Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(7)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38612536

RESUMO

The endometrial epithelium and underlying stroma undergo profound changes to support and limit embryo adhesion and invasion, which occur in the secretory phase of the menstrual cycle during the window of implantation. This coincides with a peak in progesterone and estradiol production. We hypothesized that the interplay between hormone-induced changes in the mechanical properties of the endometrial epithelium and stroma supports this process. To study it, we used hormone-responsive endometrial adenocarcinoma-derived Ishikawa cells growing on substrates of different stiffness. We showed that Ishikawa monolayers on soft substrates are more tightly clustered and uniform than on stiff substrates. Probing for mechanical alterations, we found accelerated stress-relaxation after apical nanoindentation in hormone-stimulated monolayers on stiff substrates. Traction force microscopy furthermore revealed an increased number of foci with high traction in the presence of estradiol and progesterone on soft substrates. The detection of single cells and small cell clusters positive for the intermediate filament protein vimentin and the progesterone receptor further underscored monolayer heterogeneity. Finally, adhesion assays with trophoblast-derived AC-1M-88 spheroids were used to examine the effects of substrate stiffness and steroid hormones on endometrial receptivity. We conclude that the extracellular matrix and hormones act together to determine mechanical properties and, ultimately, embryo implantation.


Assuntos
Matriz Extracelular , Progesterona , Feminino , Humanos , Epitélio , Ciclo Menstrual , Estradiol
2.
BMC Cancer ; 23(1): 995, 2023 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-37853361

RESUMO

BACKGROUND: Endometrial carcinoma (EC) is a malignant tumor of the female reproductive tract that has been associated with increased morbidity and mortality. This study aimed to identify biomarkers and potential therapeutic targets for EC. METHODS: A publicly available transcriptome data set comprising 587 EC cases was subjected to a comprehensive bioinformatics analysis to identify candidate genes responsible for EC occurrence and development. Next, we used clinical samples and cell experiments for validation. RESULTS: A total of 1,617 differentially expressed genes (DEGs) were identified. Analysis of patient survival outcomes revealed that BUB1, BUB1B, CCNA2, and CDCA8 were correlated with prognosis in patients with EC. Moreover, assessment of clinical samples confirmed that BUB1, BUB1B, CCNA2 and CDCA8 were strongly expressed in EC tissues. Additionally, bioinformatics and luciferase reporter assays confirmed that miR-524-5p can target and regulate these four genes. Overexpression of miR-524-5p significantly inhibited EC Ishikawa cells viability, migration and invasion. Inhibition of miR-524-5p showed the opposite results. CONCLUSIONS: Expression of miR-524-5p reduced the migration and invasion of Ishikawa EC cells, and decreased BUB1, BUB1B, CCNA2, and CDCA8 expression. miR-524-5p, as well as BUB1, BUB1B, CCNA2, and CDCA8, may be clinically relevant biomarkers for the diagnosis and treatment of EC.


Assuntos
Neoplasias do Endométrio , MicroRNAs , Humanos , Feminino , MicroRNAs/genética , MicroRNAs/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Biomarcadores , Neoplasias do Endométrio/diagnóstico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ciclina A2/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
3.
Gynecol Obstet Invest ; 87(2): 89-99, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35130539

RESUMO

OBJECTIVES: Melatonin (MLT) shows antitumor effects in various tumor types, including endometrial carcinoma. However, the molecular mechanism involved is unclear. In the current study, we investigated the effect of MLT on the estrogen-induced epithelial-mesenchymal transition (EMT) in endometrial adenocarcinoma cells and explored the pathway that might be involved. DESIGN: Laboratory study was via cultured endometrial cancer cells. Design refers only to in vitro experiments. METHODS: In cell culture experiments, cell growth was examined using CCK-8 assays. The expression of Numb and EMT markers in Ishikawa cells was examined using Western blot analysis and real-time PCR. Cell invasion was examined using transwell assays. Cell migration was examined using wound-healing assays and transwell assays. Using immunohistochemistry analysis, the expression of Numb in human endometrial cancers was examined. RESULTS: In immunohistochemistry experiments, we found that 15.2% of atypical endometrial hyperplasia and 15.6% of endometrial carcinoma did not express Numb. In cell culture experiments, MLT inhibited cell proliferation, invasion, and migration induced by 17ß-estradiol (E2) in endometrial cancer cells. MLT decreased the expression of vimentin and Slug and increased the expression of Numb and E-cadherin in Ishikawa cells. Numb knockdown in cancer cells significantly increased cell proliferation, invasion, and migration. LIMITATIONS: No animal experiments were performed. CONCLUSIONS: MLT blocked E2-induced cell growth and EMT in endometrial cancer cells via upregulating Numb expression.


