Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Br J Cancer ; 130(4): 585-596, 2024 03.
Article in English | MEDLINE | ID: mdl-38172534

ABSTRACT

BACKGROUND: The enriched proteins within in vitro fertilisation (IVF)-generated human embryonic microenvironment could reverse progestin resistance in endometrial cancer (EC). METHODS: The expression of thymic stromal lymphopoietin (TSLP) in EC was evaluated by immunoblot and IHC analysis. Transcriptome sequencing screened out the downstream pathway regulated by TSLP. The role of TSLP, androgen receptor (AR) and KANK1 in regulating the sensitivity of EC to progestin was verified through a series of in vitro and in vivo experiments. RESULTS: TSLP facilitates the formation of a BMP4/BMP7 heterodimer, resulting in activation of Smad5, augmenting AR signalling. AR in turn sensitises EC cells to progestin via KANK1. Downregulation of TSLP, loss of AR and KANK1 in EC patients are associated with tumour malignant progress. Moreover, exogenous TSLP could rescue the anti-tumour effect of progestin on mouse in vivo xenograft tumour. CONCLUSIONS: Our findings suggest that TSLP enhances the sensitivity of EC to progestin through the BMP4/Smad5/AR/KANK1 axis, and provide a link between embryo development and cancer progress, paving the way for the establishment of novel strategy overcoming progestin resistance using embryo original factors.


Subject(s)
Endometrial Neoplasms , Thymic Stromal Lymphopoietin , Animals , Female , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Cytokines/metabolism , Cytoskeletal Proteins/metabolism , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Progestins/pharmacology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction , Tumor Microenvironment
2.
J Med Virol ; 96(3): e29468, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38415499

ABSTRACT

Cervical human papillomavirus (HPV) infection is believed to increase the risks of pregnancy failure and abortion, however, whether the uterine cavity HPV infection reduces pregnancy rate or increases miscarriage rate remains unclarified in infertile women undergoing assisted reproductive technology (ART) treatment. Therefore, we aimed to assess ART outcomes in the presence of intrauterine HPV. This was a hospital-based multicenter (five reproductive medicine centers) matched cohort study. This study involved 4153 infertile women undergoing in vitro fertilization (IVF) or intracytoplasmic sperm injection treatment in five reproductive medicine centers between October 2018 and 2020. The spent embryo transfer media sample with endometrium tissue were collected and performed with flow-through hybridization and gene chips to detect HPV DNA. According to basic characteristics, HPV-positive and negative patients were matched in a ratio of 1:4 by age, body mass index transfer timing, transfer type, and number of embryos transferred. The primary outcome was pregnancy and clinical miscarriage rates in the transfer cycle underwent HPV detection. 92 HPV-positive and 368 HPV-negative patients were screened and analyzed statistically. Univariate analysis showed uterine cavity HPV infection resulted in lower rates of ongoing pregnancy (31.5% vs. 44.6%; p = 0.023), implantation (32.3% vs. 43.1%; p = 0.026), biochemical pregnancy (47.8% vs. 62.5%; p = 0.010), and clinical pregnancy (40.2% vs. 54.3%; p = 0.015) compared with HPV negative group. The infertile female with positive HPV also had a slightly higher frequency of biochemical miscarriage (15.9% vs. 13.0%; p = 0.610) and clinical miscarriage (24.3% vs. 15.5%; p = 0.188). These findings suggest that HPV infection in the uterine cavity is a high risk for ART failure. HPV screening is recommended before ART treatment, which may be benefit to improving pregnancy outcome.


Subject(s)
Abortion, Spontaneous , Infertility, Female , Papillomavirus Infections , Pregnancy , Humans , Male , Female , Papillomavirus Infections/diagnosis , Infertility, Female/therapy , Human Papillomavirus Viruses , Cohort Studies , Semen , Embryo Transfer/methods , Reproductive Techniques, Assisted , Fertilization in Vitro , Treatment Failure
3.
Lab Invest ; 102(12): 1335-1345, 2022 12.
Article in English | MEDLINE | ID: mdl-36038734

ABSTRACT

Progestin resistance is the main obstacle for the conservative therapy to maintain fertility in women with endometrial cancer. Brusatol was identified as an inhibitor of the NRF2 pathway; however, its impact on progestin resistance and the underlying mechanism remains unclear. Here, we found that brusatol sensitized endometrial cancer to progestin by suppressing NRF2-TET1-AKR1C1-mediated progestin metabolism. Brusatol transcriptionally suppressed AKR1C1 via modifying the hydroxymethylation status in its promoter region through TET1 inhibition. Suppression of AKR1C1 by brusatol resulted in decreased progesterone catabolism and maintained potent progesterone to inhibit endometrial cancer growth. This inhibition pattern has also been found in the established xenograft mouse and organoid models. Aberrant overexpression of AKR1C1 was found in paired endometrial hyperplasia and cancer samples from the same individuals with progestin resistance, whereas attenuated or loss of AKR1C1 was observed in post-treatment samples with well progestin response as compared with paired pre-treatment tissues. Our findings suggest that AKR1C1 expression pattern may serve as an important biomarker of progestin resistance in endometrial cancer.


Subject(s)
Endometrial Hyperplasia , Endometrial Neoplasms , Humans , Female , Mice , Animals , Endometrial Hyperplasia/drug therapy , Endometrial Hyperplasia/genetics , Progestins/pharmacology , NF-E2-Related Factor 2/metabolism , Progesterone , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins/metabolism , DNA-Binding Proteins
4.
J Assist Reprod Genet ; 37(9): 2053-2079, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32562095

ABSTRACT

PURPOSE: Patients with Klinefelter syndrome (KS) who receive assisted reproductive technology (ART) treatment often experience poor pregnancy rates due to decreased fertilization, cleavage, and implantation rates and even an increased miscarriage rate. Mounting evidence from recent studies has shown that various technological advances and approaches could facilitate the success of ART treatment for KS patients. In this review, we summarize the methods for guiding KS patients during ART and for developing optimal strategies for preserving fertility, improving pregnancy rate and live birth rate, and avoiding the birth of KS infants. METHODS: We searched PubMed and Google Scholar publications related to KS patients on topics of controlled ovarian stimulation protocols, sperm extraction, fertility preservation, gamete artificial activation, round spermatid injection (ROSI), and non-invasive prenatal screening (PGD) methods. RESULTS: This review outlines the different ovulation-inducing treatments for female partners according to the individual sperm status in the KS patient. We further summarize the methods of retrieving sperm, storing, and freezing rare sperm. We reviewed different methods of gamete artificial activation and discussed the feasibility of ROSI for sterile KS patients who absolutely lack sperm. The activation of eggs in the process of intracytoplasmic sperm injection and non-invasive PGD are urgently needed to prevent the birth of KS infants. CONCLUSION: The integrated strategies will pave the way for the establishment of ART treatment approaches and improve the clinical outcome for KS patients.


Subject(s)
Embryo Implantation/genetics , Klinefelter Syndrome/therapy , Reproductive Techniques, Assisted/trends , Birth Rate , Female , Fertility Preservation/trends , Humans , Klinefelter Syndrome/genetics , Klinefelter Syndrome/pathology , Male , Pregnancy , Pregnancy Rate , Sperm Injections, Intracytoplasmic/trends
5.
Biochem Biophys Res Commun ; 482(4): 857-862, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27889612

ABSTRACT

Large amount of clinical evidence has demonstrated that insulin resistance is closely related to oncogenesis of endometrial cancer (EC). Despite recent studies showed the up-regulatory role of insulin in G protein-coupled estrogen receptor (GPER/GPR30) expression, GPER expression was not decreased compared to control when insulin receptor was blocked even in insulin treatment. The purpose of this study was to explore the possible mechanism by which insulin up-regulates GPER that drives EC cell proliferation. For this purpose, we first investigated the GPER expression in tissues of endometrial lesions, further explored the effect of GPER on EC cell proliferation in insulin resistance context. Then we analyzed the role of Ten-Eleven Translocation 1 (TET1) in insulin-induced GEPR expression and EC cell proliferation. The results showed that GPER was highly expressed in endometrial atypical hyperplasia and EC tissues. Mechanistically, insulin up-regulated TET1 expression and the latter played an important role in up-regulating GPER expression and activating PI3K/AKT signaling pathway. TET1 mediated GPER up-regulation was another mechanism that insulin promotes EC cell proliferation.


Subject(s)
Cell Proliferation , Endometrial Neoplasms/pathology , Endometrium/pathology , Insulin/metabolism , Signal Transduction , Cell Line, Tumor , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Female , Humans , Insulin Resistance , Mixed Function Oxygenases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism
6.
Tumour Biol ; 37(9): 12263-12271, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27251670

ABSTRACT

Ovarian cancer is one of the most common cancers which cause female mortality. The knowledge of ovarian cancer initiation and progression is critical to develop new therapeutic strategies to treat and prevent it. Recently, SOX3 has been reported to play a pivotal role in tumor progression. However, the clinical significance of SOX3 in human ovarian cancer remains elusive, and the identity of SOX3 in ovarian cancer initiation, progression, and the related underlying mechanism is unknown. In this study, we showed that SOX3 expression increased from benign and borderline to malignant ovarian tumors. Subsequently, we found that overexpression of SOX3 in EOC cells promoted proliferation, migration, and invasion, while restrained apoptosis and adhesion of ovarian cancer cells. In contrast, silencing of SOX3 gained the opposite results. Finally, we discovered SOX3 targeted Src kinase in EOC cells. These data imply that SOX3, acting as an oncogene in EOC, is not only a crucial factor in the carcinogenesis but also a promising therapeutic target for EOC.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , SOXB1 Transcription Factors/genetics , src-Family Kinases/genetics , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Blotting, Western , Carcinoma, Ovarian Epithelial , Cell Adhesion/genetics , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Humans , Immunohistochemistry , Microscopy, Confocal , Middle Aged , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Oncogenes/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/metabolism , Young Adult , src-Family Kinases/metabolism
7.
Tumour Biol ; 37(7): 9671-80, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26801672

ABSTRACT

NANOG is a key transcription factor that is overexpressed and plays an important role in various cancers. Its overexpression is associated with highly tumorigenic, drug-resistant, and poor prognosis. However, the underlying mechanism of action of NANOG in ovarian cancer remains unclear. Epithelial-mesenchymal transition (EMT), which is a critical process in cancer invasion and metastasis, is also associated with drug resistance. We determined whether NANOG is associated with EMT and chemoresistance in epithelial ovarian cancer cells. NANOG expression was increased in epithelial ovarian cancer cells (HEY and SKOV3) compared with normal epithelial ovarian cells (Moody). Low expression of NANOG increased the expression of E-cadherin and decreased the expression of vimentin, ß-catenin, and Snail. Furthermore, the cell migration and invasion abilities were decreased. The multidrug resistance genes MDR-1 and GST-π were also downregulated when NANOG was lowly expressed. The cells that were transfected with the si-NANOG plasmid were more sensitive to cisplatin compared with the cells that were transfected with empty vector. The data demonstrated that Stat3 was correlated with NANOG-mediated EMT and drug resistance. The silencing of Stat3 expression abrogated NANOG-mediated EMT changes and increased the sensitivity of the cells to chemotherapy. These results suggest that NANOG mediates EMT and drug resistance through activation of the Stat3 pathway in epithelial ovarian cancer.


Subject(s)
Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Nanog Homeobox Protein/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , STAT3 Transcription Factor/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cisplatin/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms, Glandular and Epithelial/etiology , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/etiology , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Snail Family Transcription Factors/genetics , Transfection/methods , Vimentin/genetics , beta Catenin/genetics
8.
Biochem Biophys Res Commun ; 461(3): 525-32, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25911324

ABSTRACT

Our previous study showed that Octamer-binding transcription factor 4 (OCT4) expression was upregulated and significantly associated with histological grade through the analysis of OCT4 expression in 159 ovarian cancer tissue samples, and OCT4 mediated follicle-stimulating hormone (FSH)-induced anti-apoptosis in epithelial ovarian cancer. Nevertheless, whether OCT4 participates in FSH-induced invasion in ovarian cancer is still unknown. Therefore, the present study aimed to define whether FSH-induced ovarian cancer invasion is mediated by OCT4. In present study, we showed that FSH induced not only the epithelial-mesenchymal transition (EMT) and invasive phenotype but also the upregulation of OCT4 expression in a dose- and time-dependent manner in epithelial ovarian cancer cells. In addition, the expression of FSH receptor (FSHR) was upregulated by FSH induction, and knockdown of FSHR inhibited FSH-stimulated OCT4 expression. ERK1/2 signaling pathway participated in the enhanced expression of OCT4 and Snail induced by FSH. We further showed that the activated expression of Snail and N-cadherin, the suppressed expression of E-cadherin and the morphological change of the cells stimulated by FSH were blocked by OCT4-specific small interfering RNA. Moreover, our results showed that OCT4 mediated the increase in invasive capacity induced by FSH in ovarian cancer cells. Taken together, our work reveals that OCT4 is an essential mediator in FSH-induced EMT and invasion in epithelial ovarian cancer and may act as a potential therapeutic target.


Subject(s)
Follicle Stimulating Hormone/physiology , MAP Kinase Signaling System , Neoplasm Invasiveness , Neoplasms, Glandular and Epithelial/pathology , Octamer Transcription Factor-3/physiology , Ovarian Neoplasms/pathology , Base Sequence , Carcinoma, Ovarian Epithelial , Humans , Neoplasms, Glandular and Epithelial/enzymology , Neoplasms, Glandular and Epithelial/metabolism , Octamer Transcription Factor-3/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/metabolism , Polymerase Chain Reaction , RNA, Small Interfering , Receptors, FSH/physiology
9.
Tumour Biol ; 36(3): 1529-37, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25608836

ABSTRACT

The purpose of the study was to explore the role and mechanism of ataxia-telangiectasia mutated (ATM) protein in endometrial carcinogenesis. A reverse-phase protein array (RPPA) was used to analyze the expression of ATM signal pathway proteins in Ishikawa and progesterone-insensitive Ishikawa. ATM expression was detected in endometrium specimens by immunohistochemistry, including 8 cases with proliferative endometrium, 6 cases with secretory endometrium, 10 cases with simple hyperplasia (SH), 13 cases of complex hyperplasia (CH), 11 cases of endometrial atypical hyperplasia (EAH), and 83 cases with type I endometrial cancer. The relationship between ATM expression and other clinicopathological indicators was also examined in type I endometrial cancer patients. The mechanisms of ATM were explored in vitro with the endometrial cell lines Ishikawa and RL95-2. A cell counting kit-8 (CCK-8) test and Western blot analysis were performed to test proliferation and protein expression. Statistical analysis was performed with SPSS19.0. The significance level was set at 0.05. ATM was increased with medroxyprogesterone acetate (MPA) stimulation in Ishikawa in RPPA. ATM expression gradually decreased in endometrial hyperplasic lesions compared with the normal proliferative and secretory endometrium and was the lowest in type I endometrial cancer. ATM expression was negatively correlated with pathological grades in type I endometrial cancer. In vitro, ATM silencing retarded proliferation inhibition in Ishikawa and RL95-2 treated with MPA. ATM silencing could down-regulate the MPA-stimulated signal proteins, including Chk2, P53, and caspase-3 in vitro. MPA might exert its role through activating the ATM-associated pathway, ATM-Chk2-P53-caspase-3 (active), preserving normal endometrium and protecting it from malignancies. ATM might be a promising indicator for endometrial hyperplasia and cancer.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinogenesis/metabolism , Endometrial Neoplasms/metabolism , Progesterone/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Proliferation/genetics , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/metabolism , Endometrium/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Progesterone/metabolism , Protective Factors , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
Int J Gynecol Cancer ; 24(9): 1564-74, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25340291

ABSTRACT

PURPOSE: It has previously been shown that follicle-stimulating hormone (FSH) and its receptor contribute to epithelial ovarian cancer (EOC) development. Epithelial-mesenchymal transition (EMT) is the early event of metastasis in cancer. Therefore, the aim of this study was to investigate the roles of FSH and the FSH receptor (FSHR) in EMT of EOC. METHODS: Ovarian cancer cells treated with various doses of FSH were used to investigate the effect of FSH on EMT. Small interfering RNA-mediated FSHR depletion or reexpression of FSHR by acute transfecting pcDNA-hFSHR plasmid was performed to determine the role of FSHR in FSH-induced EMT. Moreover, LY294002, a potent and specific cell-permeable inhibitor of phosphatidylinositol 3-kinases (PI3K), was selected to pretreat ovarian cancer cells to confirm whether PI3K/Akt signaling is involved in this event. RESULTS: In the current study, FSH was found to induce the phenotypes of EMT including migration and invasion in EOC cells. Elevated FSHR levels promoted EMT, migration, and invasion, whereas small interfering RNA-mediated FSHR knockdown inhibited these processes. Moreover, the inhibition of FSH-induced PI3K/Akt signaling pathway attenuated Snail expression and the EMT process. CONCLUSIONS: Collectively, the findings of the current study indicate that FSH induced the EMT of ovarian cancer cells through the FSHR-PI3K/Akt-Snail signaling pathway.


Subject(s)
Cystadenocarcinoma, Papillary/pathology , Cystadenocarcinoma, Serous/pathology , Epithelial-Mesenchymal Transition/drug effects , Follicle Stimulating Hormone/pharmacology , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, FSH/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cystadenocarcinoma, Papillary/drug therapy , Cystadenocarcinoma, Papillary/metabolism , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Female , Fluorescent Antibody Technique , Hormones/pharmacology , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, FSH/antagonists & inhibitors , Receptors, FSH/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
11.
Int J Gynecol Cancer ; 23(2): 318-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23318911

ABSTRACT

OBJECTIVES: The objective of this study was to analyze the relationship among the protein levels of MCM7, p63, and human papillomavirus (HPV) in different cervical lesion tissues and appraise their predictive value in evaluating severity of cervical disease. METHODS: Twelve normal cervix or chronic cervicitis, 42 squamous intraepithelial lesions, and 53 cervical carcinoma tissues were enrolled, and the protein levels of MCM7, p63, and HPV were detected by immunohistochemistry. RESULTS: The positive examination rates of all the MCM7, p63, and HPV proteins increased gradually and significantly from normal cervix and chronic cervicitis tissues, low-grade squamous intraepithelial lesions, high-grade squamous intraepithelial lesions to cervical carcinomas, respectively. As to predict high-grade squamous intraepithelial lesions and carcinogenesis is concerned, the MCM7 protein had a sensitivity of 94.0%, a specificity of 56.5%, a positive predictive value of 88.8%, and a negative predictive value of 72.2%. The p63 protein had a sensitivity of 78.6%, a specificity of 81.8%, a positive predictive value of 94.3%, and a negative predictive value of 50.0%. Protein level of MCM7 was positively correlated with that of p63 in cervical tissues (r = 0.806, P < 0.01), and the p63 was also positively correlated with histopathologic type (P < 0.05). CONCLUSIONS: Protein levels of MCM7 and p63 were associated significantly with high-grade cervical lesion, and aberrant p63 protein level may distinguish different histopathologic types of cervical carcinoma. They may act as co-predictive index in both HPV-dependent and HPV-independent high-grade cervical lesion with high sensitivity and specificity.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Neoplasms/diagnosis , Adult , Aged , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cervix Uteri/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Minichromosome Maintenance Complex Component 7 , Predictive Value of Tests , Prognosis , Severity of Illness Index , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Dysplasia/pathology
12.
Mol Med ; 18: 1197-208, 2012 Oct 24.
Article in English | MEDLINE | ID: mdl-22801793

ABSTRACT

Ovarian cancers are heterogeneous and contain stemlike cells that are able to self-renew and are responsible for sustained tumor growth. Metastasis in the peritoneal cavity occurs more frequently in ovarian cancer than in other malignancies, but the underlying mechanism remains largely unknown. We have identified that ovarian cancer stemlike cells (CSCs), which were defined as side population (SP) cells, were present in patients' ascitic fluid and mesenchymally transformed cell lines, ES-2 and HO-8910PM. SP cells, which were sorted from both cell lines and implanted into immunocompromised mice, were localized to the xenografted tumor boundary. In addition, SP cells exhibited an epithelial phenotype and showed a distinct gene expression profile with reduced expression of cell adhesion molecules (CAMs), indicating that SP cells exert an important role in ovarian cancer progression on the basis of their delicate interaction with the surrounding microenvironment and anatomical localization in tumors. In contrast, non-SP cells exhibited a more mesenchymal phenotype and showed more increased invasive potential than SP cells. This heterogeneity was observed as an endogenous transformation via the epithelial-mesenchymal transition (EMT) process. Inhibition of the EMT process by Snail1 silencing reduced the SP cell frequency, and affected their invasive capacity and engraftment. These findings illustrate the interplay between epithelial ovarian CSCs and the EMT, and exert a link to explain tumor heterogeneity and its necessity for ovarian cancer maintenance, metastasis and progression.


Subject(s)
Cell Lineage , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition , Mesoderm/pathology , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/pathology , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Animals , Ascitic Fluid/drug effects , Ascitic Fluid/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Injections, Subcutaneous , Mesoderm/drug effects , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasms, Glandular and Epithelial/genetics , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/genetics , Side-Population Cells/drug effects , Side-Population Cells/pathology , Snail Family Transcription Factors , Transcription Factors/metabolism , Transforming Growth Factor beta1/pharmacology , Xenograft Model Antitumor Assays
13.
J Obstet Gynaecol Res ; 38(8): 1077-85, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22540333

ABSTRACT

AIM: Metformin plays an important role in the inhibition of cancer cell growth and prolongs remission durations. It reverses progestin-resistance in endometrial cancer cells by downregulating glyoxalase I (GloI) expression. This study aimed to investigate the effect of metformin on endometrial cancer cell chemotherapeutic sensitivity and explore the underlying molecular mechanisms. MATERIAL AND METHODS: MTT assay was performed to determine the rate of cell death after cisplatin and paclitaxel with or without metformin. Western blot was carried out to analyze GloI expression. SiRNA-targeting of GloI was used to knockdown GloI expression before further treatment with chemotherapeutic agents to examine the effect of GloI downregulation on chemotherapy-induced cell killing. In addition, plasmid transfection was used to overexpress GloI and determine whether high GloI levels blocked metformin-enhanced cell sensitivity to chemotherapy. PCR was used to analyze the efficiency of RNA interference and plasmid transfection. RESULTS: The addition of metformin enhanced the sensitivity of endometrial cells to cisplatin and paclitaxel, which was associated with reduced levels of GloI expression. Moreover, low-dose chemotherapeutic drugs alone could not significantly reduce GloI expression, whereas the addition of metformin potently downregulated GloI protein levels. Cisplatin and paclitaxel markedly inhibited the proliferative ability of GloI-depleted endometrial cancer cells. However, the overexpression of GloI abolished the effect of metformin-enhanced cell sensitivity to chemotherapeutic drugs. CONCLUSION: Metformin enhances the rate of cell-killing induced by chemotherapeutic agents by repressing GloI expression.


Subject(s)
Carcinoma, Endometrioid/drug therapy , Endometrial Neoplasms/drug therapy , Hypoglycemic Agents/therapeutic use , Lactoylglutathione Lyase/metabolism , Metformin/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Endometrioid/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/therapeutic use , Drug Evaluation, Preclinical , Drug Synergism , Endometrial Neoplasms/enzymology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Hypoglycemic Agents/pharmacology , Lactoylglutathione Lyase/genetics , Metformin/pharmacology , Paclitaxel/therapeutic use
14.
Arch Gynecol Obstet ; 286(3): 729-37, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22569715

ABSTRACT

OBJECTIVE: To determine the role of oestrogen receptor α (ERα) in the regulation of survivin expression by 17ß-estradiol (E(2)) in ovarian cancer cells and to evaluate the mechanism of E(2) action on ovarian cancer cell migration. METHODS: We performed RT-PCR and Western blot analysis to assess the expression of ERα in the ovarian cancer cell lines NIH:OVCAR-3 and SKOV-3. Full-length ERα cDNA was reintroduced into SKOV-3 cells through stable transfection. After treatment with E(2), with or without pre-incubation of anti-oestrogen compound ICI 182780, RT-PCR and Western blot analysis were performed to detect survivin expression at the mRNA and protein levels. RNA interference (RNAi) was used to inhibit the expression of survivin in SKOV-3 cells. Wound healing-induced migration and Matrigel invasion experiments were performed to determine the motility of ovarian cancer cells. RT-PCR and gelatin zymography were used to detect the expression and activity of MMP-9 in SKOV-3 cells. RESULTS: A stably transfected clone with over-expression of ERα, SKOV-α, was isolated. Exogenous or endogenous expression of ERα in SKOV-3 or NIH:OVCAR-3 cells resulted in a significant up-regulation of survivin in the presence of E(2). Pre-treatment with ICI 182780 attenuated the up-regulation of survivin by E(2). Previous data from our laboratory showed that E(2) enhanced the motility of ovarian cancer cells. RNAi strongly inhibited survivin expression in SKOV-3 cells. Knock-down of survivin expression reduced the migration and invasion of SKOV-3 cells, which correlated with down-regulation of MMP9 mRNA expression and activity. CONCLUSIONS: ERα may be responsible for the up-regulation of survivin after E(2) treatment in ovarian cancer cells. The mechanism of oestrogen-promoted ovarian cancer metastasis may due to the up-regulation of survivin conducted through the ERα signalling pathway.


Subject(s)
Cell Movement , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Ovarian Neoplasms/metabolism , Animals , Cell Line, Tumor , DNA, Complementary , Female , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Survivin , Transfection , Up-Regulation
15.
Cancers (Basel) ; 14(24)2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36551694

ABSTRACT

With a younger tendency in morbidity age, endometrial cancer (EC) incidence has grown year after year. Worse, even more commonly occurring is endometrial hyperplasia (EH), which is a precancerous endometrial proliferation. For young women with early EC and EH who want to preserve fertility, progestin therapy has been utilized as a routine fertility-preserving treatment approach. Nevertheless, progestin medication failure in some patients is mostly due to progestin resistance and side effects. In order to further analyze the potential mechanisms of progestin resistance in EH and EC, to provide theoretical support for effective therapeutic strategies, and to lay the groundwork for searching novel treatment approaches, this article reviews the current therapeutic effects of progestin in EH and EC, as well as the mechanisms and molecular biomarkers of progestin resistance, and systematically expounds on the potential therapeutic methods to overcome progestin resistance.

16.
Front Endocrinol (Lausanne) ; 13: 1037220, 2022.
Article in English | MEDLINE | ID: mdl-36277710

ABSTRACT

Objective: To examine the efficacy of gonadotropin releasing hormone (GnRH) antagonist (GnRH-ant) protocol and the long GnRH agonist (GnRH-a) protocol during in vitro fertilization (IVF) therapy in patients with severe male infertile factors. Methods: A total of 983 women with severe male factor infertility undergoing IVF therapy from 2017 to 2020 at one center were retrospectively analyzed. Patients were divided into the GnRH-ant group (n=527) and the GnRH-a group (n=456) according to their ovarian stimulation protocols. Patient baseline characteristics, ovarian stimulation characteristics, and clinical pregnancy outcomes were compared between the groups. The live birth rate was considered the main pregnancy outcome. Results: GnRH-a group had a higher live birth rate compared with the GnRH-ant group (41.0% versus 31.3%, p=0.002). Moreover, the implantation (32.8% vs. 28.1%, p=0.033), biochemical pregnancy (52.4% versus 44.8%, p=0.017), clinical pregnancy (49.3% versus 39.7%, p=0.002) and ongoing pregnancy rates (43.2% vs. 34.9%, p=0.008) were higher in GnRH-a group. For patients with one embryo transferred, the GnRH-a group demonstrated higher live birth (37.0% vs. 19.4%, p=0.010) and ongoing pregnancy rate (38.9% vs. 24.5%, p=0.046) than the GnRH-ant group. Among patients with two embryos transferred, the live birth rate was also higher in the GnRH-a group than in the GnRH-ant group, with no statistical difference. No significant differences were observed in the biochemical abortion rate, clinical miscarriage rate, early miscarriage rate, late miscarriage rate, heterotopic pregnancy rate, twin pregnancy rate, and birth sex ratio between the two groups. Conclusion: For individuals with severe male infertility undergoing IVF, the GnRH-a protocol is considered a more efficient and feasible strategy with a higher live birth rate compared to the GnRH-ant protocol, especially in single embryo transfer.


Subject(s)
Abortion, Spontaneous , Infertility, Male , Humans , Male , Female , Pregnancy , Retrospective Studies , Ovulation Induction/methods , Hormone Antagonists/therapeutic use , Fertilization in Vitro/methods , Gonadotropin-Releasing Hormone , Infertility, Male/drug therapy
17.
Cancer Sci ; 102(3): 557-64, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21205080

ABSTRACT

Progestin resistance is the main obstacle to successful conservative therapy in young endometrial cancer patients. To investigate the molecular events that lead to progestin resistance and to find a possible way to reverse progestin resistance in endometrial cancer, we established a progestin-resistant Ishikawa cell line by long-term progestin treatment to downregulate progesterone receptor (PR) expression. Both medoxyprogesterone acetate (MPA) and LY294002, a phosphatidylinositol 3-kinase (PI3K) inhibitor, were assayed for their effects on the proliferation of progestin-sensitive and progestin-resistant cancer cells, respectively. The MPA inhibited the PI3K/Akt pathway and suppressed cell proliferation in progestin-sensitive Ishikawa cells, but activated the PI3K/Akt pathway and had no effect on cell proliferation in progestin-resistant Ishikawa cells or HEC-1A cells. Inhibiting the PI3K/Akt pathway by LY294002 upregulated PR expression and diminished cell growth, especially in progestin-resistant endometrial cancer cells. In vivo endometrial cancer xenograft studies in nude mice also showed that inhibiting the PI3K/Akt pathway reversed progestin resistance in endometrial cancer. Our results indicate that activation of the PI3K/Akt pathway by progestin without PR mediation plays an important role in progestin resistance to endometrial cancer cells. In addition, inhibiting the PI3K/Akt pathway might reverse progestin resistance in endometrial cancer.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Chromones/pharmacology , Endometrial Neoplasms/drug therapy , Medroxyprogesterone Acetate/pharmacology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Endometrial Neoplasms/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Receptors, Progesterone/analysis , Receptors, Progesterone/physiology
18.
BMC Cancer ; 11: 22, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21244707

ABSTRACT

BACKGROUND: ARHI is a Ras-related imprinted gene that inhibits cancer cell growth and motility. ARHI is downregulated in the majority of breast cancers, and loss of its expression is associated with its progression from ductal carcinoma in situ (DCIS) to invasive disease. In ovarian cancer, re-expression of ARHI induces autophagy and leads to autophagic death in cell culture; however, ARHI re-expression enables ovarian cancer cells to remain dormant when they are grown in mice as xenografts. The purpose of this study is to examine whether ARHI induces autophagy in breast cancer cells and to evaluate the effects of ARHI gene re-expression in combination with paclitaxel. METHODS: Re-expression of ARHI was achieved by transfection, by treatment with trichostatin A (TSA) or by a combination of TSA and 5-aza-2'-deoxycytidine (DAC) in breast cancer cell cultures and by liposomal delivery of ARHI in breast tumor xenografts. RESULTS: ARHI re-expression induces autophagy in breast cancer cells, and ARHI is essential for the induction of autophagy. When ARHI was re-expressed in breast cancer cells treated with paclitaxel, the growth inhibitory effect of paclitaxel was enhanced in both the cell culture and the xenografts. Although paclitaxel alone did not induce autophagy in breast cancer cells, it enhanced ARHI-induced autophagy. Conversely, ARHI re-expression promoted paclitaxel-induced apoptosis and G2/M cell cycle arrest. CONCLUSIONS: ARHI re-expression induces autophagic cell death in breast cancer cells and enhances the inhibitory effects of paclitaxel by promoting autophagy, apoptosis, and G2/M cell cycle arrest.


Subject(s)
Autophagy/genetics , Gene Expression Regulation, Neoplastic/genetics , Paclitaxel/pharmacology , rho GTP-Binding Proteins/genetics , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Decitabine , Gene Expression Regulation, Neoplastic/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hydroxamic Acids/pharmacology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Microscopy, Fluorescence , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , rho GTP-Binding Proteins/metabolism
19.
Int J Gynecol Cancer ; 21(2): 213-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21270604

ABSTRACT

INTRODUCTION: A long-term treatment with progestin commonly results in progestin resistance in endometrial cancer. So, the aim of this study was to investigate the role of glyoxalase I (GloI), a mediator of chemotherapy resistance, in metformin reversal of progestin resistance in endometrial carcinoma. METHODS: The proliferation variety of endometrial cancer cells was determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-Diphenyltetrazolium (MTT) assay after exposure to medroxyprogesterone acetate, metformin, or both reagents; apoptosis rates were assessed by flow cytometry. Real-time polymerase chain reaction was used to evaluate the effect of small interfering RNA sequence on target gene expression. Western immunoblotting was performed to determine the expression of GloI and the molecules of the mammalian target of rapamycin (mTOR) pathway. RESULT: Knocking down GloI sensitized progestin-resistant Ishikawa cells to progestin. Metformin downregulated GloI expression, reversed progestin resistance, enhanced progestin-induced cell proliferation inhibition, and induced apoptosis in progestin-resistant Ishikawa cells. In addition, medroxyprogesterone acetate-induced mTOR phosphorylation was blocked by metformin. Metformin abolishes mTOR phosphorylation and inhibits GloI expression, attenuating proliferation and inducing apoptosis in progestin-resistant Ishikawa cells. CONCLUSIONS: Dysregulation of GloI expression in endometrial cancer may be part of the molecular mechanisms for progestin resistance.


Subject(s)
Carcinoma, Endometrioid/drug therapy , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Lactoylglutathione Lyase/metabolism , Metformin/pharmacology , Progestins/metabolism , Apoptosis , Carcinoma, Endometrioid/metabolism , Down-Regulation , Endometrial Neoplasms/metabolism , Female , Humans , Tumor Cells, Cultured
20.
Int J Cancer ; 126(9): 2102-11, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19795442

ABSTRACT

Snail, a key inducer of epithelial-mesenchymal transition (EMT), plays an important role in cancer metastasis. To better understand the role of Snail in the metastasis of ovarian carcinoma, expression of Snail was knocked down by antisense-Snail in the highly metastatic ovarian cancer cell line HO8910PM. Gene array analysis revealed that blocking Snail expression suppressed the activity of matrix metalloproteinases (MMPs) and upregulated TIMP3, an MMP inhibitor. These findings suggest that Snail interacts with MMP during tumor invasion and metastasis. In addition, we examined the role of Snail in an ovarian cancer orthotopic model by using the antisense-Snail HO8910PM cell line. We found that the size of primary ovarian cancer tumor and the number of metastatic lesions were significantly reduced when Snail was knocked down. Confirming our initial findings, the activity of MMP2 was greatly inhibited in tumors from antisense-Snail cells. Furthermore, immunohistochemical analysis on ovarian cancer progression tissue array demonstrated that the expression of Snail was significantly higher in metastatic lesions, and Snail expression correlated with the stage of ovarian cancer. Interestingly, in early-stage tumors, Snail was localized in both the cytoplasm and nucleus. In late stage and metastatic lesions, the level of Snail was elevated, and Snail was localized to the nucleus. The expression level and nuclear localization of Snail were also inversely correlated with E-cadherin expression. Overall, our study indicates that Snail plays a critical role in tumor growth and metastasis of ovarian carcinoma through regulation of MMP activity.


Subject(s)
Ovarian Neoplasms/pathology , Transcription Factors/physiology , Adult , Aged , Animals , Cadherins/analysis , Cell Line, Tumor , Cell Proliferation , Epithelium/pathology , Female , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mesoderm/pathology , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Snail Family Transcription Factors , Tissue Inhibitor of Metalloproteinase-3/physiology , Transcription Factors/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL