Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 292
Filter
Add more filters

Publication year range
1.
Biochem Cell Biol ; 99(5): 666-674, 2021 10.
Article in English | MEDLINE | ID: mdl-33974808

ABSTRACT

Liver fibrosis is the most common pathway in most types of chronic liver damage, characterized by an imbalance of ECM degradation and synthesis. Saikosaponin-d (SSd) possesses anti-inflammatory and anti-fibrotic effects. However, the underlying mechanism by which SSd represses hepatic stellate cell (HSC) activation remains unclear. Here, we found that SSd remarkably alleviated carbon tetrachloride (CCl4)-induced liver fibrosis, as evidenced by decreased collagen levels and profibrotic marker (COl1a1 and α-smooth muscle actin (SMA)) expression. SSd repressed CCl4-induced NOD-like receptor family pyrin-domain-containing-3 (NLRP3) activation in fibrotic livers, as suggested by decreased levels of NLRP3, IL-18, and IL-ß. The primary HSCs of CCl4 mice exhibited a significant increase in profibrotic marker expression and NLRP3 activation, but SSd treatment reversed this effect. SSd also repressed TGF-ß-induced profibrotic marker expression and NLRP3 activation in vitro. Mechanistically, TGF-ß decreased the expression of estrogen receptor-ß (ERß) in HSCs, whereas SSd treatment reversed this effect. ERß inhibition enhances NLRP3 activation in HSCs. More importantly, ERß or NLRP3 inhibition partially destroyed the function of SSd in liver fibrosis. In summary, the current data suggest that SSd prevents hepatic fibrosis by regulating the ERß/NLRP3 inflammasome pathway and suggests SSd as a potential agent for treating liver fibrosis.


Subject(s)
Estrogen Receptor beta/antagonists & inhibitors , Inflammasomes/drug effects , Liver Cirrhosis/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Oleanolic Acid/analogs & derivatives , Protective Agents/pharmacology , Saponins/pharmacology , Animals , Carbon Tetrachloride , Cells, Cultured , Estrogen Receptor beta/metabolism , Inflammasomes/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oleanolic Acid/pharmacology
2.
Bioorg Chem ; 116: 105364, 2021 11.
Article in English | MEDLINE | ID: mdl-34560558

ABSTRACT

(±)-Caryopterisines A (1) and B (2) featuring an unprecedented 6/5/5/5/6 pentacyclic rings system were isolated from Caryopteris glutinosa. The structures were determined by spectroscopic and X-ray crystallographic data analyses as well as theoretical calculations. Chiral HPLC resolution of both racemic 1 and 2 afforded their corresponding enantiotropic enantiomers. A plausible biogenesis for 1 and 2 may be originated from Diels-Alder reaction between pyridine-containing oxerine derivatives. The enantiotropic conversion mechanism of the enantiomers was demonstrated by H-D exchange and 18O incorporation studies. Compounds 1 and 2 showed moderate inhibition of estrogen E2 biosynthesis in human ovarian granulosa-like KGN cells. These two alkaloids reduced kynurenine biosynthesis at moderate level via inhibition of indoleamine 2,3-dioxygenase. Alkaloid 2 exhibited moderate inhibition of the release of interleukin-1ß.


Subject(s)
Alkaloids/pharmacology , Estrogen Receptor beta/antagonists & inhibitors , Lamiaceae/chemistry , Monoterpenes/pharmacology , Alkaloids/chemistry , Alkaloids/isolation & purification , Cell Line , Dose-Response Relationship, Drug , Estrogen Receptor beta/metabolism , Humans , Molecular Structure , Monoterpenes/chemistry , Monoterpenes/isolation & purification , Stereoisomerism , Structure-Activity Relationship
3.
Biol Pharm Bull ; 44(10): 1524-1529, 2021.
Article in English | MEDLINE | ID: mdl-34602561

ABSTRACT

Bisphenol A (BPA) has been shown to induce the activation of nuclear estrogen receptor α/ß (ERα/ß) in both in vitro and in vivo settings. We originally obtained a 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene (MBP), a possible active metabolite of BPA, strongly activating the ERs-mediated transcription in MCF-7 cells with an EC50 of 2.8 nM (i.e., BPA's EC50 = 519 nM). Environmental estrogens can also target G protein-coupled estrogen receptor 1 (GPER1), a membrane-type ER. However, the effects of BPA/MBP on GPER1, have not yet been fully resolved. In this study, we used MCF-7, a ERα/ERß/GPER1-positive human breast cancer cell line, as a model to investigate the effects of the exposure to BPA or MBP. Our results revealed that at concentrations below 1 nM MBP, but not BPA, downregulates the expression of GPER1 mRNA via upregulated ERß, and the MCF-7 cells pre-treated with MBP display resistance to GPER1 agonist G-1-mediated anti-proliferative effects. Because GPER1 can act as a tumor suppressor in several types of cancer including breast cancer, the importance of MBP-mediated decrease in GPER1 expression in breast cancer cells is discussed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cyclopentanes/pharmacology , Estrogen Receptor beta/antagonists & inhibitors , Phenols/pharmacology , Quinolines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclopentanes/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Female , Humans , MCF-7 Cells , Phenols/therapeutic use , Quinolines/therapeutic use , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
4.
Bioorg Med Chem ; 28(3): 115274, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31879182