Assuntos
Adenocarcinoma , Neoplasias do Endométrio , Melatonina , Adenocarcinoma/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Transição Epitelial-Mesenquimal , Estradiol/farmacologia , Feminino , Humanos , Melatonina/farmacologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/farmacologia
4.
Lasers Med Sci ; 37(8): 3193-3201, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35727394

RESUMO

This study was conducted to investigate the inhibitory effects of light-emitting diodes (LEDs) on exosome biogenesis and angiogenesis capacity in Ishikawa endometrial cancer cells. To this end, cells were exposed to different energy densities (fluences) of 4, 8, 16, 32, and 64 J/cm2 for 5 days (once every 24 h), and cell viability was determined using an MTT assay. Based on data from the MTT panel, cells were exposed to 4 and 16 J/cm2 for subsequent analyses. Exosome biogenesis was also monitored via monitoring the expression of CD63, ALIX, and Rab27a and b. The size and morphology of exosomes in the supernatant were measured using scanning electron microscopy (SEM), and dynamic light scattering (DLS). Using Transwell insert, the migration capacity of these cells was studied. The angiogenic effects of irradiated Ishikawa cell secretome at different fluences were monitored on human endothelial cells using in vitro tubulogenesis. Results indicated LED can reduce the viability of Ishikawa cells in a dose-dependent manner. According to our data, 4 and 64 J/cm2 groups exhibited minimum and maximum cytotoxic effects compared to the control cells. Data revealed a close proportional relationship between the power of laser and exosome average size compared to the non-treated control cells (p < 0.05). Real-time PCR analysis showed the suppression of Rab27b and up-regulation of Rab27a in irradiated cells exposed to 4 and 16 J/cm2 (p < 0.05). These effects were evident in the 16 J/cm2 group. Likewise, LED can inhibit the migration of Ishikawa cells in a dose-dependent manner (p < 0.05). Tubulogenesis activity of endothelial cells was suppressed after incubation with the secretome of irradiated Ishikawa cells (p < 0.05). These data showed tumoricidal properties of LED irradiation on human adenocarcinoma Ishikawa cells via the inhibition of exosome biogenesis and suppression of angiogenesis capacity.


Assuntos
Adenocarcinoma , Exossomos , Sobrevivência Celular/efeitos da radiação , Células Endoteliais , Exossomos/metabolismo , Feminino , Humanos , Regulação para Cima
5.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-34830456

RESUMO

Selective estrogen receptor modulators (SERMs) act as estrogen receptor (ERα) agonists or antagonists depending on the target issue. Tamoxifen (TAM) (a non-steroidal triphenylethylene derivative) was the first SERM approved as anti-estrogen for the treatment of metastatic breast cancer. On the hunt for novel SERMs with potential growth inhibitory activity on breast cancer cell lines yet no potential to induce endometrial carcinoma, we designed and synthesized 28 novel TAM analogs. The novel analogs bear a triphenylethylene scaffold. Modifications on rings A, B, and C aim to attenuate estrogenic/anti-estrogenic activities of the novel compounds so they can potentially inhibit breast cancer and provide positive, beneficial estrogenic effects on other tissues with no risk of developing endometrial hyperplasia. Compound 12 (E/Z-1-(2-{4-[1-(4-Chloro-phenyl)-2-(4-methoxy-phenyl)-propenyl]-phenoxy}-ethyl)-piperidine) showed an appreciable relative ERα agonistic activity in a yeast estrogen screen (YES) assay. It successfully inhibited the growth of the MCF-7 cell line with GI50 = 0.6 µM, and it was approximately three times more potent than TAM. It showed no potential estrogenicity on Ishikawa endometrial adenocarcinoma cell line via assaying alkaline phosphatase (AlkP) activity. Compound 12 was tested in vivo to assess its estrogenic properties in an uterotrophic assay in an ovariectomized rat model. Compared to TAM, it induced less increase in wet uterine wet weight and showed no uterotrophic effect. Compound 12 is a promising candidate for further development due to its inhibition activity on MCF-7 proliferation with moderate AlkP activity and no potential uterotrophic effects. The in vitro estrogenic activity encourages further investigations toward potential beneficial properties in cardiovascular, bone, and brain tissues.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias do Endométrio/tratamento farmacológico , Receptor alfa de Estrogênio/genética , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Antagonistas de Estrogênios/síntese química , Antagonistas de Estrogênios/farmacologia , Feminino , Humanos , Células MCF-7 , Ratos , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/genética , Moduladores Seletivos de Receptor Estrogênico/síntese química , Estilbenos/síntese química , Estilbenos/farmacologia , Tamoxifeno/análogos & derivados
6.
Int J Mol Sci ; 20(16)2019 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-31394756

RESUMO

The metabolite 2-methoxyestradiol (2ME) is an endogenous estrogen metabolite with potential therapeutic properties in reproductive cancers. However, the molecular mechanisms by which 2ME exerts its anticancer activity are not well elucidated. The purpose of this study was to determine the molecular signals associated with the apoptotic effects of 2ME in a human endometrial cancer cell line. Ishikawa cells were treated with non-apoptotic (0.1 µM) or apoptotic concentrations (5 µM) of 2ME, and 12 hours later mRNA levels for Scd2, Snx6, and Spon1 were determined by real-time PCR. We then investigated by immunofluorescence and Western blot the expression and distribution of F-spondin, encoded by Spon1, in Ishikawa cells treated with 2ME 5 µM at 6, 12, or 24 h after treatment. The role of estrogen receptors (ER) in the effect of 2ME on the Spon1 level was also investigated. Finally, we examined whether 2ME 5 µM induces cell death in Ishikawa cells pre-incubated with a neutralizing F-spondin antibody. Non-apoptotic or apoptotic concentrations of 2ME decreased Scd2 and increased Snx6. However, Spon1 was only increased with the 2ME apoptotic concentration. F-spondin protein was also increased at 12 and 24 h after 2ME treatment, while 2ME-induced Spon1 increase was independent of ER. Neutralization of F-spondin blocked the effect of 2ME on the cell viability. These results show that F-spondin signaling is one of the components in the apoptotic effects of 2ME on Ishikawa cells and provide experimental evidence underlying the mechanism of action of this estrogen metabolite on cancer cells.


Assuntos
2-Metoxiestradiol/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Endométrio/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Biomarcadores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Humanos , Espaço Intracelular/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Estrogênio/metabolismo
7.
Apoptosis ; 22(4): 570-584, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28176146

RESUMO

Centchroman (CC) or Ormeloxifene has been shown to induce apoptosis and cell cycle arrest in various types of cancer cells. This has, however, not been addressed for endometrial cancer cells where its (CC) mechanism of action remains unclear. This study focuses on the basis of antineoplasticity of CC by blocking the targets involved in the cell cycle, survival and apoptosis in endometrial cancer cells. Ishikawa Human Endometrial Cancer Cells were cultured under estrogen deprived medium, exposed to CC and analyzed for proliferation and apoptosis. Additionally, we also analyzed oxidative stress induced by CC. Cell viability studies confirmed the IC50 of CC in Ishikawa cells to be 20 µM after 48 h treatment. CC arrests the cells in G0/G1 phase through cyclin D1 and cyclin E mediated pathways. Phosphatidylserine externalization, nuclear morphology changes, DNA fragmentation, PARP cleavage, and alteration of Bcl-2 family protein expression clearly suggest ongoing apoptosis in the CC treated cells. Activation of caspase 3 & 9, up-regulation of AIF and inhibition of apoptosis by z-VAD-fmk clearly explains the participation of the intrinsic pathway of programmed cell death. Further, the increase of ROS, loss of MMP, inhibition of antioxidant (MnSOD, Cu/Zn-SOD and GST) and inhibition of apoptosis with L-NAC suggests CC induced oxidative stress leading to apoptosis via mitochondria mediated pathway. Therefore, CC could be a potential therapeutic agent for the treatment of Endometrial Cancer adjunct to its utility as a contraceptive and an anti-breast cancer agent.


Assuntos
Adenocarcinoma/patologia , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Centocromano/farmacologia , Neoplasias do Endométrio/patologia , Caspases/fisiologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Ciclina D1/fisiologia , Ciclina E/fisiologia , Fragmentação do DNA/efeitos dos fármacos , Feminino , Humanos , Concentração Inibidora 50 , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Neoplasias/fisiologia , Oxirredução , Transporte Proteico/efeitos dos fármacos
8.
Gynecol Oncol ; 143(1): 168-178, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27473926

