Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Chin Med J (Engl) ; 134(2): 200-205, 2021 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-33443938

RESUMO

BACKGROUND: It has been a global trend that increasing complications related to pelvic floor surgeries have been reported over time. The current study aimed to outline the development of Chinese pelvic floor surgeries related to pelvic organ prolapse (POP) over the past 14 years and investigate the potential influence of enhanced monitoring conducted by the Chinese Association of Urogynecology since 2011. METHODS: A total of 44,594 women with POP who underwent pelvic floor surgeries between October 1, 2004 and September 30, 2018 were included from 22 tertiary academic medical centers. The data were reported voluntarily and obtained from a database. We compared the proportion of each procedure in the 7 years before and 7 years after September 30, 2011. The data were analyzed by performing Z test (one-sided). RESULTS: The number of different procedures during October 1, 2011-September 30, 2018 was more than twice that during October 1, 2004-September 30, 2011. Regarding pelvic floor surgeries related to POP, the rate of synthetic mesh procedures increased from 38.1% (5298/13,906) during October 1, 2004-September 30, 2011 to 46.0% (14,107/30,688) during October 1, 2011-September 30, 2018, whereas the rate of non-mesh procedures decreased from 61.9% (8608/13,906) to 54.0% (16,581/30,688) (Z = 15.53, P < 0.001). Regarding synthetic mesh surgeries related to POP, the rates of transvaginal placement of surgical mesh (TVM) procedures decreased from 94.1% (4983/5298) to 82.2% (11,603/14,107) (Z = 20.79, P < 0.001), but the rate of laparoscopic sacrocolpopexy (LSC) procedures increased from 5.9% (315/5298) to 17.8% (2504/14,107). CONCLUSIONS: The rate of synthetic mesh procedures increased while that of non-mesh procedures decreased significantly. The rate of TVM procedures decreased while the rate of LSC procedures increased significantly. TRIAL REGISTRATION NUMBER: NCT03620565, https://register.clinicaltrials.gov.


Assuntos
Diafragma da Pelve , Prolapso de Órgão Pélvico , China , Feminino , Procedimentos Cirúrgicos em Ginecologia/efeitos adversos , Humanos , Diafragma da Pelve/cirurgia , Prolapso de Órgão Pélvico/cirurgia , Telas Cirúrgicas/efeitos adversos , Resultado do Tratamento , Vagina
2.
J Ovarian Res ; 13(1): 31, 2020 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-32192508

RESUMO

As one of the main gynecological cancers, ovarian cancer (OC) has an unfavourable outcomes owing to its high recurrence and metastasis rate. Our previous studies have revealed that LINC01296 functions as an oncogene in OC, but the underlying mechanism has not been explored. The aim of this paper was to further investigate that how LINC01296 plays a role in OC. Through online software prediction, miR-29c-3p has been discriminated as the target miRNA of LINC01296 for further research, and subsequent luciferase assay confirmed bioinformatics prediction. Then the data obtained from the two databases (GSE119055 and GSE83693) were analyzed by GEO2R for differential gene analysis. The results indicated that the miR-29c-3p was lowly expressed in OC tissues than that in normal ovarian tissues, and its expression in recurrent OC tissues was lower than that in primary OC tissues. Simultaneously, Kaplan-Meier survival analysis illustrated that the lower expression of miR-29c-3p was interrelated to unfavourable outcomes of OC. Further, the qRT-PCR data revealed that the miR-29c-3p expression in OC cell lines (SKOV-3 and OVCAR-3) was markedly declined than that in normal control cells (IOSE80). Subsequently, the functional experiments, such as CCK8, colony formation and Transwell assays, prompted that inhibition of miR-29c-3p can obviously increase the proliferation, invasion and migration of OVCAR3 and SKOV3 cells compared with control group, while downregulation of LINC01296 showed an opposite result. It is worth noting that downregulation of LINC01296 can reverse the effect of miR-29c-3p suppression on OC cells. Finally, we detected the changes of EMT-related proteins by western blot experiment, and reached a similar conclusion that knockdown of LINC01296 reversed the EMT caused by miR-29c-3p inhibition. In sum up, the cancer-promoting function of LINC01296 was achieved by regulating the expression of miR-29c-3p, and LINC01296/miR-29c-3p axis mediates the mechanical regulation of EMT in OC cells, hoping to provide the novel biomarkers and possibilities for OC therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Interferência de RNA , RNA Longo não Codificante/genética , Regiões 3' não Traduzidas , Biomarcadores Tumorais , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/terapia
3.
Cell Signal ; 62: 109341, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31176022

RESUMO

BACKGROUND: Recently, long intergenic non-coding RNA 01296 (LINC01296) has been demonstrated to regulate the initiation and progression of several cancers, but the functions of LINC01296 in ovarian cancer still remain unclear. The objective of our study was to determine the expression, biological roles, and clinical significance of LINC01296 in ovarian cancer. METHODS: LINC01296 expression was measured in ovarian cancer tissues or cell lines. Next, the relationships between LINC01296 levels and the clinical factors of ovarian cancer, such as progression-free survival and overall survival were analyzed. Additionally, cell proliferation, migration and invasion capacities, apoptosis, cell cycle distribution were investigated after silencing of LINC01296. To confirm whether LINC01296 mediates EMT initiation in ovarian cancer cells, the effect of LINC01296 silence on E-cadherin, N-cadherin and vimentin was assessed in SKOV3 and OVCAR3 cells. RESULTS: We found that LINC01296 was over-expressed in ovarian cancer tissues and cell lines, when comparing with adjacent normal tissue samples and normal cells. Higher LINC01296 expression was significantly correlated with shorter progression-free survival and overall survival. For the functional experiments, knockdown of LINC01296 suppressed cell proliferation, inhibited colony formation ability, abrogated cell migration and invasion potential, and enhanced cell apoptosis. Cell cycle analysis suggested that LINC01296 positively regulated cell cycle progression in ovarian cancer cells. Moreover, western blotting analysis displayed that knockdown of LINC01296 significantly increased E-cadherin, but reduced N-cadherin and vimentin expressions in SKOV3 and OVCAR3 cells, compared with no-transfection cells. CONCLUSIONS: LINC01296 plays an important role in promoting the progression of ovarian cancer. Over-expression of LINC01296 might function as an indicator for diagnosis and prognosis of ovarian cancer patients.


Assuntos
Proliferação de Células/genética , Invasividade Neoplásica/genética , Neoplasias Ovarianas/genética , RNA Longo não Codificante/genética , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Progressão da Doença , Intervalo Livre de Doença , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Humanos , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/patologia , Prognóstico , RNA Longo não Codificante/antagonistas & inibidores
4.
Arch Gynecol Obstet ; 299(6): 1627-1639, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31006841

RESUMO

PURPOSE: Curcumin (Cur), a yellow-colored dietary flavor from the plant (Curcuma longa), has been demonstrated to potentially resist diverse diseases, including ovarian cancer, but drug resistance becomes a major limitation of its success clinically. The key molecule or mechanism associated with curcumin resistance in ovarian cancer still remains unclear. The aim of our study was to investigate the effects of curcumin on autophagy in ovarian cancer cells and elucidate the underlying mechanism. METHODS: In our study, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), EdU proliferation assay and colony-forming assay were used to assess cell viability. Apoptosis was detected by western blot and flow cytometric analysis of apoptosis. Autophagy was defined by both electron microscopy and immunofluorescence staining markers such as microtubule-associated protein 1 light chain 3 (LC3). Plasmid construction and shRNA transfection helped us to confirm the function of curcumin. RESULTS: Curcumin reduced cell viability and induced apoptotic cell death by MTT assay in human ovarian cancer cell lines SK-OV-3 and A2780 significantly. Electron microscopy, western blot and immunofluorescence staining proved that curcumin could induce protective autophagy. Moreover, treatment with autophagy-specific inhibitors or stable knockdown of LC3B by shRNA could markedly enhance curcumin-induced apoptosis. Finally, the cells transiently transfected with AKT1 overexpression plasmid demonstrated that autophagy had a direct relationship with the AKT/mTOR/p70S6K pathway. CONCLUSIONS: Curcumin can induce protective autophagy of human ovarian cancer cells by inhibiting the AKT/mTOR/p70S6K pathway, indicating the synergistic effects of curcumin and autophagy inhibition as a possible strategy to overcome the limits of current therapies in the eradication of epithelial ovarian cancer.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Curcumina/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linhagem Celular Tumoral , Curcumina/farmacologia , Feminino , Humanos , Transfecção
5.
Exp Ther Med ; 13(6): 3097-3102, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28587385

