• Title/Summary/Keyword: Estrogen pathway

Search Result 69, Processing Time 0.029 seconds

Reconstruction and Exploratory Analysis of mTORC1 Signaling Pathway and Its Applications to Various Diseases Using Network-Based Approach

  • Buddham, Richa;Chauhan, Sweety;Narad, Priyanka;Mathur, Puniti
    • Journal of Microbiology and Biotechnology
    • /
    • v.32 no.3
    • /
    • pp.365-377
    • /
    • 2022
  • Mammalian target of rapamycin (mTOR) is a serine-threonine kinase member of the cellular phosphatidylinositol 3-kinase (PI3K) pathway, which is involved in multiple biological functions by transcriptional and translational control. mTOR is a downstream mediator in the PI3K/Akt signaling pathway and plays a critical role in cell survival. In cancer, this pathway can be activated by membrane receptors, including the HER (or ErbB) family of growth factor receptors, the insulin-like growth factor receptor, and the estrogen receptor. In the present work, we congregated an electronic network of mTORC1 built on an assembly of data using natural language processing, consisting of 470 edges (activations/interactions and/or inhibitions) and 206 nodes representing genes/proteins, using the Cytoscape 3.6.0 editor and its plugins for analysis. The experimental design included the extraction of gene expression data related to five distinct types of cancers, namely, pancreatic ductal adenocarcinoma, hepatic cirrhosis, cervical cancer, glioblastoma, and anaplastic thyroid cancer from Gene Expression Omnibus (NCBI GEO) followed by pre-processing and normalization of the data using R & Bioconductor. ExprEssence plugin was used for network condensation to identify differentially expressed genes across the gene expression samples. Gene Ontology (GO) analysis was performed to find out the over-represented GO terms in the network. In addition, pathway enrichment and functional module analysis of the protein-protein interaction (PPI) network were also conducted. Our results indicated NOTCH1, NOTCH3, FLCN, SOD1, SOD2, NF1, and TLR4 as upregulated proteins in different cancer types highlighting their role in cancer progression. The MCODE analysis identified gene clusters for each cancer type with MYC, PCNA, PARP1, IDH1, FGF10, PTEN, and CCND1 as hub genes with high connectivity. MYC for cervical cancer, IDH1 for hepatic cirrhosis, MGMT for glioblastoma and CCND1 for anaplastic thyroid cancer were identified as genes with prognostic importance using survival analysis.

Ginsenoside Rg1 enhances the healing of injured tendon in achilles tendinitis through the activation of IGF1R signaling mediated by oestrogen receptor

  • Wu, Tianyi;Qi, Wenxiao;Shan, Haojie;Tu, Bin;Jiang, Shilin;Lu, Ye;Wang, Feng
    • Journal of Ginseng Research
    • /
    • v.46 no.4
    • /
    • pp.526-535
    • /
    • 2022
  • Background: During the pathogenesis of tendinopathy, the chronic inflammation caused by the injury and apoptosis leads to the generation of scars. Ginsenoside Rg1 (Rg1) is extracted from ginseng and has anti-inflammatory effects. Rg1 is a unique phytoestrogen that can activate the estrogen response element. This research aimed to explore whether Rg1 can function in the process of tendon repair through the estrogen receptor. Methods: In this research, the effects of Rg1 were evaluated in tenocytes and in a rat model of Achilles tendinitis (AT). Protein levels were shown by western blotting. qRT-PCR was employed for evaluating mRNA levels. Cell proliferation was evaluated through EdU assay and cell migration was evaluated by transwell assay and scratch test assay. Results: Rg1 up-regulated the expression of matrix-related factors and function of tendon in AT rat model. Rg1 reduced early inflammatory response and apoptosis in the tendon tissue of AT rat model. Rg1 promoted tenocyte migration and proliferation. The effects of Rg1 on tenocytes were inhibited by ICI182780. Rg1 activates the insulin-like growth factor-I receptor (IGF1R) and MAPK signaling pathway. Conclusion: Rg1 promotes injured tendon healing in AT rat model through IGF1R and MAPK signaling pathway activation.

Regulation of Estrogen Receptor Under Hypoxia in Breast Cancer Cells

  • Lee, Young-Joo
    • Proceedings of the Korean Society of Applied Pharmacology
    • /
    • 2008.04a
    • /
    • pp.55-74
    • /
    • 2008
  • Previously, we have shown that hypoxia, through HIF-1, induces ligand-independent $ER{\alpha}$ activation and the physical interaction of HIF-1 and $ER{\alpha}$. However, the effect of hypoxia on the transactivation of $ER{\beta}$ is not yet known. In the present study, we found that hypoxia activated the $ER{\beta}$-mediated transcriptional response in the HEK 293 cell line, as determined by the transient expression of$ER{\beta}$ and ER-responsive reporter plasmids. The hypoxia-induced estrogen response element-mediated transcriptional response was dependent on $ER{\beta}$ expression and was inhibited by the ER antagonist ICI 182,780. Transactivation of $ER{\beta}$ was induced by the expression of HIF-$1{\alpha}$ under normoxic conditions, as determined by the expression of oxygen-independent stable GFP-HIF-$1{\alpha}$. HIF-$1{\alpha}$-induced $ER{\beta}$ transactivation was abolished by the inhibition of HIF-$1{\alpha}$ activation. This was determined by using chemical inhibitors for the MAPK pathway. In addition, HIF-$1{\alpha}$ interacted with $ER{\beta}$ in a mammalian-two hybrid assay. We conclude that hypoxia activates $ER{\beta}$ in a ligand-independent manner, possibly through the interaction of HIF-$1{\alpha}$ and $ER{\beta}$.

  • PDF

Raloxifene, a Selective Estrogen Receptor Modulator, Inhibits Lipopolysaccharide-induced Nitric Oxide Production by Inhibiting the Phosphatidylinositol 3-Kinase/Akt/Nuclear Factor-kappa B Pathway in RAW264.7 Macrophage Cells

  • Lee, Sin-Ae;Park, Seok Hee;Kim, Byung-Chul
    • Molecules and Cells
    • /
    • v.26 no.1
    • /
    • pp.48-52
    • /
    • 2008
  • We here demonstrate an anti-inflammatory action of raloxifene, a selective estrogen receptor modulator, in lipopolysaccharide (LPS)-induced murine macrophage RAW264.7 cells. Treatment with raloxifene at micromolar concentrations suppressed the production of nitric oxide (NO) by down-regulating expression of the inducible nitric oxide synthase (iNOS) gene in LPS-activated cells. The decreased expression of iNOS and subsequent reduction of NO were due to inhibition of nuclear translocation of transcription factor NF-${\kappa}B$. These effects were significantly inhibited by exposure to the phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor, LY294002, or by expression of a dominant negative mutant of PI 3-kinase. In addition, pretreatment with raloxifene reduced LPS-induced Akt phosphorylation as well as NF-${\kappa}B$ DNA binding activity and NF-${\kappa}B$-dependent reporter gene activity. Thus our findings indicate that raloxifene exerts its anti-inflammatory action in LPS-stimulated macrophages by blocking the PI 3-kinase-Akt-NF-${\kappa}B$ signaling cascade, and eventually reduces expression of pro-inflammatory genes such as iNOS.

TNFα-induced Down-Regulation of Estrogen Receptor α in MCF-7 Breast Cancer Cells

  • Lee, Sang-Han;Nam, Hae-Seon
    • Molecules and Cells
    • /
    • v.26 no.3
    • /
    • pp.285-290
    • /
    • 2008
  • Estrogen-induced proliferation in estrogen receptor (ER)-positive breast cancer cells is primarily mediated through two distinct intracellular receptors, $ER{\alpha}$ and $ER{\beta}$. Although tumor necrosis factor alpha ($TNF{\alpha}$) and $E2/ER{\alpha}$ are known to exert opposing effects on cell proliferation in MCF-7 cells, the mechanism by which $TNF{\alpha}$ antagonizes $E2/ER{\alpha}$-mediated cell proliferation is not well understood. The present study suggests that reduced cell survival in response to $TNF{\alpha}$ treatment in MCF-7 cells may be associated with the down-regulation of $ER{\alpha}$ protein. The decrease in $ER{\alpha}$ protein level was accompanied by an inhibition of $ER{\alpha}$ gene transcription. Cell viability was decreased synergistically by the combined treatment with $ER{\alpha}$-siRNA and $TNF{\alpha}$. Furthermore, pretreatment of cells with the PI3-kinase (PI3K)/ Akt inhibitor, LY294002, markedly enhanced $TNF{\alpha}$-induced down-regulation of the $ER{\alpha}$ protein, suggesting that the PI3K/Akt pathway might be involved in control of the $ER{\alpha}$ level. Moreover, down-regulation of $ER{\alpha}$ by $TNF{\alpha}$ was not inhibited in cells that were pretreated with the proteasome inhibitors, MG132 and MG152, which suggests that proteasome-dependent proteolysis does not significantly influence $TNF{\alpha}$-induced down-regulation of $ER{\alpha}$ protein. In contrast, the effect of the PI3K/Akt inhibitor on $ER{\alpha}$ was blocked in cells that were treated with LY294002 in the presence of the proteasome inhibitors. Collectively, our findings show that the $TNF{\alpha}$ may partly regulate the growth of MCF-7 breast cancer cells through the down-regulation of $ER{\alpha}$ expression, which is primarily mediated by a PI3K/Akt signaling.

Study on the Estrogen Receptor Mediated Toxicity of Cadmium and Protective Effects of Antioxidant (에스트로겐 수용체를 통한 카드뮴 독성 및 항산화제에 의한 독성경감에 관한 연구)

  • Kim Tae-Sung;Kang Tae-Seok;Kang Ho-Il;Moon Hyun-Ju;Kang Il-Hyun;Lee Young-Joo;Choi Eun-Hee;Hong Jin-Tae;Han Soon-Young;Hong Jin-Hwan
    • Environmental Mutagens and Carcinogens
    • /
    • v.26 no.1
    • /
    • pp.12-19
    • /
    • 2006
  • Cadmium, a human carcinogen, can induce toxicity in various cell lines and organs. Despite extensive research, the mechanisms of cadmium-induced cell toxicity and estrogenic potential in human are not clear. This study was performed to investigate cadmium-induced toxicity on human breast cancer cells: MCF-7 cells, an estrogen receptor (ER) positive breast cancer cells, and MDA-MB-231 cells, an ER negative breast cancer cells. MCF-7 cells was proved to be more sensitive than the other cell lines (IC50 = $50\;{\mu}M$ at MCF-7 cells and $120{\mu}M$ at MDA-MB-231). The expression of JNK and AP-1 transcription factors such as c-Jun and c-Fos dependent transcription were increased by cadmium treatment. Inhibition of ER activation by ER antagonist (tamoxifen or ICI 182,780) significantly recovered the viablity and inhibited apoptotic cell death. This suggested that cadmium-induced cell death in ER (+) cells was mediated by JNK/AP-1 pathway and this pathway was more stimulated by ER activated by cadmium. Co-treatment of antioxidants such as selenium (Se), butylated hydroxyanisole (BHA), glutathione (GSH), or N-acetyl-L-cysteine (NAC) recovered the cadmium-induced cell death in MCF-7 cells. Cadmium-induced lipid peroxidation was decreased by GSH, NAC, or BHA in MCF-7 cells. The expression of SOD protein was decreased by cadmium ($100{\mu}M$) but recovered by GSH, NAC, BHA, or Se. Our data showed that the cadmium-induced cell toxicity in human breast cancer cells could be protected by the antioxidants (Se, BHA, NAC, GSH, or NAC) and ER antagonist (tamoxifen or ICI 182,780). Therefore, toxicity of cadmium in breast cancer were mediated by oxidative stress and $ER{\alpha}$.

  • PDF

Bacterial Lipopolysaccharides Induce Steroid Sulfatase Expression and Cell Migration through IL-6 Pathway in Human Prostate Cancer Cells

  • Im, Hee-Jung;Park, Na-Hee;Kwon, Yeo-Jung;Shin, Sangyun;Kim, Donghak;Chun, Young-Jin
    • Biomolecules & Therapeutics
    • /
    • v.20 no.6
    • /
    • pp.556-561
    • /
    • 2012
  • Steroid sulfatase (STS) is responsible for the conversion of estrone sulfate to estrone that can stimulate growth in endocrine-dependent tumors such as prostate cancer. Although STS is considered as a therapeutic target for the estrogen-dependent diseases, cellular function of STS are still not clear. Previously, we found that tumor necrosis factor (TNF)-${\alpha}$ significantly enhances steroid sulfatase expression in PC-3 human prostate cancer cells through PI3K/Akt-dependent pathways. Here, we studied whether bacterial lipopolysaccharides (LPS) which are known to induce TNF-${\alpha}$ may increase STS expression. Treatment with LPS in PC-3 cells induced STS mRNA and protein in concentration- and time-dependent manners. Using luciferase reporter assay, we found that LPS enhanced STS promoter activity. Moreover, STS expression induced by LPS increased PC-3 tumor cell migration determined by wound healing assay. We investigated that LPS induced IL-6 expression and IL-6 increased STS expression. Taken together, these data strongly suggest that LPS induces STS expression through IL-6 pathway in human prostate cancer cells.

Vasorelaxing Effect of Isoflavonoids Via Rho-kinase Inhibition in Agonist-Induced Vasoconstriction (Isoflavonoids에 의한 혈관이완효과에 있어 Rho-kinase의 역할)

  • Je, Hyun-Dong
    • YAKHAK HOEJI
    • /
    • v.50 no.4
    • /
    • pp.293-299
    • /
    • 2006
  • The aim of present study was to investigate the possible influence of Rho-kinase inhibition on the plant-derived estrogen-like compounds-induced arterial relaxation. Agonist- or depolarization-induced vascular smooth muscle contractions involve the activation of Rho-kinase pathway. However there are no reports addressing the question whether this pathway is involved in genistein-or daidzein-induced vascular relaxation in rat aortae precontracted with phenylephrine or thromboxane $A_2$ mimetic U-46619. We hypothesized that Rho-kinase inhibition plays a role in vascular relaxation evoked by genistein or daidzein in rat aortae. Endothelium-intact and denuded arterial rings from male Sprague-Dawley rats were used and isometric contractions were recorded using a computerized data acquisition system. Genistein concentration-dependently inhibited phenylephrine or thromboxane $A_2-induced$ contraction regardless of endothelial function. Surprisingly, in the agonists-induced contraction, similar results were also observed in aortae treated with daidzein, the inactive congener for protein tyrosine kinase inhibition, suggesting that Rho-kinase might act upstream of tyrosine kinases in phenylephrine-induced contraction. In conclusion, in the agonists-precontracted rat aortae, genistein and daidzein showed similar relaxant response regardless of tyrosine kinase inhibition or endothelial function.

Advances in the Development and Validation of Test Methods in the United States

  • Casey, Warren M.
    • Toxicological Research
    • /
    • v.32 no.1
    • /
    • pp.9-14
    • /
    • 2016
  • The National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods (NICEATM) provides validation support for US Federal agencies and the US Tox21 interagency consortium, an interagency collaboration that is using high throughput screening (HTS) and other advanced approaches to better understand and predict chemical hazards to humans and the environment. The use of HTS data from assays relevant to the estrogen receptor signaling data pathway is used as an example of how HTS data can be combined with computational modeling to meet the needs of US agencies. As brief summary of US efforts in the areas of biologics testing, acute toxicity, and skin sensitization will also be provided.

Anti-proliferative Effects of Daidzein, Baicalein, Hesperidin and Ursolic Acid on Human Breast Cancer Cells Stimulated by Estrogenic Compounds (에스트로겐성 물질에 의해 자극된 인간 유방암 세포 증식에 대한 다이드제인, 바이칼레인, 헤스페리딘 및 우르솔산의 억제 효과)

  • Lee, Mi-Nam;Lee, Su-Yel;Lee, Hyun-Jae;Seok, Jeong-Ho;Lee, Choong-Jae
    • YAKHAK HOEJI
    • /
    • v.54 no.3
    • /
    • pp.168-173
    • /
    • 2010
  • We investigated whether three flavonoids including daidzein, baicalein, hesperidin and ursolic acid, a triterpene acid, affect proliferation of MCF-7 human breast cancer cells stimulated by estrogenic compounds. Ursolic acid and baicalein inhibited proliferation of MCF-7 cells induced by PhIP, a food-derived carcinogen with estrogenic activity. Daidzein and hesperidin inhibited estradiol-induced proliferation of MCF-7 cells. These compounds should be further investigated for the possible involvement in signaling pathway after estrogen receptor binding in breast cancer cells.