RESUMO

OBJECTIVE: Endometrial carcinoma is one of the most common malignancies of the female reproductive system, but the aetiology and pathogenesis are not well understood, although adipokines such as visfatin may be involved. Our study provides insight into the mechanism underlying the tumorigenic effects of visfatin in endometrial carcinoma. METHODS: We investigated the effect of visfatin on endometrial carcinoma cell proliferation, cell cycle, and apoptosis using well-differentiated Ishikawa cells and poorly differentiated KLE cells. We also assessed the effect of visfatin on tumour growth in vivo. RESULTS: Visfatin stimulated the proliferation of both Ishikawa and KLE cells, and visfatin treatment promoted G1/S phase progression and inhibited endometrial carcinoma cell apoptosis. Visfatin promoted endometrial carcinoma tumour growth in BALB/c-nu mice. Transplanted tumour tissues from an endometrial carcinoma mouse model were analysed using immunohistochemical staining, which revealed much stronger positive signals for Ki-67 with over-abundant visfatin. Western blot analysis revealed that insulin receptor (IR), insulin receptor substrate (IRS)1/2 and key components of the phosphoinositide 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK)1/2 signalling pathways were highly expressed in endometrial carcinoma cells exposed to visfatin. Treated cells showed increased C-MYC and cyclin D1 and reduced caspase-3 expression. The effects of visfatin on proliferation and apoptosis were abrogated by treatment with the PI3K inhibitor LY294002 and MEK inhibitor U0126. CONCLUSIONS: Visfatin promotes the malignant progression of endometrial carcinoma via activation of IR and PI3K/Akt and MAPK/ERK signalling. Visfatin may serve as a therapeutic target in the treatment of endometrial carcinoma.


Assuntos
Neoplasias do Endométrio/patologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nicotinamida Fosforribosiltransferase/farmacologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/efeitos dos fármacos , Acrilamidas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Camundongos , Inibidores de Fosfoinositídeo-3 Quinase , Piperidinas/farmacologia
9.
Biomed Chromatogr ; 28(6): 729-34, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24861737

RESUMO

Concerned about the safety of conventional estrogen replacement therapy, women are using botanical dietary supplements as alternatives for the management of menopausal symptoms such as hot flashes. Before botanical dietary supplements can be evaluated clinically for safety and efficacy, botanically authenticated and standardized forms are required. To address the demand for a standardized, estrogenic botanical dietary supplement, an extract of hops (Humulus lupulus L.) was developed. Although valued in the brewing of beer, hop extracts are used as anxiolytics and hypnotics and have well-established estrogenic constituents. Starting with a hop cultivar used in the brewing industry, spent hops (the residue remaining after extraction of bitter acids) were formulated into a botanical dietary supplement that was then chemically and biologically standardized. Biological standardization utilized the estrogen-dependent induction of alkaline phosphatase in the Ishikawa cell line. Chemical standardization was based on the prenylated phenols in hops that included estrogenic 8-prenylnaringenin, its isomer 6-prenylnaringenin, and pro-estrogenic isoxanthohumol and its isomeric chalcone xanthohumol, all of which were measured using high-performance liquid chromatography-tandem mass spectrometry. The product of this process was a reproducible botanical extract suitable for subsequent investigations of safety and efficacy.


Assuntos
Suplementos Nutricionais/normas , Estrogênios/química , Estrogênios/normas , Humulus/química , Extratos Vegetais/química , Extratos Vegetais/normas , Linhagem Celular , Suplementos Nutricionais/análise , Estrogênios/farmacologia , Feminino , Humanos , Extratos Vegetais/farmacologia
10.
J ASEAN Fed Endocr Soc ; 38(1): 13-22, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37234927

RESUMO

Objective: To determine whether bisphenol S (BPS), a common substitute for bisphenol A (BPA), induces cell proliferation and migration in human endometrial epithelial cells (Ishikawa) and adult mouse uterine tissues. Methodology: Human endometrial Ishikawa cells were exposed to low doses of BPS (1 nM and 100 nM) for 72 hours. Cell proliferation was assessed through the viability assays MTT and CellTiter-Glo®. Wound healing assays were also used to evaluate the migration potential of the cell line. The expression of genes related to proliferation and migration was also determined. Similarly, adult mice were exposed to BPS at a dose of 30 mg/kg body weight/day for 21 days, after which, the uterus was sent for histopathologic assessment. Results: BPS increased cell number and stimulated migration in Ishikawa cells, in association with the upregulation of estrogen receptor beta (ESR2) and vimentin (VIM). In addition, mice exposed to BPS showed a significantly higher mean number of endometrial glands within the endometrium. Conclusion: Overall, in vitro and in vivo results obtained in this study showed that BPS could significantly promote endometrial epithelial cell proliferation and migration, a phenotype also observed with BPA exposure. Hence, the use of BPS in BPA-free products must be reassessed, as it may pose adverse reproductive health effects to humans.


Assuntos
Endométrio , Útero , Feminino , Camundongos , Humanos , Animais , Linhagem Celular , Células Epiteliais , Contagem de Células
11.
Phytomedicine ; 120: 155062, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37683586

RESUMO

BACKGROUND: Icaritin has a wide range of pharmacological activities, including significant an-titumor activity. However, the mechanism of action of icaritin in endometrial cancer (UCEC) remains unknown. FOX proteins are a highly conserved transcription factor superfamily that play important roles in epithelial cell differentiation, tumor metastasis, angiogenesis, and cell cycle regulation. FOXC1 is an important member of the FOX protein family. FOXC1 is aberrantly expressed in endometrial cancer and may play a role in the migration and invasion of endometrial cancer; however, its mechanism of action has not yet been reported. O-GlcNAc glycosylation is a common post-translational modification. In endometrial cancer, high levels of O-GlcNAcylation promote cell proliferation, migration, and invasion. Cancer development is often accompanied by O-GlcNAc modification of proteins; however, O-GlcNAc modification of the transcription factor FOXC1 has not been reported to date. PURPOSE: To investigate the inhibitory effects of icaritin on RL95-2 and Ishikawa endometrial cancer cells in vitro and in vivo and to elucidate the possible molecular mechanisms. METHODS/STUDY DESIGN: CCK8, colony formation, migration, and invasion assays were used to determine the inhibitory effects of icaritin on endometrial cancer cells in vitro. Cell cycle regulation was assayed by flow cytometry. Protein levels were measured based on western blotting. The level of FOXC1 expression in endometrial cancer tissues was determined by immunohistochemistry. To assess whether icaritin also has activity in vivo, its effect on tumor xenografts was evaluated. RESULTS: Immunohistochemical analysis of clinical samples revealed that FOXC1 expression was significantly higher in endometrial cancer tissues than in normal tissues. Downregulation of FOXC1 inhibited the proliferative, colony formation, migration, and invasive abilities of RL95-2 and Ishikawa endometrial cancer cells. Icaritin inhibited the proliferation, colony formation, migration, and invasion of endometrial cancer cells and blocked the cell cycle in S phase. Icaritin affected O-GlcNAc modification of FOXC1 and thus the stability of FOXC1, which subsequently triggered the inhibition of endometrial cancer cell proliferation. CONCLUSION: The anti-endometrial cancer effect of icaritin is related to the inhibition of abnormal O-GlcNAc modification of FOXC1, which may provide an important theoretical foundation for the use of icaritin against endometrial cancer.


Assuntos
Neoplasias do Endométrio , Humanos , Feminino , Neoplasias do Endométrio/tratamento farmacológico , Flavonoides/farmacologia , Divisão Celular , Proliferação de Células , Fatores de Transcrição Forkhead
12.
Mol Cell Endocrinol ; 543: 111540, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34965452

RESUMO

Hops (Humulus lupulus) is used as an alternative to hormone replacement therapy due to the phytoestrogen, 8-prenylnaringenin (8-PN). To examine the potential risks/benefits of hops extract and its compounds (8-PN and 6-prenylnaringenin, 6-PN), we aimed to evaluate the estrogen receptor α (ERα) and aryl hydrocarbon receptor (AHR) signaling pathways in human endometrial cancer cells. Hops extract, 8-PN and 6-PN showed estrogenic activity. Hops extract and 6-PN activated both ERα and AHR pathways. 6-PN increased the expression of the tumor suppressor gene (AHRR), and that of genes involved in the estrogen metabolism (CYP1A1, CYP1B1). Although 6-PN might activate the detoxification and genotoxic pathways of estrogen metabolism, hops extract as a whole only modulated the genotoxic pathway by an up-regulation of CYP1B1 mRNA expression. These data demonstrate the relevant role of 6-PN contained in the hops extract as potential modulator of estrogen metabolism due to its ERα and AHR agonist activity.


Assuntos
Humulus , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Humanos , Humulus/metabolismo , Extratos Vegetais/farmacologia , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo
13.
Integr Cancer Ther ; 20: 15347354211007560, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33926283

RESUMO

Cancer of the reproductive tract includes diseases with higher prevalence in the female population. This investigation examined whether an anthocyanin-enriched extract of Aristotelia chilensis, commonly known as "maqui," could affect some hallmarks of endometrial cancer. Cultures of the human endometrial cancer cell line Ishikawa were treated with a hydroethanolic maqui extract at 1, 3, 10, 30, 100, 300, or 1000 µg/mL to determine the effect on cell viability by MTT assay. Then, we used the 50% Effective Concentration (EC50) to evaluate whether the effect of the maqui extract is mediated via an arrest of the cell cycle or induction of apoptosis using flow cytometry or Annexin V-FITC assays, respectively. The effects of sublethal doses of the maqui extract on migration and invasiveness of Ishikawa cells were also evaluated by the wound healing and Boyden Chamber assay, respectively. Our results show that the hydroethanolic maqui extract inhibits the cell viability with an EC50 of 472.3 µg/mL via increased apoptosis, and that reduces the invasive capacity but not migration of Ishikawa cells. These findings suggest that the hydroethanolic maqui extract has antineoplastic properties for endometrial cancer and merits further studies to corroborate its efficiency as anticancer therapy in reproductive organs.


Assuntos
Neoplasias do Endométrio , Frutas , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Neoplasias do Endométrio/tratamento farmacológico , Humanos , Extratos Vegetais/farmacologia
14.
Front Endocrinol (Lausanne) ; 12: 798866, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35185785

RESUMO

Follicle-stimulating hormone (FSH) and its G protein-coupled receptor, FSHR, represents a paradigm for receptor signaling systems that activate multiple and complex pathways. Classically, FSHR activates Gαs to increase intracellular levels of cAMP, but its ability to activate other G proteins, and ß-arrestin-mediated signaling is well documented in many different cell systems. The pleiotropic signal capacity of FSHR offers a mechanism for how FSH drives multiple and dynamic downstream functions in both gonadal and non-gonadal cell types, including distinct diseases, and how signal bias may be achieved at a pharmacological and cell system-specific manner. In this study, we identify an additional mechanism of FSH-mediated signaling and downstream function in the endometrial adenocarcinoma Ishikawa cell line. While FSH did not induce increases in cAMP levels, this hormone potently activated pertussis toxin sensitive Gαi/o signaling. A selective allosteric FSHR ligand, B3, also activated Gαi/o signaling in these cells, supporting a role for receptor-mediated activation despite the low levels of FSHR mRNA. The low expression levels may attribute to the lack of Gαs/cAMP signaling as increasing FSHR expression resulted in FSH-mediated activation of the Gαs pathway. Unlike prior reports for FSH-mediated Gαs/cAMP signaling, FSH-mediated Gαi/o signaling was not affected by inhibition of dynamin-dependent receptor internalization. While chronic FSH did not alter cell viability, FSH was able to increase lipid droplet size. The ß-arrestins are key adaptor proteins known to regulate FSHR signaling. Indeed, a rapid, FSH-dependent increase in interactions between ß-arrestin1 and Gαi1 was observed via NanoBiT complementation in Ishikawa cells. Furthermore, both inhibition of Gαi/o signaling and siRNA knockdown of ß-arrestin 1/2 significantly reduced FSH-induced lipid droplet accumulation, implying a role for a Gαi/o/ß-arrestin complex in FSH functions in this cell type. As FSH/FSHR has been implicated in distinct hormone-dependent cancers, including endometrial cancer, analysis of the cancer genome database from 575 human endometrial adenocarcinoma tumors revealed that a subpopulation of samples expressed FSHR. Overall, this study highlights a novel mechanism for FSHR signal pleiotropy that may be exploited for future personalized therapeutic approaches.


Assuntos
Neoplasias do Endométrio , Hormônio Foliculoestimulante , Linhagem Celular , Feminino , Hormônio Foliculoestimulante/metabolismo , Humanos , Gotículas Lipídicas/metabolismo , Receptores do FSH/genética , Receptores do FSH/metabolismo , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo
15.
Front Cell Dev Biol ; 9: 709902, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34434932

RESUMO

Embryo implantation is a complex and tightly regulated process. In humans, uterine luminal epithelium functions as a biosensor gauging the embryo quality and transmitting this information to the underlying endometrial stromal cells. This quality control ensures that only high quality embryos are implanted, while aberrant ones are rejected. The mechanisms of the embryo-uterine mucosa crosstalk remain incompletely understood. Trypsin, a serine protease secreted by the blastocyst, has been implicated in the cross-signaling. Here we address the mechanisms by which trypsin triggers the intracellular calcium signaling in uterine epithelium. We found that protease-activated G-protein coupled receptors are the main mechanism mediating the effects of trypsin in human uterine epithelium. In addition, trypsin activates the epithelial sodium channels thus increasing the intracellular Na+ concentration and promoting Ca2+ entry on the reverse mode of the sodium/calcium exchanger.

16.
Oncol Lett ; 20(5): 249, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32994812

RESUMO

Metformin (MET) constitutes the first-line treatment against type 2 diabetes. Growing evidence linking insulin resistance and cancer risk has expanded the therapeutic potential of MET to several cancer types. However, the oncostatic mechanisms of MET are not well understood. MET has been shown to promote the expression of progesterone receptor (PGR) and other antitumor biomarkers in patients with non-diabetic endometrial cancer (EC) and in Ishikawa EC cells cultured in normal glucose (5.5 mM) media. Therefore, the present study aimed to assess the effects of MET on EC cells under conditions simulating diabetes. Ishikawa cells treated with 10 nM 17ß-estradiol (E2) and/or 100 µM MET and exposed to normal and high (17.5 mM) concentrations of glucose were evaluated for proliferative and PGR expression status. Under normal glucose conditions, MET attenuated E2-induced cell proliferation and cyclin D1 gene expression, and increased total PGR and PGR-B transcript levels. MET inhibited Ishikawa cell spheroid formation only in the absence of E2 treatment. In E2-treated cells under high glucose conditions, MET showed no effects on cell proliferation and spheroid formation, and increased total PGR but not PGR-B transcript levels. Transfection with Krüppel-like factor 9 small interfering RNA increased PGR-A transcript levels, irrespective of glucose environment. Medroxyprogesterone acetate downregulated PGR-A expression more effectively with metformin under high compared with normal glucose conditions. To evaluate the potential mechanisms underlying the targeting of PGR by MET, E2-treated cells were incubated with MET and the AMPK inhibitor Compound C, or with the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), under normal glucose conditions. Compound C abrogated the effects of MET on PGR-B while AICAR increased PGR-B transcript levels, albeit less effectively compared with MET. The present results demonstrate the glucose-dependent effects of MET on PGR-B isoform expression, which may inform the response to progestin therapy in diabetic women with EC.

17.
Biochem Biophys Rep ; 22: 100740, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32099910

RESUMO

Endometrial cancer is the most common gynecologic malignancy and is associated with increased morbidity each year, including young people. However, its mechanisms of proliferation and progression are not fully elucidated. It is well known that abnormal glycosylation is involved in oncogenesis, and fucosylation is one of the most important types of glycosylation. In particular, fucosyltransferase 8 (FUT8) is the only FUT responsible for α1, 6-linked fucosylation (core fucosylation), and it is involved in various physiological as well as pathophysiological processes, including cancer biology. Therefore, we aimed to identify the expression of FUT8 in endometrial endometrioid carcinoma and investigate the effect of the partial silencing of the FUT8 gene on the cell proliferation of Ishikawa cells, an epithelial-like endometrial cancer cell line. Quantitative real-time PCR analysis showed that FUT8 gene expression was significantly elevated in the endometrial endometrioid carcinoma, compared to the normal endometrium. The immunostaining of FUT8 and Ulex europaeus Agglutinin 1 (UEA-1), a kind of lectin family specifically binding to fucose, was detected endometrial endometrioid carcinoma. The proliferation assay showed FUT8 partial knockdown by transfection of siRNA significantly suppressed the proliferation of Ishikawa cells, concomitant with the upregulation in the gene expressions associated with the interesting pathways associated with de-ubiquitination, aspirin trigger, mesenchymal-epithelial transition (MET) et al. It was suggested that the core fucosylation brought about by FUT8 might be involved in the proliferation of endometrial endometrioid carcinoma cells.

18.
Cells ; 9(5)2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32403233

RESUMO

Embryo implantation into the uterine wall is a highly modulated, complex process. We previously demonstrated that Annexin A1 (AnxA1), which is a protein secreted by epithelial and inflammatory cells in the uterine microenvironment, controls embryo implantation in vivo. Here, we decipher the effects of recombinant AnxA1 in this phenomenon by using human trophoblast cell (BeWo) spheroids and uterine epithelial cells (Ishikawa; IK). AnxA1-treated IK cells demonstrated greater levels of spheroid adherence and upregulation of the tight junction molecules claudin-1 and zona occludens-1, as well as the glycoprotein mucin-1 (Muc-1). The latter effect of AnxA1 was not mediated through IL-6 secreted from IK cells, a known inducer of Muc-1 expression. Rather, these effects of AnxA1 involved activation of the formyl peptide receptors FPR1 and FPR2, as pharmacological blockade of FPR1 or FPR1/FPR2 abrogated such responses. The downstream actions of AnxA1 were mediated through the ERK1/2 phosphorylation pathway and F-actin polymerization in IK cells, as blockade of ERK1/2 phosphorylation reversed AnxA1-induced Muc-1 and claudin-1 expression. Moreover, FPR2 activation by AnxA1 induced vascular endothelial growth factor (VEGF) secretion by IK cells, and the supernatant of AnxA1-treated IK cells evoked angiogenesis in vitro. In conclusion, these data highlight the role of the AnxA1/FPR1/FPR2 pathway in uterine epithelial control of blastocyst implantation.


Assuntos
Anexina A1/metabolismo , Blastocisto/metabolismo , Receptores de Formil Peptídeo/metabolismo , Útero/fisiologia , Actinas/metabolismo , Animais , Linhagem Celular , Claudina-1/metabolismo , Implantação do Embrião , Células Epiteliais/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL , Mucina-1/metabolismo , Neovascularização Fisiológica , Polimerização , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Trofoblastos/citologia , Trofoblastos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
19.
Practical Oncology Journal ; (6): 395-402, 2023.
Artigo em Chinês | WPRIM | ID: wpr-1020870

RESUMO

Objective The aim of this study was to investigate the effects of β-elemene on the biological behavior of endo-metrial cancer Ishikawa cells and its mechanism of action.Methods Different concentrations(0,50,100,150,200,250,300 μg/mL)of β-elemene were used to treat Ishikawa cells.The cell viability was measured using the CCK-8 assay to calculate the half-maxi-mal inhibitory concentration(IC50)of β-elemene and to determining low,medium,and high treatment concentrations.A 0 μg/mL of β-elemene was set up a blank control group and 2 μg/mL of cisplatin for a positive control group.The apoptotic rate was detected u-sing Annexin V-FITC/PI double staining.The scratch wound healing assay and Transwell assay were used to detect the abilities of cell migration and invasion.Western blot was performed to evaluate the expression of PI3 K/Akt/mTOR signaling pathway related pro-teins in Ishikawa cells.Results β-Elemene significantly inhibited the proliferation of Ishikawa cells in a dose-dependent manner.The IC50 values of β-elemene at 24 and 48 hours were 168.9 μg/mL and 157.1 μg/mL,respectively.β-Elemene induced apoptosis in Ishikawa cells and after treatment with β-elemene at 0,85,170,and 255 μg/mL for 24 hours,the apoptotic rates of Ishikawa cells were(9.41±0.52)%,(16.67±0.25)%,(21.27±0.30)%and(25.55±0.89)%,respectively.There was a statistically signifi-cant difference in multiple comparisons between groups(P<0.001).β-Elemene inhibited the migration and invasion ability of Ish-ikawa cells,and the inhibitory effects increased with concentration(P<0.001).In addition,β-elemene reduced the levels of PI3K,p-PI3K,mTOR and p-mTOR proteins in Ishikawa cells(P<0.05),and the expression of p-PI3K and p-mTOR proteins showed a significant concentration dependence,and β-elemene at the 255 μg/mL of group showed a significant reduction in the expression of PI3K,p-PI3K,Akt and p-mTOR when compared to the 0 μg/mL of β-elemene group(P<0.001).Conclusion β-Elemene can inhibit the proliferation,migration,and invasion of endometrial cancer Ishikawa cells,promote their apoptosis,and its mechanism may be related to the inhibitory activation of the PI3 K/Akt/mTOR signaling pathway in Ishikawa cells.

20.
Biochem Insights ; 10: 1178626417721676, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28804245

RESUMO

Based on a previous study, glabridin displayed a dose-dependent increase in estrogenic activity and cell proliferative activity in Ishikawa cells. However, when treated in combination with 17ß-E2, synergistic estrogenic effect was observed but without the same synergistic increase in cell proliferative effect. This study aimed to identify the estrogen and nonestrogen-regulated activities induced by glabridin and in combination with 17ß-E2 in comparison with 17ß-E2. The results showed that 10 µM glabridin and the combination treatment of 100 nM glabridin with 1 nM 17ß-E2 regulated both the genomic and nongenomic estrogen pathways to possibly provide benefits of estrogens in cardiovascular, circulatory, and vasculature systems. Meanwhile, the combination of 100 nM glabridin with 1 nM 17ß-E2 seems to be more suitable to be used as an estrogen replacement. Finally, the results of this study have added on to the present knowledge of glabridin's function as a phytoestrogen and suggested new ideas for the usage of glabridin.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa