Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Phytomedicine ; 113: 154681, 2023 May.
Article in English | MEDLINE | ID: mdl-36893674

ABSTRACT

BACKGROUND: Ginsenoside Re is an active component in ginseng that confers protection against myocardial ischemia/reperfusion (I/R) injury. Ferroptosis is a type of regulated cell death found in various diseases. PURPOSE: Our study aims to investigate the role of ferroptosis and the protective mechanism of Ginsenoside Re in myocardial ischemia/reperfusion. METHODS: In the present study, we treated rats for five days with Ginsenoside Re, then established the myocardial ischemia/reperfusion injury rat model to detect molecular implications in myocardial ischemia/reperfusion regulation and to determine the underlying mechanism. RESULTS: This study identifies the mechanism behind ginsenoside Re's effect on myocardial ischemia/reperfusion injury and its regulation of ferroptosis through miR-144-3p. Ginsenoside Re significantly reduced cardiac damage caused by ferroptosis during myocardial ischemia/reperfusion injury and glutathione decline. To determine how Ginsenoside Re regulated ferroptosis, we isolated exosomes from VEGFR2+ endothelial progenitor cells after ischemia/reperfusion injury and performed miRNA profiling to screen the miRNAs aberrantly expressed in the process of myocardial ischemia/reperfusion injury and ginsenoside Re treatment. We identified that miR-144-3p was upregulated in myocardial ischemia/reperfusion injury by luciferase report and qRT-PCR. We further confirmed that the solute carrier family 7 member 11 (SLC7A11) was the target gene of miR-144-3p by database analysis and western blot. In comparison with ferropstatin-1, a ferroptosis inhibitor, in vivo studies confirmed that ferropstatin-1 also diminished myocardial ischemia/reperfusion injury induced cardiac function damage. CONCLUSION: We demonstrated that ginsenoside Re attenuates myocardial ischemia/reperfusion induced ferroptosis via miR-144-3p/SLC7A11.


Subject(s)
Ferroptosis , MicroRNAs , Myocardial Ischemia , Myocardial Reperfusion Injury , Reperfusion Injury , Rats , Animals , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Ischemia
2.
Exp Ther Med ; 24(3): 545, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35978936

ABSTRACT

Coronary heart disease (CHD) mainly refers to coronary atherosclerotic heart disease and its pathogenesis is complex. Ginsenoside Rg1 (Rg1) has a wide range of pharmacological activities, such as antitumor effects, enhancing immunity and exerting protective effects on the vascular system. In the present study, the effect of Rg1 on vascular endothelial cells in CHD was investigated. Oxidized low-density lipoprotein (ox-LDL) was used to induce human umbilical vein endothelial cells (HUVECs) and cells were treated with 1, 5 or 10 µM Rg1. Cell Counting Kit-8 assay, TUNEL staining, western blot analysis of apoptosis-related proteins and senescence-related proteins, senescence-associated ß-galactosidase staining, ELISA and other techniques including related kits of oxidative stress markers were used to detect the viability, apoptosis, oxidative stress, inflammatory cytokines including IL-1ß, IL-6 and TNF-α and senescence of ox-LDL-induced HUVECs induced by Rg1. Western blot analysis was used to detect the expression levels of the AMP-activated protein kinase (AMPK)/sirtuin 3 (SIRT3)/p53 signaling pathway-related proteins. In addition, the associated mechanism was further determined using the AMPK pathway inhibitor compound C (CC). Rg1 increased the viability, and inhibited the apoptosis, senescence, oxidative stress and inflammation of ox-LDL-induced HUVECs. Pretreatment with CC partially reversed the protective effect of Rg1 on ox-LDL-induced HUVECs. In conclusion, Rg1 ameliorated apoptosis, senescence and oxidative stress of ox-LDL-induced HUVECs, at least in part, via the AMPK/SIRT3/p53 signaling pathway.

3.
Front Cardiovasc Med ; 9: 1099043, 2022.
Article in English | MEDLINE | ID: mdl-36588555

ABSTRACT

Objective: To investigate the effectiveness and safety of sacubitril valsartan sodium in the treatment of resistant hypertension (RH). Methods: This study is a single-center, prospective, randomized controlled study. According to the inclusion and exclusion criteria, patients with RH who met the criteria were screened, and all patients adjusted their drug treatment (valsartan 80 mg, amlodipine 5 mg, and hydrochlorothiazide 12.5 mg). After 4 weeks of drug elution, the random envelope method was used for random grouping. The treatment group took sacubitril valsartan sodium 200 mg, amlodipine 5 mg, hydrochlorothiazide 12.5 mg, and the control group took valsartan 80 mg, amlodipine 5 mg, and hydrochlorothiazide 12.5 mg for 8 weeks. The 24 h ambulatory blood pressure (BP) and the echocardiography index using the office sphygmomanometer were observed in the patients. Results: A total of 100 patients with RH were included in the two groups, with 50 cases in each group. There were no significant differences in sex, age, or comorbid diseases between the two groups. During the 8-week follow-up, the office BP of the research group were significantly decreased (24.78/17.86 mmHg) compared with those of the control group. In the research group the 24 h average BP, daytime average BP, and nighttime average BP were 144.84/79.82, 147.10/82.06, and 138.67/76.31 mmHg at baseline, and reduced to 128.96/73.32, 131.50/74.94, and 122.11/69.27 mmHg at week 8, which were significantly decreased (P < 0.05 or P < 0.01), and the left ventricular ejection fraction was significantly increased (P < 0.05), compared with the control group. Conclusion: Sacubitril valsartan sodium can effectively reduce BP and improve cardiac function in RH.

4.
Front Oncol ; 11: 690618, 2021.
Article in English | MEDLINE | ID: mdl-34621667

ABSTRACT

BACKGROUND: Implantation metastasis is the main means of dissemination in ovarian cancer. Our previous studies showed that SET and MYND domain-containing protein 3 (SMYD3) expression was higher in ovarian cancer spheroids than in monolayers. SMYD3 enhancement of spheroid invasion and adhesion is mediated by the downstream effectors ITGB6 and ITGAM. However, the potential mechanisms underlying the SMYD3/integrin-mediated invasion and adhesion of spheroids still need to be explored. METHODS: Western blotting was used to examine the expression of SMYD3, ITGB6 and downstream molecules under different treatments. Immunofluorescence was used to detect the expression of F-actin, E-cadherin and N-cadherin. Anti-ITGB6 antibody-based inhibition and dual-luciferase reporter assays were used to confirm the binding between ITGB6 and latent TGFß1. Transwell invasion, adherence and 3D tumor spheroid invasion assays were employed to test the effects of TGFß1 on the invasion and adhesion of ovarian cancer spheroids. ELISA was performed to assess the release of latent TGFß1 from ovarian cancer spheroids. RESULTS: SMYD3 and ITGB6 activated the TGFß1/Smad3 pathway and then induced the upregulation of Snail, Vimentin and N-cadherin and the downregulation of E-cadherin in 3D-cultured ovarian cancer spheroids. In this process, latent TGFß1 could bind to ITGB6 and become activated to stimulate the Smad3 pathway. Moreover, SMYD3 and ITGB6 could facilitate the release of latent TGFß1 from 3D-cultured ovarian cancer spheroids. Interestingly, TGFß1 could promote the expression of SMYD3 and ITGB6 via feedback. This positive feedback loop could further amplify the biological effect and promote the invasion and adhesion of ovarian cancer spheroids. CONCLUSION: Our results demonstrated that the SMYD3/ITGB6/TGFß1-Smad3 positive feedback loop could promote the invasion and adhesion of ovarian cancer spheroids by upregulating the expression of N-cadherin, Snail, and Vimentin and downregulating the expression of E-cadherin. Thus, our study unmasked the mechanism of SMYD3- and ITGB6-induced ovarian cancer metastasis and provides new ideas for targeted ovarian cancer treatment.

5.
Cancer Manag Res ; 13: 6585-6597, 2021.
Article in English | MEDLINE | ID: mdl-34456588

ABSTRACT

PURPOSE: Ovarian cancer, manifested by malignant ascites, is the most lethal gynaecological cancer. Suspended ascites-derived spheroids may contribute to ovarian cancer metastasis. MicroRNAs (miRNAs) are also associated with ovarian cancer metastasis. Here, we aimed to investigate the differentially expressed miRNAs (DE-miRNAs) in ascites-derived spheroids compared with primary tumour tissues, which may regulate ovarian cancer metastasis. METHODS: The DE-miRNAs between ovarian cancer primary tumour tissues and ascites-derived spheroids were identified by GEO2R screening in samples from 3 high-grade serous ovarian cancer (HGSOC) patients of dataset GSE65819. We used MiRTarBase, TargetScanHuman7.2 and STRING to predict the target hub genes of DE-miRNAs and DAVID to perform functional analysis of hub genes. ALGGEN PROMO and TransmiR v2.0 were used to predict transcription factors (TFs) that potentially regulate DE-miRNAs expression. The observed differences in DE-miRNAs expression were validated with samples from 12 HGSOC patients and 2 ovarian cancer cell lines using PCR. The functions of DE-miRNAs on ovarian cancer progression were verified by invasion, adherent, and angiogenesis assays. RESULTS: Through bioinformatics screening and experimental validation, miR-199a-3p, miR-199b-3p, miR-199a-5p, miR-126-3p and miR-145-5p were identified as being significantly downregulated in ascites-derived spheroids compared with primary tumour tissues. In addition, TFAP2A was identified as a potentially common upstream TF regulating the expression of the above mentioned DE-miRNAs. The overexpression of miR-199a-3p, miR-199b-3p, miR-199a-5p lead to invasion inhibition, and the overexpression of miR-126-3p, miR-145-5p, miR-199a-5p and miR-199b-3p lead to adhesion inhibition of suspended ovarian cancer cells. High-expressed miR-126-3p, miR-199a-3p, miR-199a-5p and miR-199b-3p contributed to apoptosis of suspended ovarian cancer cells. CONCLUSION: The downregulated expression of miR-199a-3p, miR-199b-3p, miR-199a-5p, miR-126-3p and miR-145-5p in ascites-derived spheroids plays a key role in promoting ovarian cancer progression, which may represent novel molecules for targeted therapy for ovarian cancer.

6.
Int J Cancer ; 146(6): 1553-1567, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31503345

ABSTRACT

Detachment of cancer cells from the primary tumor and formation of spheroids in ascites is required for implantation metastasis in epithelial ovarian cancer (EOC), but the underlying mechanism of this process has not been thoroughly elucidated. To mimic this process, ovarian cancer cells were grown in 3D and 2D culture. Hey and OVCA433 spheroids exhibited decreased cell proliferation and enhanced adhesion and invasion. SMYD3 expression was elevated in ovarian carcinoma spheroids in association with increased H3K4 methylation. Depletion of SMYD3 by transient siRNA, stable shRNA knockdown and the SMYD3 inhibitor BCI-121 all decreased spheroid invasion and adhesion. Gene expression arrays revealed downregulation of integrin family members. Inhibition assays confirmed that invasion and adhesion of spheroids are mediated by ITGB6 and ITGAM. SMYD3-deficient cells regained the ability to invade and adhere after forced overexpression of SMYD3, ITGB6 and ITGAM. However, this biological ability was not restored by forced overexpression of SMYD3 in ITGB6- and/or ITGAM-deficient cancer cells. SMYD3 and H3K4me3 binding at the ITGB6 and ITGAM promoters was increased in spheroids compared to that in monolayer cells, and the binding was decreased when SMYD3 expression was inhibited, consistent with the expression changes in integrins. SMYD3 expression and integrin-mediated adhesion were also activated in an intraperitoneal xenograft model and in EOC patient spheroids. In vivo, SMYD3 knockdown inhibited tumor metastasis and reduced ascites volume in both the intraperitoneal xenograft model and a PDX model. Overall, our results suggest that the SMYD3-H3K4me3-integrin pathway plays a crucial role in ovarian cancer metastasis to the peritoneal surface.


Subject(s)
Ascites/pathology , Carcinoma, Ovarian Epithelial/secondary , Histone-Lysine N-Methyltransferase/metabolism , Integrins/genetics , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Adult , Aged , Ascites/etiology , Carcinoma, Ovarian Epithelial/genetics , Cell Adhesion/genetics , Cell Culture Techniques , Cell Line, Tumor , DNA Methylation , Down-Regulation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Middle Aged , Ovarian Neoplasms/genetics , Peritoneal Neoplasms/genetics , Promoter Regions, Genetic/genetics , Spheroids, Cellular , Xenograft Model Antitumor Assays
7.
J Cancer ; 10(17): 4072-4084, 2019.
Article in English | MEDLINE | ID: mdl-31417652

ABSTRACT

Background: Epigenetic regulation has been verified as a key mechanism in tumorigenesis. SET and MYND domain-containing protein 3 (SMYD3), a histone methyltransferase, is a promising epigenetic therapeutic target and is overexpressed in numerous human tumors. SMYD3 can promote oncogenic progression by methylating lysines to integrate cytoplasmic kinase signaling cascades or by methylating histone lysines to regulate specific gene transcription. However, the exact role of SMYD3 in the progression of ovarian cancer is still unknown. Methods: Immunohistochemistry was employed to test SMYD3 expression in ovarian cancer tissues from clinical patients. CCK-8 assay, Real-time cell analysis (RTCA), colony formation assay, cell cycle and apoptosis tested by Flow cytometer were employed to test the effects of SMYD3 on cell proliferation and apoptosis in ovarian cancer cell lines. A PCR array was used to identify the downstream targets of SMYD3. And, PCR and Western blot were used to verify their expression. The binding of SMYD3 on the promoter of target genes were tested by ChIP assays. We also use nude mice subcutaneous tumor model and patient-derived xenograft (PDX) model to investigate the tumor promotive function of SMYD3 in vivo. Results: SMYD3 expression was higher in ovarian cancer tissues and cell lines than in normal ovarian epithelial tissue and human ovarian surface epithelial cells (HOSEpiC). After silencing SMYD3, the proliferation of ovarian cancer cells was significantly inhibited in vitro. In addition, the SMYD3-specific small-molecule inhibitor BCI-121 suppressed ovarian cancer cell proliferation. Downregulation of SMYD3 led to S phase arrest and increased the cell apoptosis rate. Furthermore, a PCR array revealed that SMYD3 knockdown caused the upregulation of the cyclin-dependent kinase (CDK) inhibitors CDKN2A (p16INK4), CDKN2B (p15INK4B), CDKN3 and CDC25A, which may be responsible for the S phase arrest. In addition, the upregulation of CD40LG and downregulation of BIRC3 may explain the increased cell apoptosis rate after silencing SMYD3. We also discovered that SMYD3 bound on the promoter of CDKN2A and down-regulated its expression by triple-methylating H4K20. In addition, SMYD3 bound on the promoter of BIRC3 and up-regulated its expression by triple-methylating H3K4. Finally, knocking down SMYD3 could inhibit ovarian cancer growth in nude mice subcutaneous tumor model and PDX model. Conclusion: Our results demonstrated that SMYD3 was overexpressed in ovarian cancer and contributes to the regulation of tumor proliferation and apoptosis via SMYD3-H4K20me3-CDKN2A pathway and SMYD3-H3K4me3-BIRC3 pathway. Thus, SMYD3 is a promising epigenetic therapeutic target for ovarian cancer.

8.
J Int Med Res ; 47(6): 2492-2498, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31020889

ABSTRACT

OBJECTIVE: This study aimed to retrospectively investigate the safety of ovarian preservation of premenopausal women with stage 1a endometrial carcinoma. METHODS: We performed a population-based study to identify surgically treated stage Ia endometrial cancer of premenopausal women who were diagnosed between August 1989 and December 2015 in our center. Survival outcomes and recurrence rate were examined for premenopausal women who underwent ovarian preservation. Recurrence-free survival rates were calculated following generation of Kaplan-Meier curves and were compared with the log-rank test. Cox regression analysis was performed to identify the independent factors affecting the recurrence rate. RESULTS: Patients with ovarian preservation tended to be significantly younger at diagnosis, have less myometrial invasion, and were less likely to undergo lymphadenectomy compared with women treated with bilateral salpingo-oophorectomy. There was no significant difference in recurrence-free survival between the two groups. In the Cox regression model, ovarian preservation remained an independent prognostic factor for improved overall survival. CONCLUSION: Ovarian preservation does not have a negative effect on oncological outcomes. Ovarian preservation can be applied to premenopausal women with stage Ia endometrial carcinoma.


Subject(s)
Endometrial Neoplasms/surgery , Fertility Preservation/statistics & numerical data , Neoplasm Recurrence, Local/surgery , Organ Sparing Treatments/methods , Ovary/surgery , Premenopause , Adult , Case-Control Studies , Endometrial Neoplasms/mortality , Endometrial Neoplasms/pathology , Female , Follow-Up Studies , Humans , Hysterectomy/mortality , Lymph Node Excision/mortality , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Ovariectomy/mortality , Retrospective Studies , Survival Rate , Treatment Outcome
9.
J Exp Psychol Gen ; 148(11): 1882-1897, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30802096

ABSTRACT

The Dunning-Kruger effect (DKE) is the finding that, across a wide range of tasks, poor performers greatly overestimate their ability, whereas top performers make more accurate self-assessments. The original account of the DKE involves the idea that metacognitive insight requires the same skills as task performance, so that unskilled people perform poorly and lack insight. However, global measures of self-assessment are prone to statistical and other biases that could explain the same pattern. We used psychophysical methods to examine metacognitive insight in simple movement and spatial memory tasks: pointing at a dot or recalling its position after a delay. We measured task skill in an initial block, and self-assessment in a second block, in which participants judged after every trial whether they had hit the target or not. Metacognitive calibration and sensitivity were related to task skill, but a path analysis showed that their net contribution to the DKE was weak. The major driver of the DKE was the level of task performance. In a second study, we again measured task skill in an initial block, but titrated task difficulty in the second block so that all participants performed at equivalent levels of success. Metacognitive measures were again related to task skill, but the DKE pattern was eliminated. We present a simple model of these findings, showing that metacognitive differences can contribute to the DKE but are neither necessary nor sufficient for it. This analysis clarifies and quantifies how metacognitive insight and other factors interact to determine this famous effect. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Subject(s)
Metacognition/physiology , Psychomotor Performance/physiology , Self-Assessment , Adult , Female , Humans , Male , Spatial Memory/physiology , Young Adult
10.
Mol Med Rep ; 10(5): 2433-40, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25216292

ABSTRACT

Previous studies by our group revealed that the phosphoinositide 3­kinase (PI3K)/AKT pathway was involved in estrogen­induced metastasis in ovarian cancer cells. In the present study, the role and mechanism of estrogen­induced invasion was further explored using a stable short hairpin RNA (shRNA) estrogen receptor α/ß (ER α/ß) SKOV3 cell line when ER α and ER ß were knocked down by lentiviral infection. The effects of estrogen and LY294002, a PI3K inhibitor, on the invasion of shRNA ER α/ß SKOV3 cells were evaluated in vitro and in vivo. 17­ß estradiol promoted cell invasion, activated phosphorylated AKT in a dose­ and time­dependent manner, decreased E­cadherin and increased cytoplasmic α­actinin­4 expression. When the PI3K/AKT pathway was suppressed by LY294002, the effect of estrogen was attenuated. Estrogen stimulated the growth of shRNA ER α/ß SKOV3 xenograft tumors in nude mice, whereas LY294002 inhibited the growth and antagonized the effect of estrogen. The results indicate that estrogen promotes the invasion of ovarian cancer cells via activation of the PI3K/AKT pathway, downregulation of E­cadherin and upregulation of α­actinin­4 in an ER­independent manner. Inhibiting the PI3K/AKT pathway may be a useful treatment for ovarian carcinoma.


Subject(s)
Actinin/metabolism , Cadherins/metabolism , Estrogens/physiology , Ovarian Neoplasms/metabolism , Actinin/genetics , Animals , Antigens, CD , Cadherins/genetics , Cell Line, Tumor , Female , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
11.
Epigenetics ; 8(12): 1330-46, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24135786

ABSTRACT

The initiation of angiogenesis can mark the transition from tumor dormancy to active growth and recurrence. Mechanisms that regulate recurrence in human cancers are poorly understood, in part because of the absence of relevant models. The induction of ARHI (DIRAS3) induces dormancy and autophagy in human ovarian cancer xenografts but produces autophagic cell death in culture. The addition of VEGF to cultures maintains the viability of dormant autophagic cancer cells, thereby permitting active growth when ARHI is downregulated, which mimics the "recurrence" of growth in xenografts. Two inducible ovarian cancer cell lines, SKOv3-ARHI and Hey-ARHI, were used. The expression level of angiogenesis factors was evaluated by real-time PCR, immunohistochemistry, immunocytochemistry and western blot; their epigenetic regulation was measured by bisulfite sequencing and chromatin immunoprecipitation. Six of the 15 angiogenesis factors were upregulated in dormant cancer cells (tissue inhibitor of metalloproteinases-3, TIMP3; thrombospondin-1, TSP1; angiopoietin-1; angiopoietin-2; angiopoietin-4; E-cadherin, CDH1). We found that TIMP3 and CDH1 expression was regulated epigenetically and was related inversely to the DNA methylation of their promoters in cell cultures and in xenografts. Increased H3K9 acetylation was associated with higher TIMP3 expression in dormant SKOv3-ARHI cells, while decreased H3K27me3 resulted in the upregulation of TIMP3 in dormant Hey-ARHI cells. Elevated CDH1 expression during dormancy was associated with an increase in both H3K4me3 and H3K9Ac in two cell lines. CpG demethylating agents and/or histone deacetylase inhibitors inhibited the re-growth of dormant cancer cells, which was associated with the re-expression of anti-angiogenic genes. The expression of the anti-angiogenic genes TIMP3 and CDH1 is elevated during dormancy and is reduced during the transition to active growth by changes in DNA methylation and histone modification.


Subject(s)
Epigenesis, Genetic , Neoplasm Recurrence, Local/genetics , Neoplasms, Glandular and Epithelial/genetics , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/genetics , Acetylation , Animals , Autophagy , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Heterografts , Histones/metabolism , Humans , Mice , Mice, Nude , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Promoter Regions, Genetic
12.
PLoS One ; 8(5): e63731, 2013.
Article in English | MEDLINE | ID: mdl-23667667

ABSTRACT

Traditional Chinese medicine has gained popularity due to its ability to kill tumor cells. Recently, the apoptotic and anti-angiogenic effects of Trametes robiniophila murr (Huaier) have been investigated. The aim of this study was to investigate its effect on cell mobility and tumor growth in ovarian cancer. Cell viability and motility were measured using SRB, scratch and migration assays. Cell apoptosis was analysed by annexin V/PI staining. Using a reverse-phase protein array (RPPA) assay, we analyzed the levels of 153 proteins and/or phosphorylations in Huaier-treated and untreated cells. Huaier inhibited cell viability and induced both early and late apoptosis in SKOV3, SKOV3.ip1 and Hey cells in a time- and dose-dependent manner. Cell invasiveness and migration were also suppressed significantly. The RPPA results showed significant differences (of at least 30%; P <0.05) in the levels of 7 molecules in SKOV3 cells and 10 in SKOV3.ip1 cells between the untreated and treated cells. Most of the molecules identified play roles in cell proliferation, apoptosis or cell adhesion/invasion. Western blot analysis further validated that Huaier treatment resulted in decreased AKT phosphorylation, enhanced expression of total GSK3ß, inhibition of the phosphorylation of GSK3ß on S9, reduction of both cytoplasmic ß-catenin expression and nuclear ß-catenin translocation, and transcriptional repression of several Wnt/ß-catenin target genes (DIXDC1, LRP6, WNT5A, and cyclin D1). After knocking down GSK3ß, ß-catenin expression could not be inhibited by Huaier. Finally, Huaier inhibited the growth of ovarian tumor xenografts in vivo. These studies indicate that Huaier inhibits tumor cell mobility in ovarian cancer via the AKT/GSK3ß/ß-catenin signaling pathway.


Subject(s)
Cell Movement/drug effects , Gene Expression Regulation/drug effects , Ovarian Neoplasms/physiopathology , Signal Transduction/drug effects , Tissue Extracts/pharmacology , Trametes/chemistry , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/drug effects , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Medicine, Chinese Traditional , Protein Array Analysis , Proto-Oncogene Proteins c-akt/metabolism , Tissue Extracts/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...