ABSTRACT

17ß-Estradiol (E2) is a natural steroid ligand for the structurally and physiologically independent estrogen receptors (ERs) ERα and ERß. We recently observed that CF3-containing bisphenol AF (BPAF) works as an agonist for ERα but as an antagonist for ERß. Similar results were also observed for the CCl3-containing bisphenol designated as HPTE. Both BPAF and HPTE are comprised of a tri-halogenated methyl group in the central alkyl moiety of their bisphenol structures, which strongly suggests that halogens contribute directly to the agonist/antagonist dual biological functions. We conducted this study to investigate the structure-activity relationships by assessing together newly synthesized CF3- and CBr3-containing bisphenol E analogs (BPE-X). We first tested bisphenols for their receptor binding ability and then for their transcriptional activities. Halogen-containing bisphenols were found to be fully active for ERα, but almost completely inactive for ERß. When we examined these bisphenols for their inhibitory activities for E2 in ERß, we observed that they worked as distinct antagonists. The ascending order of agonist/antagonist dual biological functions was BPE-F < BPE-Cl (HPTE) ≤ BPAF < BPE-Br, demonstrating that the electrostatic halogen bonding effect is a major driving force of the bifunctional ERα agonist and ERß antagonist activities of BPAF.


Subject(s)
Benzhydryl Compounds/pharmacology , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogens/pharmacology , Phenols/pharmacology , Benzhydryl Compounds/chemical synthesis , Benzhydryl Compounds/chemistry , Dose-Response Relationship, Drug , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/chemical synthesis , Estrogens/chemistry , HeLa Cells , Humans , Molecular Structure , Phenols/chemical synthesis , Phenols/chemistry , Structure-Activity Relationship
5.
Bioorg Chem ; 95: 103500, 2020 01.
Article in English | MEDLINE | ID: mdl-31869665

ABSTRACT

The structure-guided virtual screening (VS) has proved to be successful strategy in identification of new scaffolds for biological targets. The overactivity of NLRP3 inflammasome has been implicated in variety of inflammatory diseases including Alzheimer's disease. The up-regulation of estrogen-receptor ß (ER-ß) activity has been directly linked with inhibition of NLRP3 inflammasome activity. In the present study, we report discovery of new NLRP3 inflammasome inhibitors via ER-ß crystal structure (PDB: 5TOA) guided virtual screening of 20,000 compound library. For experimental validation, top 10 ligands were selected based on structure novelty, docking score, prime MMGB/SA binding affinity and interaction pattern analysis. Amongst the tested compounds, three thiazolidin-4-ones IIIM-1268, IIIM-1269 and IIIM-1270 and benzo[cd]indol-2-one IIIM-1266 have shown 73, 69, 75 and 77% suppression of IL-1ß release in mouse macrophages (J774A.1 cells) at 10 µM. Benzylidene-thiazolidine-2,4-diones IIIM-1268 and IIIM-1270 inhibited IL-1ß release with IC50 of 2.3 and 3.5 µM and also significantly decreased the protein expression level of mature form of IL-1ß in western-blot analysis. IIIM-1266 and IIIM-1270 displayed bidentate H-bonding with Arg 346 and Glu 305 residues in the active site of ER-ß; and they also strongly occupied the ADP-binding site of NLRP3 protein. The results presented herein, indicate that ER-ß guided VS can be successfully used to identify new NLRP3 inflammasome inhibitors, which may have potential in the development of novel anti-Alzheimer agents.


Subject(s)
Benzylidene Compounds/pharmacology , Drug Discovery , Estrogen Receptor beta/antagonists & inhibitors , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Thiazolidinediones/pharmacology , Animals , Benzylidene Compounds/chemical synthesis , Benzylidene Compounds/chemistry , Cell Line , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Estrogen Receptor beta/metabolism , Inflammasomes/metabolism , Mice , Molecular Structure , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/chemistry
6.
Bioorg Chem ; 94: 103380, 2020 01.
Article in English | MEDLINE | ID: mdl-31757413

ABSTRACT

Drug discovery and development have gained momentum due to the rational drug design by engaging computational tools and bioinformatics methodologies. Bioisosteric replacements and hybrid molecular approaches are the other inventive processes, used by medicinal chemists for the desired modifications of leads for clinical drug candidates. SERMs, ought to produce inhibitory activity in breast, uterus and agonist activity in other tissues, are beneficial for estrogen-like actions. ER subtypes α and ß are hormone dependent modulators of intracellular signaling and gene expression, and development of ER selective ligands could be an effective approach for treatment of breast cancer. This report has critically investigated the possible designing considerations of SERMs, their in silico interactions, and potent pharmacophore generation approaches viz. indole, restricted benzothiophene [3, 2-b] indole, carborane, xanthendione, combretastatin A-4, organometallic heterocycles, OBHS-SAHA hybrids, benzopyranones, tetrahydroisoquinolines, Dig G derivatives and their specifications in drug design and development, to rationally improve the understanding in drug discovery. This also includes various strategies for the development of dual inhibitors for the management of antiestrogenic resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Design , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Dose-Response Relationship, Drug , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Female , Humans , Molecular Structure , Selective Estrogen Receptor Modulators/chemistry , Structure-Activity Relationship
7.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998232

ABSTRACT

Estrogenic steroids and adenosine A2A receptors promote the wound healing and angiogenesis processes. However, so far, it is unclear whether estrogen may regulate the expression and pro-angiogenic activity of A2A receptors. Using in vivo analyses, we showed that female wild type (WT) mice have a more rapid wound healing process than female or male A2A-deficient mice (A2AKO) mice. We also found that pulmonary endothelial cells (mPEC) isolated from female WT mice showed higher expression of A2A receptor than mPEC from male WT mice. mPEC from female WT mice were more sensitive to A2A-mediated pro-angiogenic response, suggesting an ER and A2A crosstalk, which was confirmed using cells isolated from A2AKO. In those female cells, 17ß-estradiol potentiated A2A-mediated cell proliferation, an effect that was inhibited by selective antagonists of estrogen receptors (ER), ERα, and ERß. Therefore, estrogen regulates the expression and/or pro-angiogenic activity of A2A adenosine receptors, likely involving activation of ERα and ERß receptors. Sexual dimorphism in wound healing observed in the A2AKO mice process reinforces the functional crosstalk between ER and A2A receptors.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Neovascularization, Physiologic/drug effects , Receptor, Adenosine A2A/genetics , Wounds, Penetrating/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Female , Gene Expression Regulation , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Phenethylamines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor Cross-Talk , Receptor, Adenosine A2A/metabolism , Sex Factors , Signal Transduction , Wound Healing/drug effects , Wound Healing/genetics , Wounds, Penetrating/drug therapy , Wounds, Penetrating/metabolism , Wounds, Penetrating/pathology
8.
Molecules ; 25(12)2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32580515

ABSTRACT

An estrogen deficiency is the main cause of osteoporosis in postmenopausal women. In bone remodeling, estrogen receptors (ERs) can mediate estrogen-transducing signals. Methylpiperidinopyrazole (MPP) is a highly specific antagonist of ER-alpha (ERα). This study was designed to evaluate the effects of MPP on estrogen-induced energy production, subsequent osteoblast maturation, and the possible mechanisms. Exposure of primary osteoblasts isolated from neonatal rat calvarias to MPP did not affect cell morphology or survival. Estradiol can induce translocation of ERα into mitochondria from the cytoplasm. Interestingly, pretreatment of rat calvarial osteoblasts with MPP lowered estrogen-induced ERα translocation. Sequentially, estrogen-triggered expressions of mitochondrial energy production-linked cytochrome c oxidase (COX) I and COX II messenger (m)RNAs were inhibited following pretreatment with MPP. Consequently, MPP caused decreases in estrogen-triggered augmentation of the activities of mitochondrial respiratory complex enzymes and levels of cellular adenosine phosphate (ATP). During progression of osteoblast maturation, estrogen induced bone morphogenetic protein (BMP)-6 and type I collagen mRNA expressions, but MPP treatment inhibited such induction. Consequently, estrogen-induced osteoblast activation and mineralization were attenuated after exposure to MPP. Taken together, MPP suppressed estrogen-induced osteoblast maturation through decreasing chromosomal osteogenesis-related BMP-6 and type I collagen mRNA expressions and mitochondrial ATP synthesis due to inhibiting energy production-linked COX I and II mRNA expressions. MPP can appropriately be applied to evaluate estrogen-involved bioenergetics and osteoblast maturation.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogens/genetics , Osteoporosis/drug therapy , Pyrazoles/pharmacology , Animals , Bone Morphogenetic Protein 6/genetics , Cell Differentiation/drug effects , Electron Transport Complex IV/genetics , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Estrogens/metabolism , Female , Gene Expression Regulation/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Osteoblasts/drug effects , Osteocalcin/genetics , Osteogenesis/drug effects , Osteoporosis/metabolism , Osteoporosis/pathology , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Rats , Signal Transduction/drug effects
9.
Cell Physiol Biochem ; 52(3): 468-485, 2019.
Article in English | MEDLINE | ID: mdl-30873822

ABSTRACT

BACKGROUND/AIMS: Breast cancer is a clinically and molecularly heterogeneous disease. Patients with triple-negative breast cancer (TNBC) have poorer outcomes than those with other breast cancer subtypes due to lack of effective molecular targets for therapy. The present study aimed to the identification of estrogen receptor (ER)ß as a novel mitochondrial target in TNBC cells, together with underlying mechanisms. METHODS: Expression of ERß in clinical breast samples were examined by qRT-PCR, immunohistochemistry and immunoblotting. Subcellular distribution and binding of ERß-Grp75 was determined by confocal microscopic analysis, co-immunoprecipitation experiments, and limited-detergent extraction of subcellular organelles. The effect of mitocondrial ERß(mitoERß) overexpression on cell proliferation and cell cycle distribution were assessed CCK-8 assays and FACS. Mitochondrial ROS, membrane potential, and Ca²âº level were measured using the specific fluorescent probes Mito-Sox, TMRE, and Rhod-2AM. The tumorigenic effect of mitoERß overexpression was assessed using an anchorage-independent growth assay, sphere formation and a mouse orthotopic xenograft model. RESULTS: ERß expression was lower in tumor tissue than in adjacent normal tissue of patients with breast cancer, and low levels of mitochondrial ERß (mitoERß) also were associated with increased tumor recurrence after surgery. Overexpression of mitoERß inhibited the proliferation of TNBC cells and tumor masses in an animal model. Moreover, overexpression of mitoERß increased ATP production in TNBC cells and normal breast MCF10A cells, with the latter completely reversed by mitoERß knockdown in MCF10A cells. Grp75 was found to positively regulate ERß translocation into mitochondria via a direct interaction. Coimmunoprecipitation and subcellular fractionation experiments revealed that ERß-Grp75 complex is stable in mitochondria. CONCLUSION: These results suggest that the up-regulation of mitoERß in TNBC cells ensures proper mitochondrial transcription, activating the OXPHOS system to produce ATP. Studying the effects of mitoERß on mitochondrial activity and specific mitochondrial gene expression in breast cancer might help predict tumor recurrence, inform clinical decision-making, and identify novel drug targets in the treatment of TNBC.


Subject(s)
Adenosine Triphosphate/biosynthesis , Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic , HSP70 Heat-Shock Proteins/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Calcium/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Female , Fluorescent Dyes/chemistry , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/metabolism , Humans , Mice , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Neoplasm Staging , Oxidative Phosphorylation , Protein Binding , Protein Transport , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Cell Physiol Biochem ; 53(1): 186-199, 2019.
Article in English | MEDLINE | ID: mdl-31278696

ABSTRACT

BACKGROUND/AIMS: Estrogen could play a key role in the mechanisms underlying sex-related disparity in the incidence of thrombotic events. We investigated whether estrogen receptors (ERs) were expressed in human red blood cells (RBCs), and if they affected cell signaling of erythrocyte constitutive isoform of endothelial NO-synthase (eNOS) and nitric oxide (NO) release. METHODS: RBCs from 29 non-smoker volunteers (15 males and 14 females) aged between 20 and 40 years were analyzed by cytometry and western blot. In particular, content and distribution of ER-α and ER-ß, tyrosine kinases and eNOS phosphorylation and NO release were analyzed. RESULTS: We demonstrated that: i) both ER-α and ER-ß were expressed by RBCs; ii) they were both functionally active; and iii) ERs distribution and function were different in males and females. In particular, ERs modulated eNOS phosphorylation and NO release in RBCs from both sexes, but they induced the phosphorylation of specific tyrosine residues of kinases linked to eNOS activation and NO release in the RBCs from females only. CONCLUSION: Collectively, these data suggest that ERs could play a critical role in RBC intracellular signaling. The possible implication of this signaling in sex-linked risk disparity in human cardiovascular diseases, e.g. in thrombotic events, may not be ruled out.


Subject(s)
Receptors, Estrogen/metabolism , Signal Transduction , Adult , Dronabinol/pharmacology , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Female , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects , Piperidines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Signal Transduction/drug effects , Young Adult
11.
Hum Reprod ; 34(4): 646-658, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30838396

ABSTRACT

STUDY QUESTION: How is the activation of estrogen receptor ß (ERß) in endometriotic stromal cells (ESCs) involved in macrophage recruitment to promote the pathogenesis of endometriosis? SUMMARY ANSWER: ERß modulates the production of C-C motif chemokine ligand 2 (CCL2) via nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling in ESCs and thus recruits macrophages to ectopic lesions to promote pathogenesis. WHAT IS KNOWN ALREADY: Macrophages are mainly recruited to the peritoneal cavity to promote the pathogenesis of endometriosis. Recent studies have demonstrated that ERß plays an important role in the progression of endometriosis through modulating apoptosis and inflammation. STUDY DESIGN, SIZE, DURATION: An observational study consisting of 22 cases (women with endometriosis, diagnosed by laparoscopy and histological analysis) and 14 controls (without endometriosis) was carried out. PARTICIPANTS/MATERIALS, SETTING, METHODS: Tissues and stromal cells that were isolated from 22 patients with ovarian endometrioma and deeply infiltrating endometriosis were compared with tissues and stromal cells from 14 patients with normal cycling endometrium using immunohistochemistry, quantitative PCR, Western blot, cell migration assay and cloning formation assay. P values < 0.05 were considered significant, and experiments were repeated in at least three different cell preparations. MAIN RESULTS AND THE ROLE OF CHANCE: We observed that accumulated macrophages were recruited to the ectopic milieu and mainly adopted an alternatively activated macrophage (M2) phenotype. To reveal the underlying mechanism for this, we conducted a series of experiments and found that high expression of ERß led to the production of CCL2 via NF-κB signaling and macrophages were recruited to the ectopic milieu. An in vitro co-culture assay also suggested that the recruited macrophages in turn could promote the proliferation and clonogenic ability of ESCs. Overall, the activation of ERß in ESCs is involved in macrophage recruitment via NF-κB/CCL2 signaling and subsequently appears to promote the pathogenesis of endometriosis. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Due to the limitations of obtaining surgical specimens, endometrioma tissues were collected mainly from women diagnosed with middle to late stage endometriosis. We identified the predominant presence of M2 macrophages in the samples used in our study, but the underlying mechanism of how recruited macrophages acquire the M2 phenotype is undefined. WIDER IMPLICATIONS OF THE FINDINGS: This work provides novel insight into the mechanism by which ERß may modulate macrophage infiltration and promote pathogenesis, which may provide a new therapeutic target for endometriosis. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the National Natural Science Foundation of China (81671430). The authors have no conflicts of interest.


Subject(s)
Chemokine CCL2/metabolism , Endometriosis/metabolism , Endometrium/metabolism , Estrogen Receptor beta/metabolism , Macrophages/metabolism , NF-kappa B p50 Subunit/metabolism , Stromal Cells/metabolism , Adult , Animals , Cells, Cultured , Coculture Techniques , Endometriosis/pathology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Female , Humans , Mice , Mice, Inbred C57BL , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/genetics , THP-1 Cells , Transfection , Up-Regulation/genetics
12.
Cell Mol Neurobiol ; 39(8): 1139-1149, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31250245

ABSTRACT

We examined the role of the estrogen receptors alpha (ERα) and beta (ERß) in of the preoptic-anterior hypothalamic area (POA-AHA) in the regulation of ovulation in rats. The number of ERα- and ERß-immunoreactive (-ir) cells was determined at 09:00, 13:00, and 17:00 h of each stage of the estrous cycle in intact rats. Additionally, the effects of blocking ERα and ERß on ovulation rate at 09:00 h on diestrus-2 or proestrus day through the microinjection of methyl-piperidino-pyrazole (MPP) or cyclofenil in either side of POA-AHA were evaluated. The number of ERα-ir and ERß-ir cells in POA-AHA varied in each phase of estrous cycle. Either MPP or cyclofenil in the right side of POA-AHA on diestrus-2 day reduced the ovulation rate, while at proestrus day it was decreased in rats treated in either side with MPP, and in those treated with cyclofenil in the left side. MPP or cyclofenil produced a decrease in the surge of luteinizing hormone levels (LH) and an increase in progesterone and follicle stimulating hormone (FSH). Replacement with synthetic luteinizing hormone-releasing hormone in non-ovulating rats treated with MPP or cyclofenil restored ovulation. These results suggest that activation of estrogen receptors on the morning of diestrus-2 and proestrus day asymmetrically regulates ovulation and appropriately regulates the secretion of FSH and progesterone in the morning and afternoon of proestrus day. This ensures that both, the preovulatory secretion of LH and ovulation, occur at the right time.


Subject(s)
Anterior Hypothalamic Nucleus/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Ovulation , Preoptic Area/metabolism , Animals , Anterior Hypothalamic Nucleus/drug effects , Estradiol/blood , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/antagonists & inhibitors , Estrous Cycle/drug effects , Female , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/pharmacology , Neurons/drug effects , Neurons/metabolism , Ovulation/drug effects , Ovum/drug effects , Ovum/metabolism , Preoptic Area/drug effects , Progesterone/blood , Rats
13.
J Psychiatry Neurosci ; 44(1): 19, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30565903

ABSTRACT

Background: Estrogen therapy (ET), an effective treatment for perimenopausal depression, often fails to ameliorate symptoms when initiated late after the onset of menopause. Our previous work has suggested that alternative splicing of RNA might mediate these differential effects of ET. Methods: Female Sprague­Dawley rats were treated with estradiol (E2) or vehicle 6 days (early ET) or 180 days (late ET) after ovariectomy (OVX). We investigated the differential expression of RNA splicing factors and tryptophan hydroxylase 2 (TPH2) protein using a customized RT2 Profiler PCR Array, reverse-transcription polymerase chain reaction, immunoprecipitation and behaviour changes in clinically relevant early and late ET. Results: Early ET, but not late ET, prolonged swimming time in the forced swim test and reduced anxiety-like behaviours in the elevated plus maze. It reversed OVX-increased (SFRS7 and SFRS16) or OVX-decreased (ZRSR2 and CTNNB1) mRNA levels of splicing factors and ERß splicing changes in the brains of OVX rats. Early ET, but not late ET, also increased the expression of TPH2 and decreased monoamine oxidase A levels in the dorsal raphe in the brains of OVX rats. In late ET, only diarylpropionitrile (an ERß-specific agonist) achieved similar results ­ not E2 (an ERα and ERß agonist) or propylpyrazoletriol (an ERα-specific agonist). Limitations: Our experimental paradigm mimicked early and late ET in the clinical setting, but the contribution of age and OVX might be difficult to distinguish. Conclusion: These findings suggest that ERß alternative splicing and altered responses in the regulatory system for serotonin may mediate the antidepressant efficacy of ET associated with the timing of therapy initiation. It is likely that ERß-specific ligands would be effective estrogen-based antidepressants late after the onset of menopause.


Subject(s)
Antidepressive Agents/pharmacology , Estradiol/pharmacology , Immobility Response, Tonic/drug effects , Maze Learning/drug effects , RNA Splicing Factors/biosynthesis , Animals , Brain/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Female , Monoamine Oxidase/metabolism , Nitriles/pharmacology , Ovariectomy , Phenols/pharmacology , Propionates/pharmacology , Pyrazoles/pharmacology , Rats , Time Factors , Tryptophan Hydroxylase/biosynthesis
14.
Bioorg Med Chem Lett ; 29(16): 2290-2293, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31248773

ABSTRACT

Estrogen receptor (ER) exhibits two subtypes, ERα and ERß, whose biological functions are quite different despite expression in the same tissues. We developed diiodo-m-carborane derivative 3a, which showed 14-fold selectivity for ERß with high binding affinity toward ERß. Interestingly, introduction of an alkyl group into the carbon atom of the m-carborane cage of 3a markedly enhanced the binding affinity toward ERα and decreased affinity toward ERß. C-n-propyl derivative 3d showed 28-fold selectivity for ERα in an ER binding assay and promoted proliferation of MCF-7 breast cancer cells. Docking simulation studies suggest that the directions of the n-propyl group and the diiodo substituent introduced on the m-carborane cage play important roles for the control of ER subtype selectivity. As 3a and 3d showed ERß and ERα selectivity with high binding affinity, respectively, these ligands may be useful as biological tools to aid in understanding the different roles of ER subtypes.


Subject(s)
Boranes/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Phenols/pharmacology , Antineoplastic Agents , Binding Sites/drug effects , Boranes/chemical synthesis , Boranes/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/metabolism , Humans , Ligands , MCF-7 Cells , Molecular Docking Simulation , Molecular Structure , Phenols/chemical synthesis , Phenols/chemistry , Structure-Activity Relationship
15.
Biochim Biophys Acta Bioenerg ; 1859(6): 423-433, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29550215

ABSTRACT

Recent evidence highlights a role for sex and hormonal status in regulating cellular responses to ischemic brain injury and neurodegeneration. A key pathological event in ischemic brain injury is the opening of a mitochondrial permeability transition pore (MPT) induced by excitotoxic calcium levels, which can trigger irreversible damage to mitochondria accompanied by the release of pro-apoptotic factors. However, sex differences in brain MPT modulation have not yet been explored. Here, we show that mitochondria isolated from female mouse forebrain have a lower calcium threshold for MPT than male mitochondria, and that this sex difference depends on the MPT regulator cyclophilin D (CypD). We also demonstrate that an estrogen receptor beta (ERß) antagonist inhibits MPT and knockout of ERß decreases the sensitivity of mitochondria to the CypD inhibitor, cyclosporine A. These results suggest a functional relationship between ERß and CypD in modulating brain MPT. Moreover, co-immunoprecipitation studies identify several ERß binding partners in mitochondria. Among these, we investigate the mitochondrial ATPase as a putative site of MPT regulation by ERß. We find that previously described interaction between the oligomycin sensitivity-conferring subunit of ATPase (OSCP) and CypD is decreased by ERß knockout, suggesting that ERß modulates MPT by regulating CypD interaction with OSCP. Functionally, in primary neurons and hippocampal slice cultures, modulation of ERß has protective effects against glutamate toxicity and oxygen glucose deprivation, respectively. Taken together, these results reveal a novel pathway of brain MPT regulation by ERß that could contribute to sex differences in ischemic brain injury and neurodegeneration.


Subject(s)
Adenosine Triphosphatases/genetics , Carrier Proteins/genetics , Cyclophilins/genetics , Estrogen Receptor beta/genetics , Hippocampus/metabolism , Membrane Proteins/genetics , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Prosencephalon/metabolism , Adenosine Triphosphatases/metabolism , Animals , COS Cells , Calcium/metabolism , Carrier Proteins/metabolism , Chlorocebus aethiops , Peptidyl-Prolyl Isomerase F , Cyclophilins/antagonists & inhibitors , Cyclophilins/deficiency , Cyclosporine/pharmacology , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/deficiency , Female , Hippocampus/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtomy , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases , Piperidines/pharmacology , Prosencephalon/drug effects , Protein Binding , Pyrazoles/pharmacology , Sex Factors , Tissue Culture Techniques
16.
J Biol Chem ; 292(44): 18178-18191, 2017 11 03.
Article in English | MEDLINE | ID: mdl-28893903

ABSTRACT

Unlike age-matched men, premenopausal women benefit from cardiovascular protection. Estrogens protect against apoptosis of endothelial cells (ECs), one of the hallmarks of endothelial dysfunction leading to cardiovascular disorders, but the underlying molecular mechanisms remain poorly understood. The inflammatory cytokine TNFα causes EC apoptosis while dysregulating the Notch pathway, a major contributor to EC survival. We have previously reported that 17ß-estradiol (E2) treatment activates Notch signaling in ECs. Here, we sought to assess whether in TNFα-induced inflammation Notch is involved in E2-mediated protection of the endothelium. We treated human umbilical vein endothelial cells (HUVECs) with E2, TNFα, or both and found that E2 counteracts TNFα-induced apoptosis. When Notch1 was inhibited, this E2-mediated protection was not observed, whereas ectopic overexpression of Notch1 diminished TNFα-induced apoptosis. Moreover, TNFα reduced the levels of active Notch1 protein, which were partially restored by E2 treatment. Moreover, siRNA-mediated knockdown of estrogen receptor ß (ERß), but not ERα, abolished the effect of E2 on apoptosis. Additionally, the E2-mediated regulation of the levels of active Notch1 was abrogated after silencing ERß. In summary, our results indicate that E2 requires active Notch1 through a mechanism involving ERß to protect the endothelium in TNFα-induced inflammation. These findings could be relevant for assessing the efficacy and applicability of menopausal hormone treatment, because they may indicate that in women with impaired Notch signaling, hormone therapy might not effectively protect the endothelium.


Subject(s)
Apoptosis , Endothelium, Vascular/metabolism , Estradiol/metabolism , Estrogen Receptor beta/agonists , Receptor, Notch1/agonists , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Apoptosis/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protease Inhibitors/pharmacology , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, Notch1/chemistry , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
17.
Cell Physiol Biochem ; 50(5): 1638-1658, 2018.
Article in English | MEDLINE | ID: mdl-30384372

ABSTRACT

BACKGROUND/AIMS: Accumulating evidences have demonstrated that long noncoding RNAs (lncRNA) play important roles in hepatic lipid metabolism in mammals. However, no systematic screening of the potential lncRNAs in the livers of laying hens has been performed, and few studies have been reported concerning the effects of the lncRNAs on lipid metabolism in the livers of chickens during egg-laying period. The purpose of this study was to compare the difference in lncRNA expression in the livers of pre-laying and peak-laying hens at the age of 20 and 30 weeks old by transcriptome sequencing and to investigate the interaction networks among lncRNAs, mRNAs and miRNAs. Moreover, the regulatory mechanism and biological function of lncLTR, a significantly differentially expressed lncRNA in the liver between pre- and peak-laying hens, was explored in vitro and in vivo. METHODS: Bioinformatics analyses were conducted to identify the differentially expressed (DE) lncRNAs between the two groups of hens. The target genes of the DE lncRNA were predicated for further functional enrichment. An integrated analysis was performed among the DE lncRNA datasets, DE mRNAs and DE miRNA datasets obtained from the same samples to predict the interaction relationship. In addition, in vivo and in vitro trials were carried out to determine the expression regulation of lncLTR, and polymorphism association analysis was conducted to detect the biological role of ncLTR. RESULTS: A total of 124 DE lncRNAs with a P-value ≤ 0.05 were identified. Among them, 44 lncRNAs including 30 known and 14 novel lncRNAs were significant differentially expressed (SDE) with FDR ≤ 0.05. Thirty-two lncRNAs were upregulated and 12 were downregulated in peak-laying group compared with pre-laying group. The functional enrichment results revealed that target genes of some lncRNAs are involved in the lipid metabolism process. Integrated analysis suggested that some of the genes involved in lipid metabolism might be regulated by both the lncRNA and the miRNA. In addition, an upregulated lncRNA, designated lncLTR, was demonstrated to be induced by estrogen via ERß signaling. The c242. G>A SNP in lncLTR was significantly associated with chicken carcass weight, evisceration weight, semi-evisceration weight, head weight, double-wing weight, claw weight traits, and blood biochemical index, especially for the blood triglyceride content. CONCLUSION: A series of lncRNAs associated with lipid metabolism in the livers of chickens were identified by transcriptome sequencing and functional analysis, providing a valuable data resource for further studies on chicken hepatic metabolism activities. LncLTR was regulated by estrogen via ERß signaling and associated with chicken carcass trait and blood triglyceride content.


Subject(s)
Estradiol/pharmacology , Lipid Metabolism/genetics , RNA, Long Noncoding/metabolism , Transcriptome/drug effects , Animals , Chick Embryo , Chickens/genetics , Chickens/metabolism , Down-Regulation/drug effects , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Polymorphism, Genetic , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects
18.
Toxicol Appl Pharmacol ; 345: 19-25, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29499249

ABSTRACT

Many diseases and disorders are linked to exposure to endocrine disrupting chemicals (EDCs) that mimic the function of natural estrogen hormones. Here we present a Rapid Adaptable Portable In-vitro Detection biosensor platform (RAPID) for detecting chemicals that interact with the human estrogen receptor ß (hERß). This biosensor consists of an allosteric fusion protein, which is expressed using cell-free protein synthesis technology and is directly assayed by a colorimetric response. The resultant biosensor successfully detected known EDCs of hERß (BPA, E2, and DPN) at similar or better detection range than an analogous cell-based biosensor, but in a fraction of time. We also engineered cell-free protein synthesis reactions with RNAse inhibitors to increase production yields in the presence of human blood and urine. The RAPID biosensor successfully detects EDCs in these human samples in the presence of RNAse inhibitors. Engineered cell-free protein synthesis facilitates the use of protein biosensors in complex sample matrices without cumbersome protein purification.


Subject(s)
Biosensing Techniques/methods , Cell-Free System/metabolism , Endocrine Disruptors/blood , Endocrine Disruptors/urine , Protein Biosynthesis/physiology , Cell-Free System/drug effects , Dose-Response Relationship, Drug , Endocrine Disruptors/pharmacology , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Humans , Protein Biosynthesis/drug effects
19.
Article in English | MEDLINE | ID: mdl-29080952

ABSTRACT

Elevations of sex steroids induced by social cues can rapidly modulate social behavior, but we know little about where they act within the nervous system to produce such effects. In male goldfish, testosterone (T) rapidly increases approach responses to the visual cues of females through its conversion to estradiol. Because aromatase is expressed in the retina, we tested if T can acutely influence retina responses to visual stimuli, and investigated the receptor mechanisms that may mediate such effects. Specifically, we measured FOS protein immunoreactivity to determine if T affects cellular responses to visual stimuli that include females, and used electrophysiology to investigate whether T can generally affect light sensitivity. We found that T acutely increased FOS responses to the simultaneous onset of light and the presence of female visual stimuli, both of which would normally be associated with early morning spawning, and increased electrophysiological responses to low intensity light pulses. Both effects were blocked by an estrogen receptor beta (ERß) antagonist, indicating that T is likely being converted to estradiol (E2) and acting through an ERß mediated mechanism to acutely modulate visual processing. Changes in sensory processing could subsequently influence approach behavior to increase reproductive success in competitive mating environments.


Subject(s)
Estrogen Receptor beta/metabolism , Fish Proteins/metabolism , Goldfish/metabolism , Retina/metabolism , Testosterone/metabolism , Visual Perception/physiology , Animals , Competitive Behavior/physiology , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor beta/antagonists & inhibitors , Female , Fish Proteins/antagonists & inhibitors , Male , Proto-Oncogene Proteins c-fos/metabolism , Reproduction/physiology , Retina/drug effects , Sexual Behavior, Animal/physiology , Visual Perception/drug effects
20.
J Mol Cell Cardiol ; 107: 41-51, 2017 06.
Article in English | MEDLINE | ID: mdl-28457941

ABSTRACT

Steroid hormone receptors including estrogen receptors (ER) classically function as ligand-regulated transcription factors. However, estrogens also elicit cellular effects through binding to extra-nuclear ER (ERα, ERß, and G protein-coupled ER or GPER) that are coupled to kinases. How extra-nuclear ER actions impact cardiac ischemia-reperfusion (I/R) injury is unknown. We treated ovariectomized wild-type female mice with estradiol or an estrogen-dendrimer conjugate (EDC), which selectively activates extra-nuclear ER, or vehicle interventions for two weeks. I/R injury was then evaluated in isolated Langendorff perfused hearts. Two weeks of treatment with estradiol significantly decreased infarct size and improved post-ischemic contractile function. Similarly, EDC treatment significantly decreased infarct size and increased post-ischemic functional recovery compared to vehicle-treated hearts. EDC also caused an increase in myocardial protein S-nitrosylation, consistent with previous studies showing a role for this post-translational modification in cardioprotection. In further support of a role for S-nitrosylation, inhibition of nitric oxide synthase, but not soluble guanylyl cyclase blocked the EDC mediated protection. The administration of ICI182,780, which is an agonist of G-protein coupled estrogen receptor (GPER) and an antagonist of ERα and ERß, did not result in protection; however, ICI182,780 significantly blocked EDC-mediated cardioprotection, indicating participation of ERα and/or ERß. In studies determining the specific ER subtype and cellular target involved, EDC decreased infarct size and improved functional recovery in mice lacking ERα in cardiomyocytes. In contrast, protection was lost in mice deficient in endothelial cell ERα. Thus, extra-nuclear ERα activation in endothelium reduces cardiac I/R injury in mice, and this likely entails increased protein S-nitrosylation. Since EDC does not stimulate uterine growth, in the clinical setting EDC-like compounds may provide myocardial protection without undesired uterotrophic and cancer-promoting effects.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Ischemia/genetics , Reperfusion Injury/genetics , Animals , Endothelium/metabolism , Endothelium/pathology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/antagonists & inhibitors , Estrogens/genetics , Estrogens/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Ischemia/metabolism , Ischemia/pathology , Mice , Ovariectomy , Protein Processing, Post-Translational/drug effects , Receptors, Estrogen/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL