• Title/Summary/Keyword: endometrial cell

Search Result 124, Processing Time 0.033 seconds

IL-17A and Th17 Cells Contribute to Endometrial Cell Survival by Inhibiting Apoptosis and NK Cell Mediated Cytotoxicity of Endometrial Cells via ERK1/2 Pathway

  • Young-Ju Kang;Hee Jun Cho;Yunhee Lee;Arum Park;Mi Jeong Kim;In Cheul Jeung;Yong-Wook Jung;Haiyoung Jung;Inpyo Choi;Hee Gu Lee;Suk Ran Yoon
    • IMMUNE NETWORK
    • /
    • v.23 no.2
    • /
    • pp.14.1-14.14
    • /
    • 2023
  • Immune status including the immune cells and cytokine profiles has been implicated in the development of endometriosis. In this study, we analyzed Th17 cells and IL-17A in peritoneal fluid (PF) and endometrial tissues of patients with (n=10) and without (n=26) endometriosis. Our study has shown increased Th17 cell population and IL-17A level in PF with endometriosis patients. To determine the roles of IL-17A and Th17 cells in the development of endometriosis, the effect of IL-17A, major cytokine of Th17, on endometrial cells isolated from endometriotic tissues was examined. Recombinant IL-17A promoted survival of endometrial cells accompanied by increased expression of anti-apoptotic genes, including Bcl-2 and MCL1, and the activation of ERK1/2 signaling. In addition, treatment of IL-17A to endometrial cells inhibited NK cell mediated cytotoxicity and induced HLA-G expression on endometrial cells. IL-17A also promoted migration of endometrial cells. Our data suggest that Th17 cells and IL-17A play critical roles in the development of endometriosis by promoting endometrial cell survival and conferring a resistance to NK cell cytotoxicity through the activation of ERK1/2 signaling. Targeting IL-17A has potential as a new strategy for the treatment of endometriosis.

Metformin Down-regulates Endometrial Carcinoma Cell Secretion of IGF-1 and Expression of IGF-1R

  • Zhang, Yu;Li, Meng-Xiong;Wang, Huan;Zeng, Zheng;Li, Xiao-Mao
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.1
    • /
    • pp.221-225
    • /
    • 2015
  • As metformin can inhibit endometrial carcinoma (EC) cell growth and the insulin growth factor (IGF) system is active in EC, the question of whether it can regulate endometrial carcinoma cell secretion of IGF-1 or expression of IGF-1 receptor (IGF-1R) is of interest. In this study, serum IGF-1 levels in EC patients were found to be comparable with that in the non EC patients (p>0.05). However, the IGF-1 level in the medium of cultured cells after treatment with metformin was decreased (p<0.05). IGF-1R was highly expressed in human endometrial carcinoma paraffin sections, but IGF-1R and phosphor-protein kinase B/protein kinase B (p-Akt/Akt) expression was down-regulated after metformin treatment (p<0.05). In summary, metformin can reduce the secretion of IGF-1 by Ishikawa and JEC EC cell lines and their expression of IGF-1R to deactivate downstream signaling involving the PI-3K/Akt pathway to inhibit endometrial carcinoma cell growth.

Effects of Keratinocyte Growth Factor on the Uterine Endometrial Epithelial Cells in Pigs

  • Ka, Hak-Hyun;Bazer, Fuller W.
    • Asian-Australasian Journal of Animal Sciences
    • /
    • v.18 no.12
    • /
    • pp.1708-1714
    • /
    • 2005
  • Keratinocyte growth factor (KGF) functions in epithelial growth and differentiation in many tissues and organs. KGF is expressed in the uterine endometrial epithelial cells during the estrous cycle and pregnancy in pigs, and receptors for KGF (KGFR) are expressed by conceptus trophectoderm and endometrial epithelia. KGF has been shown to stimulate the proliferation and differentiation of conceptus trophectoderm. However, the role of KGF on the endometrial epithelial cells has not been determined. Therefore, this study determined the effect of KGF on proliferation and differentiation of endometrial epithelial cells in vitro and in vivo using an immortalized porcine luminal epithelial (pLE) cell line and KGF infusion into the uterine lumen of pigs between Days 9 and 12 of estrous cycle. Results showed that KGF did not stimulate proliferation of uterine endometrial epithelial cells in vitro and in vivo determined by the $^3$H]thymidine incorporation assay and the proliferating cell nuclear antigen staining, respectively. Effects of KGF on expression of several markers for epithelial cell differentiation, including integrin receptor subunits $\alpha$4, $\alpha$5 and $\beta$1, plasmin/trypsin inhibitor, uteroferrin and retinol-binding protein were determined by RT-PCR, Northern and slot blot analyses, and immunohistochemisty, and KGF did not affect epithelial cell differentiation in vitro and in vivo. These results show that KGF does not induce epithelial cell proliferation and differentiation, suggesting that KGF produced by endometrial epithelial cells acts on conceptus trophectoderm in a paracrine manner rather than on endometrial epithelial cells in an autocrine manner.

Identification of a High-yield Technique for Isolating Endometrial Epithelial Cells from the Mouse Uterus : A Comparison of Mechanical and Sedimentation-adherence Methods

  • Sohn, Jie Ohn;Jo, Yoon Mi;Park, Hye Jin;Ahn, Ji Yeon;Song, Hyun Jin;Lim, Jeong Mook;Lee, Seung Tae
    • Journal of Embryo Transfer
    • /
    • v.31 no.1
    • /
    • pp.73-80
    • /
    • 2016
  • An in vitro assay following culture of endometrial epithelial cells is essential for understanding epithelial cell function in reproduction. Several diverse techniques have been developed for isolating endometrial epithelial cells, although an optimal technique has not been identified. In this study, we describe a sedimentation-adherence (S-A) isolation technique with a high-yield cell-separating ability to isolate endometrial epithelial cells from 8-week-old female C57BL/6 mice. We analyzed total cell number, viability, morphology, and expression of cytokeratin 18 as an endometrial epithelial cell-specific marker in cells isolated using a mechanical method compared to the S-A technique. There were no significant differences in the total number, viability, or morphology of the putative endometrial epithelial cells with either method. In contrast, significantly more endometrial epithelial cells harvested using the S-A method were positively stained for cytokeratin 18 than those isolated using the mechanical method. These results confirm that the S-A method is more efficient for retrieving endometrial epithelial cells than a mechanical method.

A case of canine bilateral ovary granulosa cell tumor and mammary complex carcinoma

  • Chung, Yung-Ho;Hong, Sunhwa;Han, Sang-Jun;Kim, Okjin
    • Korean Journal of Veterinary Service
    • /
    • v.36 no.2
    • /
    • pp.127-132
    • /
    • 2013
  • An 11-year-old poodle bitch was presented for investigation of multicentric mammary masses. Abdominal sonography and radiography demonstrated abnormal enlargement of uterus and ovaries. Blood analysis revealed high progesterone concentration. The ovariohysterectomy and mastectomy were performed. Histopathologically, the mammary masses revealed complex carcinoma-tubulopapillary carcinoma with papillary pattern and tubule pattern. In the uterus, cystic endometrial hyperplasia was observed. Scattered inflammatory cells were observed in the endometrial stroma and mucinous material was protruded from endometrial surface. Also, in the ovaries, bilateral ovary granulosa cell tumor was detected. The bitch made a complete recovery following the ovariohysterectomy and mastectomy. This case was a very rare multiple tumor occurrence with bilateral ovary granulosa cell tumor and mammary complex carcinoma. High progesterone concentration was characterized clinically in the bitch.

Endometrial Intraepithelial Neoplasia (EIN) in Endometrial Biopsy Specimens Categorized by the 1994 World Health Organization Classification for Endometrial Hyperplasia

  • Li, Xiao-Chao;Song, Wen-Jing
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.10
    • /
    • pp.5935-5939
    • /
    • 2013
  • Our study is to determine the presence of endometrial intraepithelial neoplasia (EIN) in endometrial biopsy specimens classified by the 1994 World Health Organization (WHO) criteria for endometrial hyperplasia. Endometrial biopsy specimens that were stained with hematoxylin and eosin (HE) were examined and categorized by the WHO 1994 criteria and for the presence of EIN as defined by the International Endometrial Collaborative Group. ${\beta}$-catenin expression was examined by immunohistochemistry. A total of 474 cases of HE stained endometrial biopsy tissues were reviewed. There were 379 cases of simple endometrial hyperplasia, 16 with simple atypical endometrial hyperplasia, 48 with complex endometrial hyperplasia, and 31 with complex atypical endometrial hyperplasia. Among the 474 endometrial hyperplasia cases, there were 46 (9.7%) that were classified as EIN. Of these 46 cases, 11(2.9%) were classified as simple endometrial hyperplasia, 1 (6.3%) as simple atypical endometrial hyperplasia, 6 (12.5%) as complex endometrial hyperplasia, and 28 (90.3%) as complex atypical endometrial hyperplasia. EIN was associated with a higher rate of ${\beta}$-catenin positivity than endometrium classified as benign hyperplasia (72% vs. 22.5%, respectively, P<0.001), but a lower rate than endometrial adenocarcinoma (72% vs. 96.2%, respectively, P<0.001). In benign endometrial hyperplasia, high ${\beta}$-catenin expression was noted in the cell membranes, whereas in EIN and endometrial adenocarcinoma high expression was noted in the cytoplasm. In conclusion, EIN is more accurate than the WHO classification for the diagnosis of precancerous lesions of the endometrium.

Expression and Effects of JMJD2A Histone Demethylase in Endometrial Carcinoma

  • Wang, Hong-Li;Liu, Mei-Mei;Ma, Xin;Fang, Lei;Zhang, Zong-Feng;Song, Tie-Fang;Gao, Jia-Yin;Kuang, Ye;Jiang, Jing;Li, Lin;Wang, Yang-Yang;Li, Pei-Ling
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.7
    • /
    • pp.3051-3056
    • /
    • 2014
  • Previous studies have demonstrated that JMJD2A is a potential oncogene and is overexpressed in human tumors. However, its role in the endometrial carcinoma remains largely unknown. In this study, we discovered that JMJD2A was overexpressed in endometrial carcinoma, using immunohistochemistry, quantitative realtime polymerase chain reaction, and western blotting. Downregulation of JMJD2A led to reduced endometrial carcinoma RL95-2 and ISK cell proliferation, invasion and metastasis as asessed with cell counting kit-8, cell migration and invasive assays. Collectively, our results support that JMJD2A is a promoter of endometrial carcinoma cell proliferation and survival, and is a potential novel drug target.

Knockdown of LKB1 Sensitizes Endometrial Cancer Cells via AMPK Activation

  • Rho, Seung Bae;Byun, Hyun Jung;Kim, Boh-Ram;Lee, Chang Hoon
    • Biomolecules & Therapeutics
    • /
    • v.29 no.6
    • /
    • pp.650-657
    • /
    • 2021
  • Metformin is an anti-diabetic drug and has anticancer effects on various cancers. Several studies have suggested that metformin reduces cell proliferation and stimulates cell-cycle arrest and apoptosis. However, the definitive molecular mechanism of metformin in the pathophysiological signaling in endometrial tumorigenesis and metastasis is not clearly understood. In this study, we examined the effects of metformin on the cell viability and apoptosis of human cervical HeLa and endometrial HEC-1-A and KLE cancer cells. Metformin suppressed cell growth in a dose-dependent manner and dramatically evoked apoptosis in HeLa cervical cancer cells, while apoptotic cell death and growth inhibition were not observed in endometrial (HEC-1-A, KLE) cell lines. Accordingly, the p27 and p21 promoter activities were enhanced while Bcl-2 and IL-6 activities were significantly reduced by metformin treatment. Metformin diminished the phosphorylation of mTOR, p70S6K and 4E-BP1 by accelerating adenosine monophosphate-activated kinase (AMPK) in HeLa cancer cells, but it did not affect other cell lines. To determine why the anti-proliferative effects are observed only in HeLa cells, we examined the expression level of liver kinase B1 (LKB1) since metformin and LKB1 share the same signalling system, and we found that the LKB1 gene is not expressed only in HeLa cancer cells. Consistently, the overexpression of LKB1 in HeLa cancer cells prevented metformin-triggered apoptosis while LKB1 knockdown significantly increased apoptosis in HEC-1-A and KLE cancer cells. Taken together, these findings indicate an underlying biological/physiological molecular function specifically for metformin-triggered apoptosis dependent on the presence of the LKB1 gene in tumorigenesis.

Endometrial profilin 1: A key player in embryo-endometrial crosstalk

  • Lee, Chang-Jin;Hong, Seon-Hwa;Yoon, Min-Ji;Lee, Kyung-Ah;Ko, Jung-Jae;Koo, Hwa Seon;Kim, Jee Hyun;Choi, Dong Hee;Kwon, Hwang;Kang, Youn-Jung
    • Clinical and Experimental Reproductive Medicine
    • /
    • v.47 no.2
    • /
    • pp.114-121
    • /
    • 2020
  • Objective: Despite extensive research on implantation failure, little is known about the molecular mechanisms underlying the crosstalk between the embryo and the maternal endometrium, which is critical for successful pregnancy. Profilin 1 (PFN1), which is expressed both in the embryo and in the endometrial epithelium, acts as a potent regulator of actin polymerization and the cytoskeletal network. In this study, we identified the specific role of endometrial PFN1 during embryo implantation. Methods: Morphological alterations depending on the status of PFN1 expression were assessed in PFN1-depleted or control cells grown on Matrigel-coated cover glass. Day-5 mouse embryos were cocultured with Ishikawa cells. Comparisons of the rates of F-actin formation and embryo attachment were performed by measuring the stability of the attached embryo onto PFN1-depleted or control cells. Results: Depletion of PFN1 in endometrial epithelial cells induced a significant reduction in cell-cell adhesion displaying less formation of colonies and a more circular cell shape. Mouse embryos co-cultured with PFN1-depleted cells failed to form actin cytoskeletal networks, whereas more F-actin formation in the direction of surrounding PFN1-intact endometrial epithelial cells was detected. Furthermore, significantly lower embryo attachment stability was observed in PFN1-depleted cells than in control cells. This may have been due to reduced endometrial receptivity caused by impaired actin cytoskeletal networks associated with PFN1 deficiency. Conclusion: These observations definitively demonstrate an important role of PFN1 in mediating cell-cell adhesion during the initial stage of embryo implantation and suggest a potential therapeutic target or novel biomarker for patients suffering from implantation failure.

Transforming growth factor β1 enhances adhesion of endometrial cells to mesothelium by regulating integrin expression

  • Choi, Hee-Jung;Park, Mi-Ju;Kim, Bo-Sung;Choi, Hee-Jin;Joo, Bosun;Lee, Kyu Sup;Choi, Jung-Hye;Chung, Tae-Wook;Ha, Ki-Tae
    • BMB Reports
    • /
    • v.50 no.8
    • /
    • pp.429-434
    • /
    • 2017
  • Endometriosis is the abnormal growth of endometrial cells outside the uterus, causing pelvic pain and infertility. Furthermore, adhesion of endometrial tissue fragments to pelvic mesothelium is required for the initial step of endometriosis formation outside uterus. $TGF-{\beta}1$ and adhesion molecules importantly function for adhesion of endometrial tissue fragments to mesothelium outside uterus. However, the function of $TGF-{\beta}1$ on the regulation of adhesion molecule expression for adhesion of endometrial tissue fragments to mesothelium has not been fully elucidated. Interestingly, transforming growth factor ${\beta}1$ ($TGF-{\beta}1$) expression was higher in endometriotic epithelial cells than in normal endometrial cells. The adhesion efficiency of endometriotic epithelial cells to mesothelial cells was also higher than that of normal endometrial cells. Moreover, $TGF-{\beta}1$ directly induced the adhesion of endometrial cells to mesothelial cells through the regulation of integrin of ${\alpha}V$, ${\alpha}6$, ${\beta}1$, and ${\beta}4$ via the activation of the $TGF-{\beta}1/TGF-{\beta}RI/Smad2$ signaling pathway. Conversely, the adhesion of $TGF-{\beta}1-stimulated$ endometrial cells to mesothelial cells was clearly reduced following treatment with neutralizing antibodies against specific $TGF-{\beta}1-mediated$ integrins ${\alpha}V$, ${\beta}1$, and ${\beta}4$ on the endometrial cell membrane. Taken together, these results suggest that $TGF-{\beta}1$ may act to promote the initiation of endometriosis by enhancing integrin-mediated cell-cell adhesion.