymes induction and expression of AhR, Nrf2, and ERα in the same breast cell lines. The results showed increased expression of GSTP as a result of treatment with R-SFN in breast cancer cells. An increased NQO1 transcript and protein levels were found in all breast cells, with the most significant increase in MCF7 cells. Similarly, the enhancement of Nrf2 expression was noticed in all tested cells. AhR gene transcript and protein were decreased in MCF7 cells. In MDA-MB-231, increased AhR mRNA was not confirmed at the protein level. No differences were found in the expression of ERα. Overall, the results of the present study extended our earlier suggestions on the possible interference of R-SFN with estrogens homeostasis in breast cancer cells differing in ERα status, as well as in non-tumorigenic immortalized breast epithelial cells. While some of R-SFN effects might be beneficial and useful in breast cancer prevention, the others, particularly GSTP induction, may lead to adverse effects.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/biosynthesis , Glutathione S-Transferase pi/biosynthesis , Isothiocyanates/pharmacology , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NF-E2-Related Factor 2/biosynthesis , Receptors, Aryl Hydrocarbon/biosynthesis , Sulfoxides/pharmacology , Anticarcinogenic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/metabolism , Humans , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Transcriptome
8.
J Comp Neurol ; 529(4): 786-801, 2021 03.
Article in English | MEDLINE | ID: mdl-32632943

ABSTRACT

Sex steroid hormones such as 17ß-estradiol (estradiol) regulate neuronal function by binding to estrogen receptors (ERs), including ERα and GPER1, and through differential production via the enzyme aromatase. ERs and aromatase are expressed across the nervous system, including in the striatal brain regions. These regions, comprising the nucleus accumbens core, shell, and caudate-putamen, are instrumental for a wide-range of functions and disorders that show sex differences in phenotype and/or incidence. Sex-specific estrogen action is an integral component for generating these sex differences. A distinctive feature of the striatal regions is that in adulthood neurons exclusively express membrane but not nuclear ERs. This long-standing finding dominates models of estrogen action in striatal regions. However, the developmental etiology of ER and aromatase cellular expression in female and male striatum is unknown. This omission in knowledge is important to address, as developmental stage influences cellular estrogenic mechanisms. Thus, ERα, GPER1, and aromatase cellular immunoreactivity was assessed in perinatal, prepubertal, and adult female and male rats. We tested the hypothesis that ERα, GPER1, and aromatase exhibits sex, region, and age-specific differences, including nuclear expression. ERα exhibits nuclear expression in all three striatal regions before adulthood and disappears in a region- and sex-specific time-course. Cellular GPER1 expression decreases during development in a region- but not sex-specific time-course, resulting in extranuclear expression by adulthood. Somatic aromatase expression presents at prepuberty and increases by adulthood in a region- but not sex-specific time-course. These data indicate that developmental period exerts critical sex-specific influences on striatal cellular estrogenic mechanisms.


Subject(s)
Caudate Nucleus/metabolism , Estrogen Receptor alpha/biosynthesis , Nucleus Accumbens/metabolism , Putamen/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Sex Characteristics , Animals , Caudate Nucleus/chemistry , Caudate Nucleus/growth & development , Estrogen Receptor alpha/analysis , Female , Male , Nucleus Accumbens/chemistry , Nucleus Accumbens/growth & development , Putamen/chemistry , Putamen/growth & development , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/analysis
9.
Malays J Pathol ; 42(3): 415-422, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33361723

ABSTRACT

INTRODUCTION: Papillary thyroid carcinoma (PTC) is the ninth most common malignancy among women. Although the disease prognosis is good, less favourable outcomes are predicted in those with higher disease stages and nodal metastasis. Oestrogen- α (ER-α) expression has been associated with aggressive presentation and greater disease progression and has been proposed as a predictor for lymph node metastases. The objective of this study was to evaluate the association between ER expression and clinicopathological features i.e. lymph node metastasis, tumour size, extrathyroidal extension, histological variants of PTC , age groups , ethnic and gender. METHODS: We studied ER-α expression in 84 cases of PTC obtained within an eight-year period (2011-2018) by immunohistochemical technique (IHC). Associations between ER-α expression and clinicopathological features were evaluated using Fisher's exact test. The statistical significance was set at p < 0.05. RESULTS: ER-α was expressed in 13.1% of all the PTC cases examined (n=11/84). There were no associations observed between ER-α expression and lymph node metastasis (p=1.000), tumour size (p=0.970), extrathyroidal extension (p=0.677), variants of PTC (p=1.000), age groups (p=0.188), gender (p=0.725) or race (p=0.920). CONCLUSION: There was no evidence in this study to support the application of ER-α as prediction marker for lymph node metastasis or disease aggressiveness in PTC. Given that the scope of this study was limited to the protein expression of ER- α, we also propose the inclusion of molecular analysis of ESR1 gene expression, as well as inclusion of detailed clinical and radiological findings in future research investigating the role of ER-α in prognostication of PTC.


Subject(s)
Biomarkers, Tumor/metabolism , Estrogen Receptor alpha/biosynthesis , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Female , Humans , Lymphatic Metastasis/pathology , Male , Middle Aged
10.
J Korean Med Sci ; 35(50): e420, 2020 Dec 28.
Article in English | MEDLINE | ID: mdl-33372422

ABSTRACT

BACKGROUND: Studies have reported that epithelial cell proliferation may be involved in the pathogenesis of nasal polyps (NPs). Estrogen receptor (ER)-α, one type of ER, is related to anti-inflammatory action and cell survival in certain tissues. In this study, we examined the presence or absence of ER-α in NPs and healthy inferior turbinate mucosae. We also investigated the effect of dexamethasone on ER-α expression, cell viability, and apoptosis in RPMI 2650 cells. METHODS: Immunohistochemical staining and Western blot analysis were conducted to determine the expression of ER-α in 15 NPs and 15 healthy inferior turbinate mucosae. After treating RPMI 2650 cells with dexamethasone, ER-α expression was analyzed using Western blot analysis and cell viability was determined using the MTT assay. Western blot analysis and annexin V-phycoerythrin (PE) staining were used to examine apoptotic cell death. RESULTS: Western blot analysis showed that ER-α expression was upregulated in 13 of the 15 NP tissues. Immunohistochemical staining for ER-α confirmed the results of the Western blot analysis. When RPMI 2650 cells were treated with dexamethasone, both ER-α expression and cell viability were decreased. Furthermore, the treatment of RPMI 2650 cells with dexamethasone increased apoptotic cell death, as shown by increased levels of BAX and cleaved caspase-3, decreased levels of Bcl-2, and an increased percentage of positive annexin V-PE stained cells. CONCLUSION: ER-α expression was higher in NPs than in healthy inferior turbinate mucosae. When RPMI 2650 cells were treated with dexamethasone, ER-α expression was downregulated, cell viability decreased, and apoptosis increased. The decreased cell viability may be related, at least in part, to the decreased ER-α protein levels, which likely contributed to the induction of apoptotic cell death in RPMI 2650 cells.


Subject(s)
Dexamethasone/pharmacology , Estrogen Receptor alpha/biosynthesis , Nasal Polyps/metabolism , Anti-Inflammatory Agents/pharmacology , Apoptosis , Caspase 3/metabolism , Cell Line , Cell Survival , Dexamethasone/chemistry , Endoscopy , Fulvestrant , Humans , Immunohistochemistry , Keratins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , Tetrazolium Salts , Thiazoles , Turbinates/metabolism , bcl-2-Associated X Protein/metabolism
11.
Front Endocrinol (Lausanne) ; 11: 568375, 2020.
Article in English | MEDLINE | ID: mdl-33117284

ABSTRACT

The estrogen receptor alpha (ERα) is a ligand-activated transcription factor whose activity is modulated by its interaction with multiple protein complexes. In this work, we have identified the protein interferon alpha inducible protein 27 (IFI27/ISG12) as a novel ERα-associated protein. IFI27/ISG12 transcription is regulated by interferon and estradiol and its overexpression is associated to reduced overall survival in ER+ breast cancer patients but its function in mammary gland tissue remains elusive. In this study we showed that overexpression of IFI27/ISG12 in breast cancer cells attenuates ERα transactivation activity and the expression of ERα-dependent genes. Our results demonstrated that IFI27/ISG12 overexpression in MCF-7 cells reduced their proliferation rate in 2-D and 3-D cell culture assays and impaired their ability to migrate in a wound-healing assay. We show that IFI27/ISG12 downregulation of ERα transactivation activity is mediated by its ability to facilitate the interaction between ERα and CRM1/XPO1 that mediates the nuclear export of large macromolecules to the cytoplasm. IFI27/ISG12 overexpression was shown to impair the estradiol-dependent proliferation and tamoxifen-induced apoptosis in breast cancer cells. Our results suggest that IFI27/ISG12 may be an important factor in regulating ERα activity in breast cancer cells by modifying its nuclear versus cytoplasmic protein levels. We propose that IFI27/ISG12 may be a potential target of future strategies to control the growth and proliferation of ERα-positive breast cancer tumors.


Subject(s)
Breast Neoplasms/metabolism , Down-Regulation/physiology , Estrogen Receptor alpha/biosynthesis , Karyopherins/biosynthesis , Membrane Proteins/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Transcriptional Activation/physiology , Breast Neoplasms/genetics , Databases, Genetic , Down-Regulation/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Humans , Karyopherins/genetics , MCF-7 Cells , Membrane Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Tamoxifen/pharmacology , Transcriptional Activation/drug effects , Exportin 1 Protein
12.
Sci Rep ; 10(1): 15062, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934281

ABSTRACT

Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.


Subject(s)
Androgen-Insensitivity Syndrome/metabolism , Anti-Mullerian Hormone/metabolism , Estrogen Receptor alpha/biosynthesis , Neoplasm Proteins/biosynthesis , Peutz-Jeghers Syndrome/metabolism , Response Elements , Sertoli Cells/metabolism , Androgen-Insensitivity Syndrome/pathology , Animals , Cell Line , Child , Child, Preschool , Estradiol/metabolism , Female , Humans , Male , Mice , Peutz-Jeghers Syndrome/pathology , Sertoli Cells/pathology
13.
J Reprod Dev ; 66(6): 515-522, 2020 Dec 22.
Article in English | MEDLINE | ID: mdl-32830152

ABSTRACT

Cluster of differentiation (CD) 9 and CD81 are closely-related members of the tetraspanin family that consist of four-transmembrane domain proteins. Cd9 and Cd81 are highly expressed in breast cancer cells; however, their expression in healthy mammary glands is unclear. In this study, we performed quantitative real-time PCR to analyze the expression levels of Cd9 and Cd81. Histological techniques were employed to identify Cd9- and Cd81-expressing cells in rat mammary glands during pregnancy and lactation. It was observed that Cd9 and Cd81 were expressed in the mammary glands, and their expression levels correlated with mammary gland development. To identify cells expressing Cd9 and Cd81 in the mammary glands, we performed double immunohistochemical staining for CD9 and CD81, prolactin receptor long form, estrogen receptor alpha, or Ki67. The results showed that CD9 and CD81 were co-expressed in proliferating mammary epithelial cells. Next, we attempted to isolate CD9-positive epithelial cells from the mammary gland using pluriBead cell-separation technology based on antibody-mediated binding of cells to beads of different sizes, followed by isolation using sieves with different mesh sizes. We successfully isolated CD9-positive epithelial cells with 96.8% purity. In addition, we observed that small-interfering RNAs against Cd9 and Cd81 inhibited estrogen-induced proliferation of CD9-positive mammary epithelial cells. Our current findings may provide novel insights into the proliferation of mammary epithelial cells during pregnancy and lactation as well as in pathological processes associated with breast cancer.


Subject(s)
Epithelial Cells/cytology , Gene Expression Profiling , Mammary Glands, Animal/metabolism , Tetraspanin 28/biosynthesis , Tetraspanin 29/biosynthesis , Animals , Cell Differentiation , Cell Proliferation , Diethylstilbestrol , Estrogen Receptor alpha/biosynthesis , Female , Ki-67 Antigen/biosynthesis , Lactation , Pregnancy , Pregnancy, Animal , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
14.
Sci Rep ; 10(1): 4024, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32132580

ABSTRACT

The estrogen signaling pathway has been reported to modulate prostate cancer (PCa) progression through the activity of estrogen receptors α and ß (ERα and ERß). Given that selective estrogen receptor modulators (SERMs) are used to treat breast cancer, ERs have been proposed as attractive therapeutic targets in PCa. However, many inconsistencies regarding the expression of ERs and the efficacy of SERMs for PCa treatment exist, notably due to the use of ERß antibodies lacking specificity and treatments with high SERM concentrations leading to off-target effects. To end this confusion, our objective was to study the impact of estrogenic and anti-estrogenic ligands in well-studied in vitro PCa models with appropriate controls, dosages, and ER subtype-specific antibodies. When using physiologically relevant concentrations of nine estrogenic/anti-estrogenic compounds, including five SERMs, we observed no significant modulation of PCa cell proliferation. Using RNA-seq and validated antibodies, we demonstrate that these PCa models do not express ERs. In contrast, RNA-seq from PCa samples from patients have detectable expression of ERα. Overall, our study reveals that commonly used PCa models are inappropriate to study ERs and indicate that usage of alternative models is essential to properly assess the roles of the estrogen signaling pathway in PCa.


Subject(s)
Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Humans , MCF-7 Cells , Male , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
15.
Int J Biol Sci ; 16(8): 1388-1402, 2020.
Article in English | MEDLINE | ID: mdl-32210727

ABSTRACT

Objective: The therapeutic effects of the checkpoint kinase 1 (CHK1)-targeted inhibition in tumor therapy have been confirmed, but how to choose an effective application method in breast cancer with heterogeneous molecular characteristics has remained unclear. Methods: We evaluated the status of CHK1 in breast cancer using the cancer genome atlas database. Chemosensitivity and single-agent antitumor activity of CHK1 inhibition were measured by drug sensitivity assay, cell proliferation assay, cell cycle and apoptosis analysis in breast cancer with different ER/PR status. And based on the conjoint transcriptome atlas analyses, the corresponding mechanism were explored. Results: In ER-/PR-/HER2- breast cancer, CHK1 inhibition enhanced adriamycin (ADR) chemosensitivity which was mediated by the mitotic checkpoint complex (MCC)-anaphase-promoting complex/cyclosome (APC/C)-cyclin B1 axis, Msh homeobox 2 (MSX2) and Bcl-2-like protein 11 (BIM). However, in ER+/PR+/HER2- breast cancer, because of the significant suppression for centromere protein F (CENPF)-mediated transcriptional activation of CHK1 induced by ADR itself, CHK1 inhibition fails to sensitize ADR toxicity. Interestingly, CHK1 inhibition showed the single-agent antitumor activity in ER+/PR+/HER2- breast cancer which was mediated by the cyclin dependent kinase inhibitor 1A (p21), kinesin family member 11 (Eg5) and cell surface death receptor (Fas). Conclusions: CHK1's variable role determines the application of CHK1 inhibition in breast cancer with ER/PR heterogeneity.


Subject(s)
Breast Neoplasms/metabolism , Checkpoint Kinase 1/biosynthesis , Estrogen Receptor alpha/biosynthesis , Receptors, Progesterone/biosynthesis , Apoptosis , Breast Neoplasms/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Computational Biology , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor/methods , Female , Genome, Human , Humans , Kaplan-Meier Estimate , MCF-7 Cells , RNA Interference , Treatment Outcome
16.
Development ; 147(5)2020 03 11.
Article in English | MEDLINE | ID: mdl-32098763

ABSTRACT

17ß-Estradiol induces the postnatal development of mammary gland and influences breast carcinogenesis by binding to the estrogen receptor ERα. ERα acts as a transcription factor but also elicits rapid signaling through a fraction of ERα expressed at the membrane. Here, we have used the C451A-ERα mouse model mutated for the palmitoylation site to understand how ERα membrane signaling affects mammary gland development. Although the overall structure of physiological mammary gland development is slightly affected, both epithelial fragments and basal cells isolated from C451A-ERα mammary glands failed to grow when engrafted into cleared wild-type fat pads, even in pregnant hosts. Similarly, basal cells purified from hormone-stimulated ovariectomized C451A-ERα mice did not produce normal outgrowths. Ex vivo, C451A-ERα basal cells displayed reduced matrix degradation capacities, suggesting altered migration properties. More importantly, C451A-ERα basal cells recovered in vivo repopulating ability when co-transplanted with wild-type luminal cells and specifically with ERα-positive luminal cells. Transcriptional profiling identified crucial paracrine luminal-to-basal signals. Altogether, our findings uncover an important role for membrane ERα expression in promoting intercellular communications that are essential for mammary gland development.


Subject(s)
Epithelium/metabolism , Estrogen Receptor alpha/biosynthesis , Mammary Glands, Animal/embryology , Paracrine Communication/physiology , Animals , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Estradiol/metabolism , Estrogen Receptor alpha/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Lipoylation/physiology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/transplantation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
17.
Eur Rev Med Pharmacol Sci ; 24(3): 1460-1467, 2020 02.
Article in English | MEDLINE | ID: mdl-32096196

ABSTRACT

OBJECTIVE: To explore the role of estrogen and estrogen receptors in the migration of vascular smooth muscle cells in varicose lower-extremity veins. PATIENTS AND METHODS: Tissue samples of normal lower extremity vein (56 cases) and varicose lower extremity vein (47 cases) were collected. Western blot and real-time fluorescent qPCR were performed to measure the expression of Estrogen receptor α (ERα) in tissues. Two cell co-culture systems were established for human umbilical vein endothelial cells (HUVECs) and human umbilical vein smooth muscle cells (HUVSMCs). One system was incubated under normal oxygen conditions (normal oxygen group), and the other was under oxygen-poor conditions (hypoxia group). The two systems were treated with 10-7 mM Estrogen E2, 10-7 mM BSA-conjugated Estrogen E2-BSA, 10-7 mM Estrogen E2+10-3 mM Tamoxifen (TAM), respectively for 24 h. The treated cells were subjected to cell scratch assay, transwell assay, and Western blot analysis of MMP2 and MMP9 protein expression. RESULTS: The expression of ERα in varicose lower extremity vein was significantly up-regulated compared with that in normal lower extremity vein. The cell migration rate and the number of migrating cells in untreated hypoxia group and E2-treated normal oxygen group were comparable (p>0.05) to those in untreated normal oxygen group. The cell migration rate and the number of migrating cells were significantly increased (p<0.05) in E2-treated hypoxia group, compared with E2-treated normal oxygen group and untreated hypoxia group. The cell migration rate, the number of migrating cells, and expression levels of MMP2 and MMP9 were significantly decreased in E2/TAM-treated hypoxia group, compared with those in E2-treated hypoxia group. CONCLUSIONS: In summary, E2 can promote the migration of vascular smooth muscle cells and induce varicose veins of the lower extremities, which may be related to the promotion of MMP2 and MMP9 expression through the classical pathway of ER.


Subject(s)
Cell Movement/physiology , Estrogen Receptor alpha/biosynthesis , Estrogens/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Muscle, Smooth, Vascular/enzymology , Adult , Cell Movement/drug effects , Coculture Techniques , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Enzymologic , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Middle Aged , Muscle, Smooth, Vascular/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Varicose Veins/enzymology , Varicose Veins/pathology
18.
J Ethnopharmacol ; 254: 112572, 2020 May 23.
Article in English | MEDLINE | ID: mdl-31940455

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Antipsychotics often induce hyperprolactinemia. The transforming growth factor (TGF)-beta1 signaling in the pituitary and hypothalamus inhibits prolactin synthesis and secretion, and its impairment is implicated in neuropsychiatric disorders. Longdan Xiegan Tang (LXT) alone or together with antipsychotics have been used to treat various neuropsychiatric diseases and hyperprolactinemia-associated disorders. AIM OF THE STUDY: To investigate the effect of LXT on hyperprolactinemia and involvement of the TGF-beta1 signaling. MATERIALS AND METHODS: Male rats were co-administered with olanzapine (5 mg/kg) and LXT extract (50 and 500 mg/kg) (p.o., × 8 weeks). Plasma concentrations of prolactin and TGF-beta1 were determined by ELISA. Protein expression was analyzed by Western blot. RESULTS: Treatment of rats with LXT extract suppressed olanzapine-induced increase in plasma prolactin concentration and overexpression of pituitary and hypothalamic prolactin protein. Importantly, LXT restored olanzapine-induced decrease in protein expression of the key components of the TGF-beta1 signaling, TGF-beta1, type II TGF-beta receptor, type I TGF-beta receptor and phosphorylated SMAD3 in the pituitary and hypothalamus. Further, it antagonized downregulation of pituitary and hypothalamic dopamine D2 receptor (D2R) protein level, and inhibited pituitary estrogen receptor (ER) alpha and ERbeta protein expression. CONCLUSIONS: The present results suggest that LXT ameliorates antipsychotic-induced hyperprolactinemia in rats by repairing the pituitary and hypothalamic TGF-beta1 signaling possibly via D2R, ERs or/and other pathways. Our findings may also provide scientific elucidation for use of the ancient Chinese formula to treat the impaired TGF-beta1 signaling-associated neuropsychiatric disorders.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hyperprolactinemia/prevention & control , Hypothalamus/metabolism , Olanzapine/adverse effects , Pituitary Gland/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Antipsychotic Agents/adverse effects , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Hyperprolactinemia/chemically induced , Male , Prolactin/biosynthesis , Rats , Receptors, Dopamine D2/metabolism , Signal Transduction/drug effects
19.
Neurotoxicol Teratol ; 78: 106864, 2020.
Article in English | MEDLINE | ID: mdl-31926947

ABSTRACT

Bisphenol A (BPA) is a xenoestrogen that is prevalent in the environment of industrialized nations due its use in the production of many plastic household items. Virtually all adults in the U.S. have detectable levels of BPA in urine and it can be measured in fetal serum and in breastmilk, making developmental exposure a particular concern. The present study utilizes a progesterone receptor (PR) expression bioassay to assess the estrogen receptor α (ERα)-dependent effects of BPA in fetal rodent brain following maternal exposure. Maternal ingestion of 10 µg/kg/day, but not 50 µg/kg/day, BPA from gestational day 14-22 significantly increased levels of PR immunoreactivity (PRir) in the medial preoptic nucleus (MPN) of female offspring. PR expression in the perinatal MPN is highly dependent on the activation of ERα, but not ERß, by estrogens. Indeed, injections of BPA (5 µg/kg) to neonates from postnatal day 2-4 (P2-4) significantly increased PR expression in the MPN of postnatal day 5 females compared to the MPN of females administered the oil vehicle. However, pretreatment with the ER antagonist, ICI 182,780 from P1-4 significantly attenuated the effects of BPA on PR expression, indicating an ERα-dependent mechanism. The present results also demonstrate a non-monotonic effect of BPA on the direct expression of a transcription factor in developing brain.


Subject(s)
Benzhydryl Compounds/toxicity , Estrogen Receptor alpha/biosynthesis , Estrogens, Non-Steroidal/toxicity , Phenols/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Preoptic Area/drug effects , Receptors, Progesterone/biosynthesis , Animals , Female , Pregnancy , Preoptic Area/metabolism , Rats, Sprague-Dawley
20.
Medicine (Baltimore) ; 98(46): e17954, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31725654

ABSTRACT

BACKGROUND: Published studies have investigated the prognostic roles of estrogen receptor alpha (ERα) and estrogen receptor beta (ERß) in gastroesophageal cancer patients with the controversial results. The aim of the study was to systematically evaluate the impacts of ERα and ERß on the overall survival (OS) in patients. METHOD: Relevant eligible studies were extracted from PubMed, Embase, Web of Science, CNKI and Wanfang databases (from the start date to November 2018) following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. HR (hazard ratio) with 95% confidence intervals (CIs) were used to assess the prognostic values of ERα and ERß for OS in patients. RESULTS: High ERα expression was associated with poor OS (HR = 1.58, 95% CI = 1.29-1.94, P < .001) and ERß with better OS (HR = 0.56, 95% CI = 0.37-0.83, P = .004) in gastroesophageal cancer. Furthermore, unfavorable OS was found in Chinese gastroesophageal patients with higher ERα expression (HR = 1.57, 95% CI = 1.25-1.96, P < .001) and better OS with higher ERß expression (HR = 0.51, 95% CI = 0.31-0.83, P < .01) in our subgroup analysis. Meanwhile, worse OS was found in esophageal squamous cell carcinoma (ESCC) patients with high ERα expression (HR = 1.74, 95% CI = 1.33-2.26, P < .001), and favorable OS in ESCC with ERß overexpression (HR = 0.40, 95% CI = 0.31-0.52, P < .001). Besides, high ERα expression was associated with lower tumor differentiation in ESCC (OR = 1.64; 95% CI = 1.02-2.64, P = .04) and ERß was linked with better tumor differentiation in gastric adenocarcinoma (GCA) (OR = 0.49; 95% CI = 0.26-0.94, P = .03). CONCLUSIONS: ERα and ERß might serve as potential prognostic biomarkers for gastroesophageal cancer patients. ERα overexpression predicted poor OS and lower tumor differentiation, and ERß suggested favorable OS and better tumor differentiation. Further related studies should be performed to test these results.


Subject(s)
Esophageal Neoplasms/mortality , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Stomach Neoplasms/mortality , Biomarkers, Tumor , China/epidemiology , Esophageal Neoplasms/blood , Esophageal Neoplasms/pathology , Estrogen Receptor alpha/blood , Estrogen Receptor beta/blood , Humans , Prognosis , Proportional Hazards Models , Stomach Neoplasms/blood , Stomach Neoplasms/pathology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...