RESUMO

The aim of the present study was to evaluate the influence of a gonadotropin-releasing hormone (GnRH) antagonist compared with a GnRH agonist on the in vitro fertilization cycle outcome in patients with polycystic ovary syndrome. The outcomes of pregnancy were evaluated. The area under the curve (AUC) of the receiver operating characteristic (ROC) curve was also used to evaluate whether the endometrial thickness (cm) and estradiol (E2) level (pg/ml) on the day of human chorionic gonadotropin (hCG) administration (the hCG day) had the best sensitivity and specificity for predicting a clinical pregnancy. The results demonstrated that there were significant differences in the E2 and progesterone levels between the two treatment groups on the hCG day. Furthermore, the mean number of total oocytes retrieved, mean number of 2 pronuclei oocytes, mean number of oocytes cleaved (P<0.05), mean number of embryos available (P=0.022) and mean number of embryos transferred (P=0.014) were significantly different. Additionally, the rates of ectopic pregnancy (P=0.984) and ovarian hyperstimulation syndrome (P=0.976) did not differ significantly between the treatment groups. Although the biochemical pregnancy (P=0.592), clinical pregnancy (P=0.617) and live birth (P=0.365) rates were lower with the GnRH antagonist than with the GnRH agonist, there were no significant differences in the outcomes between the two groups. Analysis of the influence of endometrial thickness with respect to the clinical pregnancy using the ROC (AUC) method revealed that when the best cutoff of 9.75 cm was used, the sensitivity was 62.5%, the specificity was 43.1% and the AUC was 0.53. Additionally, the Youden index was 0.056. Analysis of the influence of the E2 level on the hCG day on clinical pregnancy, using the ROC (AUC) method showed that the best cutoff was 2,984.5 pg/ml, which had a sensitivity of 68.8% and specificity of 52.9%, while the AUC was 0.573 (with a Youden index of 0.217). Furthermore, the results demonstrated that neither the endometrial thickness nor the E2 level on the hCG day had the best sensitivity and specificity for predicting a clinical pregnancy.

6.
J Int Med Res ; 45(6): 1731-1738, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28661216

RESUMO

Objective This retrospective study compared the effect of the luteal phase ovarian stimulation protocol (LP group) with the gonadotrophin-releasing hormone (GnRH) antagonist protocol (AN group) in women with poor ovarian responses. Methods Ovarian stimulation was initiated with 225 IU of human gonadotrophin (hMG) daily. When the dominant follicle diameter exceeded 13 mm, 0.25 mg of a GnRH antagonist was used daily until human chorionic gonadotrophin (HCG) administration in the AN group. A GnRH antagonist was not used in the LP group. Ovulation was induced with HCG for all patients when at least one follicle reached a diameter of 16 mm or one dominant follicle reached 18 mm. The highest quality embryos were transferred or cryopreserved for later transfer. Results From January 2013 to December 2015, 274 women with poor ovarian response were included. A total of 108 patients underwent the luteal phase ovarian stimulation protocol while 166 patients underwent the GnRH antagonist protocol. hMG was used for more total days in the LP group was than in the AN group. Oestradiol levels on the day of HCG administration in the LP group were significantly lower than those in the AN group. The mean number of oocytes retrieved in the LP and AN groups was 3.5 ± 2.5 and 3.5 ± 2.9, respectively. The mean number of embryos of the highest quality was 1.7 ± 1.2 and 1.7 ± 1.5, respectively. The clinical pregnancy and implantation rates in the LP and AN groups were 26.2% (22/84) and 25% (29/116), and 15.5% (24/155) and 16.3% (35/215), respectively. Conclusions The luteal phase ovarian stimulation protocol can be applied in women with poor ovarian response and attain comparable clinical pregnancy and implantation rates to those of the GnRH antagonist protocol.


Assuntos
Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Fase Luteal/fisiologia , Indução da Ovulação/métodos , Adulto , Feminino , Humanos , Gravidez , Resultado da Gravidez
7.
Tumour Biol ; 39(5): 1010428317701627, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28475014

RESUMO

Receptor tyrosine kinase-like orphan receptor 2 is an enzyme-linked receptor which specifically modulates WNT5A signaling and plays an important role in tumorigenesis, invasion, and metastasis; however, the precise role of receptor tyrosine kinase-like orphan receptor 2 in cancer is controversial. The purpose of this study was to investigate the expression and role of receptor tyrosine kinase-like orphan receptor 2 in ovarian carcinoma and clarify the biological functions and interactions of receptor tyrosine kinase-like orphan receptor 2 with non-canonical Wnt pathways in ovarian cancer. The result of the human ovary tissue microarray revealed that the receptor tyrosine kinase-like orphan receptor 2-positive rate increased in malignant epithelial ovarian cancers and was extremely higher in the metastatic tumor tissues, which was also higher than that in the malignant ovarian tumor tissues. In addition, high expression of receptor tyrosine kinase-like orphan receptor 2 was closely related with ovarian cancer grading. The expression of receptor tyrosine kinase-like orphan receptor 2 protein was higher in SKOV3 and A2780 cells than OVCAR3 and 3AO cells. Knockdown of receptor tyrosine kinase-like orphan receptor 2 inhibited ovarian cancer cell proliferation, migration, invasion, and induced morphologic as well as digestive state alterations in stably transfected SKOV3 cells. Detailed study further revealed that silencing of receptor tyrosine kinase-like orphan receptor 2 reversed the epithelial-mesenchymal transition and inhibited non-canonical Wnt signaling. Our findings suggest that receptor tyrosine kinase-like orphan receptor 2 may be an important regulator of epithelial-mesenchymal transition, primarily regulated the non-canonical Wnt signaling pathway in ovarian cancer cells, and may display a promising therapeutic target for ovarian cancer.


Assuntos
Carcinogênese/genética , Transição Epitelial-Mesenquimal/genética , Neoplasias Ovarianas/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/biossíntese , Idoso , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/antagonistas & inibidores , Proteína Wnt-5a/biossíntese , Proteína Wnt-5a/genética
8.
Biomed Res Int ; 2016: 9842619, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27034956

RESUMO

Although periostin was confirmed to facilitate the pathogenesis of endometriosis by enhancing the migration, invasion, and adhesion of human endometrial stromal cells (ESCs), its effect on the endometrial epithelial cells (EECs) is still unknown. The current study aimed to determine whether periostin enhanced the epithelial-mesenchymal transition (EMT) of EECs. EECs were isolated from 12 women with endometriosis. The migration and invasion abilities of EECs were evaluated by transwell assays. Expressions of proteins were detected by western blot. After treatment with periostin, the migration and invasion abilities of EECs were enhanced. Additionally, E-cadherin and keratin were downregulated while N-cadherin and vimentin were upregulated in EECs. Simultaneously, levels of ILK, p-Akt, slug, and Zeb1 were all upregulated in EECs. After silencing the expression of ILK in EECs, levels of p-Akt, slug, Zeb1, N-cadherin, and vimentin were downregulated while E-cadherin and keratin were upregulated. Although periostin weakened the above effects in EECs after silencing the expression of ILK, it failed to induce the EMT of EECs. Thus, periostin enhanced invasion and migration abilities of EECs and facilitated the EMT of EECs through ILK-Akt signaling pathway. Playing a pivotal role in the pathogenesis of endometriosis, periostin may be a new clinical therapy target for endometriosis.


Assuntos
Moléculas de Adesão Celular/genética , Endometriose/genética , Transição Epitelial-Mesenquimal/genética , Proteína Oncogênica v-akt/genética , Proteínas Serina-Treonina Quinases/genética , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Tumores do Estroma Endometrial/genética , Tumores do Estroma Endometrial/patologia , Endometriose/metabolismo , Endometriose/patologia , Endométrio/metabolismo , Endométrio/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteína Oncogênica v-akt/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Transdução de Sinais/genética
9.
Reprod Biol Endocrinol ; 13: 103, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26377086

RESUMO

BACKGROUND: Endometrial polyps (EP) and endometriosis are both estrogen-dependent overgrowths of the endometrium. Several studies have shown a higher frequency of EP in endometriosis patients when compared with women without endometriosis. Therefore, we performed a meta-analysis to investigate the risk of EP in women with endometriosis. METHODS: This meta-analysis searched for articles published between 1964 and 2014 in PubMed, Embase, and Cochrane Library, as well as in Chinese databases, including CNKI, VIP and Wanfang, regarding the association between endometriosis and EP. Nine cohort studies and one case-control study including 2896 women were included in this meta-analysis. The EP risk was evaluated using relative risk (RR) with a 95% confidence interval (CI). Heterogeneity, small study effect and publication bias were assessed using Higgins I(2), sensitivity analysis and funnel plots, respectively. RESULTS: The risk of EP increased in women with endometriosis compared with those without endometriosis (the pooled RR, 2.81; 95% CI, 2.48-3.18). No significant heterogeneity, small study effect or publication bias was found. The risk of EP slightly increased in women with endometriosis at stages 2-4 compared with those at stage 1 (Pooled effect size: stage 2 versus stage 1, RR, 1.22, 95% CI, 1.04 - 1.42; stage 3 versus stage 1, RR, 1.23, 95% CI, 1.06-1.42; stage 4 versus stage 1, RR, 1.29, 95% CI, 1.11-1.51; stages 2-4 versus stage 1, RR, 1.24, 95% CI, 1.10-1.40); however, no significantly different risk of EP in women with endometriosis existed between the other stages. CONCLUSION: The results suggest that it is important to identify whether patients with endometriosis also have EP and then remove any coexisting EP via hysteroscopy, especially for infertile patients. This process will be clinically helpful to treat endometriosis-related infertility in patients with endometriosis, especially for those with endometriosis that is more serious than stage 1.


Assuntos
Endometriose/diagnóstico , Endométrio/patologia , Pólipos/diagnóstico , Estudos de Casos e Controles , Estudos de Coortes , Endometriose/epidemiologia , Feminino , Humanos , Pólipos/epidemiologia , Gravidez , Fatores de Risco
10.
Arch Gynecol Obstet ; 291(2): 377-82, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25216959

RESUMO

PURPOSE: Ovarian cancer is among the top diseases in the list of malignant gynaecologic tumors. In the present study, we aim to investigate the effect of lentivirus-mediated knockdown of Krüppel-like factor 9 (KLF9) on cell viability and tumor growth in ovarian cancer. METHODS: Firstly, the expression of KLF9 was determined by real-time PCR and western blot in human ovarian cancer tissues. Then, endogenous KLF9 expression was silenced by lentivirus in SKOV3 and OVCAR3 ovarian cancer cells, and followed by MTT and BrdU incorporation assays, cell cycle analysis and tumor xenografts in nude mice. RESULTS: Our results found that the expression of KLF9 is up-regulated in human ovarian cancer. As expected, KLF9 knockdown significantly inhibited cell proliferation and resulted in cell cycle arrest in the G0/G1 phase. Besides, KLF9 deficiency significantly inhibited tumor growth in nude mice. CONCLUSION: Therefore, our data reveal that lentivirus-mediated KLF9 silencing might be promising in the treatment of human ovarian cancer.


Assuntos
Fatores de Transcrição Kruppel-Like/genética , Lentivirus/genética , Neoplasias Ovarianas/patologia , Animais , Pontos de Checagem do Ciclo Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/genética , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real
11.
Asian Pac J Cancer Prev ; 15(3): 1171-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24606436

RESUMO

OBJECTIVE: The tumor suppressor gene, Ras-association domain family (RASSF)2A, is inactivated by promoter hypermethylation in many cancers. The current study was performed to evaluate the methylation status of RASSF2A in epithelial ovarian cancer (EOC) tissues and plasma, and correlations with gene expression and clinicopathologic characteristics. METHOD: We detected methylation of the RASSF2A gene in tissues and corresponding plasma samples from 47 EOC patients and 14 patients with benign ovarian tumors and 10 with normal ovarian tissues. The methylation status was determined by methylation-specific PCR while gene expression of mRNA was examined by RT-PCR. The EOC cell line, SKOV3, was treated with 5-aza-2'-deoxycytidine (5-aza- dC). RESULTS: RASSF2A mRNA expression was significantly low in EOC tissues. The frequency of aberrant methylation of RASSF2A was 51.1% in EOC tissues and 36.2% in corresponding plasma samples, whereas such hypermethylation was not detected in the benign ovarial tumors and normal ovarian samples. The expression of RASSF2A mRNA was significantly down-regulated or lost in the methylated group compared to the unmethylated group (p<0.05). After treatment with 5-aza-dC, RASSF2A mRNA expression was significantly restored in the Skov3 cell line. CONCLUSION: Epigenetic inactivation of RASSF2A through aberrant promoter methylation may play an important role in the pathogenesis of EOC. Methylation of the RASSF2A gene in plasma may be a valuable molecular marker for the early detection of EOC.


Assuntos
Metilação de DNA/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , Proteínas Supressoras de Tumor/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Decitabina , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , Proteínas Supressoras de Tumor/sangue , Proteínas Supressoras de Tumor/metabolismo
12.
Asian Pac J Cancer Prev ; 14(5): 2915-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23803053

RESUMO

BACKGROUND: Recent studies have shown that 3-deazaneplanocin A (DZNep), a well-known histone methyltransferase inhibitor, disrupts polycomb-repressive complex 2 (PRC2), and induces apoptosis, while inhibiting proliferation and metastasis, in cancer cells, including acute myeloid leukemia, breast cancer and glioblastoma. However, little is known about effects of DZNep on ovarian cancer cells. MATERIALS AND METHODS: We here therefore studied DZNep-treated A2780 ovarian cancer cells in vitro. Proliferation of ovarian cancer cells under treatment of DZNep was assessed by MTT and apoptosis by flow cytometry. Cell wound healing was applied to detect the migration. Finally, we used q-PCR to assess the migration-related gene, E-cadherin. RESULTS: DZNep could inhibit the proliferation of A2780 and induce apoptosis Furthermore, it inhibited migration and increased the expression of E-cadherin (P<0.05). CONCLUSION: DZNep is a promising therapeutic agent for ovarian cancer cells, with potential to inhibite proliferation, induce apoptosis and decrease migration.


Assuntos
Adenosina/análogos & derivados , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Adenosina/farmacologia , Apoptose/efeitos dos fármacos , Caderinas/biossíntese , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Humanos , Complexo Repressor Polycomb 2/antagonistas & inibidores
13.
Zhonghua Yi Xue Za Zhi ; 92(5): 351-3, 2012 Feb 07.
Artigo em Chinês | MEDLINE | ID: mdl-22490843

RESUMO

OBJECTIVE: To explore the expression of cyclinD1 in ovarian carcinoma cell 3AO and analyze its relationship with the proliferation of ovarian cancer cell. METHODS: Human ovarian epithelium and ovarian cancer cells 3AO were cultured in vitro. CyclinD1 genes and proteins were detected by reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry before and after the activation of cell 3AO by cis-platinum. Also the cell activity and cell cycle were observed. RESULTS: Abnormal gene amplification and over-expressions of cyclinD1 were found in ovarian cancer cell while the expression of cyclinD1 was negative in normal ovarian epithelium. Under cis-platinum, different expressions of cyclinD1 genes were found in 3AO by RT-PCR. The higher the concentrations of cisplatin, the lower expressions of cyclinD1 genes. By flow cytometry, it was also found that there were lower expressions of cyclinD1 protein in 3AO under cisplatin than without it. With the rising concentrations of cisplatin, the low expressions of cyclinD1 protein in 3AO were detected. The mean numbers were 105.9, 15.42 and 8.59, the cell apoptotic rates 0.63%, 9.08% and 27.41% and the proliferation index (PI) numbers 38.83%, 44.54%, 37.31%. The differences were statistically significant (P < 0.01). CONCLUSION: The up-regulation of cyclinD1 is detected in ovarian cancer cell. A positive correlation is found between the lower expressions of cyclinD1 and the concentration of cisplatin. There is a close relationship between the expressions of cyclinD1 and ovarian cancer cell activity.


Assuntos
Ciclina D1/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Carcinoma Epitelial do Ovário , Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/genética , Feminino , Humanos , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia
14.
Histochem Cell Biol ; 135(5): 475-85, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21465248

RESUMO

The Slit glycoproteins and their Roundabout (Robo) receptors regulate migration and growth of many types of cells including human cancer cells. However, little is known about the expression and roles of Slit/Robo in human ovarian cancer. Herein, we examined the expression of Slit/Robo in human normal and malignant ovarian tissues and its potential participation in regulating migration and proliferation of human ovarian cancer cells using two ovarian cancer cell lines, OVCAR-3 and SKOV-3. We demonstrated that Slit2/3 and Robo1 were immunolocalized primarily in stromal cells in human normal ovaries and in cancer cells in many histotypes of ovarian cancer tissues. Protein expression of Slit2/3 and Robo1/4 was also identified in OVCAR-3 and SKOV-3 cells. However, recombinant human Slit2 did not significantly affect SKOV-3 cell migration, and OVCAR-3 and SKOV-3 cell proliferation. Slit2 also did not induce ERK1/2 and AKT1 phosphorylation in OVCAR-3 and SKOV-3 cells. The current findings indicate that three major members (Slit2/3 and Robo1) of Slit/Robo family are widely expressed in the human normal and malignant ovarian tissues and in OVCAR-3 and SKOV-3 cells. However, Slit/Robo signaling may not play an important role in regulating human ovarian cancer cell proliferation and migration.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/metabolismo , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/análise , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Proteínas de Membrana/análise , Proteínas de Membrana/biossíntese , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/biossíntese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/biossíntese , Receptores Imunológicos/análise , Receptores Imunológicos/biossíntese , Transdução de Sinais , Análise Serial de Tecidos , Proteínas Roundabout
15.
Zhonghua Yi Xue Za Zhi ; 91(47): 3367-70, 2011 Dec 20.
Artigo em Chinês | MEDLINE | ID: mdl-22333207

RESUMO

OBJECTIVE: To find the time window of normalization of tumor vasculature by endostar in tumor-bearing mice with ovarian cancer. METHODS: The nude murine model of ovarian cancer was established. The vasculature markers α-SMA (alpha-smooth muscle actin) and collagen IV covered tumor vessels and hypoxic zone following the treatment of endostar were monitored. According to the changes of microvascular morphology and tumor hypoxia zone, the time window was identified. Furthermore the treatment protocols of scheduling dosing of cisplatin plus endostar at different time points were designed. After treatment tumor volume, the parameters of microvascular density (MVD) and PCNA (proliferating cell nuclear antigen) were monitored to evaluate the effects of different protocols. RESULTS: At Days 4 and 6 post-treatment, more α-SMA and collagen IV covered vessels could be observed. The amount of microvasculature expressed α-SMA on days 4 compared with control was (15.3 ± 5.2) vs (4.3 ± 2.1)/mm(2) (P < 0.01); at Day 6, the result was (16.4 ± 4.6) vs (6.6 ± 2.4)/mm(2)(P < 0.01). The expression of collagen IV had a similar change. And the numbers of microvasculature expressing collagen IV was (14.7 ± 4.3) vs (6.7 ± 5.1)/mm(2) at Days 4 and 6 (P < 0.01); (18.4 ± 5.5) vs (7.1 ± 1.7)/mm(2) (P < 0.01). The expression of HIF-1α decreased in hypoxic area. In the rhES + DDP (d4-6) group, the value of microvessel density (MVD) decreased and the expression of PCNA significantly decreased versus other groups. CONCLUSION: Endostar may normalize the tumor vasculature. And the time window is found at Days 4-6 post-treatment. During the time of vascular normalization, a combination therapy of endostar plus cisplatin has optimal efficacies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Actinas/análise , Animais , Cisplatino/administração & dosagem , Colágeno Tipo IV/análise , Endostatinas/administração & dosagem , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microvasos , Antígeno Nuclear de Célula em Proliferação/análise , Proteínas Recombinantes , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Pharmacol Res ; 56(6): 483-92, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18029193

RESUMO

Smac/DIABLO is a recently identified protein released from mitochondria in response to apoptotic stimuli and promotes apoptosis by antagonizing inhibitor of apoptosis proteins (IAPs). In this study, we observed depressed Smac/DIABLO and increased XIAP expression in ovarian epithelial tissues ordered by normal, benign and malignant epithelia. In epithelial ovarian cancer (EOC), the expression of Smac/DIABLO decreased with the malignancy. Smac/DIABLO expression showed no correlation with TRAIL sensitivity, while lower Smac/DIABLO expression and decreased release of Smac/DIABLO from mitochondria upon apoptosis stimuli were observed in paclitaxel-resistant A2780/pac cells as compared to the sensitive controls. Ectopic Smac/DIABLO alone inhibited cell growth, arrested cells in G0/G1 phase, and sensitized drug-resistant EOC cells to TRAIL or paclitaxel-induced apoptosis. Increased apoptosis was associated with the down-regulation of XIAP, FLIP, and up-regulation of Smac/DIABLO, cytochrome c, p53, along with increased activity of caspase-3. Thus, over-expression of Smac/DIABLO is a promising strategy for drug-resistant ovarian cancer treatment.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Mitocondriais/genética , Neoplasias Ovarianas/genética , Adulto , Antineoplásicos/farmacologia , Apoptose , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pessoa de Meia-Idade , Proteínas Mitocondriais/metabolismo , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo , Paclitaxel/farmacologia , RNA Mensageiro/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Transfecção , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
17.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 25(4): 438-42, 2003 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-12974091

RESUMO

OBJECTIVE: To investigate the in vitro effect of HSV-tk/GCV using a hTERT promoter-driven vector system on Skov3 ovarian cancer cells. METHODS: An expression vector (pBTdel-279-tk) containing tk gene under the hTERT promoter was constructed by molecular biological methods, and then was transfected into Skov3 ovarian cancer cells, normal ovarian epithelial cells (NOEC) and human embryonic lung fibroblast by cationic liposome. Following the transfection with tk, GCV was added, and MTT and flow cytometry methods were applied to investigate its antitumor effect. RT-PCR was used to detect the tk gene in ovarian cancer cells and normal cells after the transfection of pcDNA3-tk or pBTdel-279-tk. RESULTS: pBTdel-279-tk/GCV system induced apoptosis in hTERT-positive ovarian cancer cells, but not in hTERT-negative normal ovarian epithelial cells and fibroblasts. The hTERT promoter system was almost as efficient in inducing cancer cell death as the CMV promoter. tk gene was expressed in Skov3 cells and NOEC after pcDNA3-tk transfection, while positive was only in ovarian cancer cells after pBTdel-279-tk transfection. CONCLUSION: The telomerasespecific transfer of the tk gene under the hTERT promoter is a novel targeting approach for the treatment of ovarian cancer and may lead to an effective and specific gene therapy.


Assuntos
Apoptose/genética , Neoplasias Ovarianas/genética , Regiões Promotoras Genéticas/genética , Telomerase/genética , Timidina Quinase/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Proteínas de Ligação a DNA , Feminino , Ganciclovir/farmacologia , Terapia Genética , Humanos , Neoplasias Ovarianas/patologia , Simplexvirus/genética , Transfecção , Células Tumorais Cultivadas
18.
Ai Zheng ; 22(5): 486-91, 2003 May.
Artigo em Chinês | MEDLINE | ID: mdl-12753708

RESUMO

BACKGROUND & OBJECTIVE: Approximately 90% of tumors have telomerase activity, whereas most normal cells do not express telomerase. Telomerase catalytic subunit or human telomerase reverse transcriptase (hTERT) is the main component of telomerase. Telomerase expression is predominantly regulated at the transcriptional level of hTERT. Telomerase, hTERT, and hTERT promoter are closely related. The aim of this study was to investigate the relationship among human telomerase reverse transcriptase (hTERT) promoter activity, hTERT mRNA expression, and telomerase activity in three ovarian cancer cell lines. METHODS: hTERT promoter activity was determined by luciferase assay after the plasmids of pBTdel-279 containing hTERT core promoter were transfected into three ovarian carcinoma-derived cell lines of OVCAR3, SKOV3, 3AO and normal human ovarian epithelial cells. hTERT mRNA expression levels of these cells were determined by semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR). Telomerase activity was determined by PCR-ELISA (enzyme-linked immunosorbent assay) in above four cells. Immortalized human embryonic kidney cell line HEK293 and the human embryonic lung fibroblasts HELF were used as positive control and negative control, respectively. RESULTS: The relative luciferase activities of OVCAR3, SKOV3, 3AO and normal ovarian epithelial cells by the hTERT promoter were 31.4%, 20.3%, 17.7%, and 0.3%, respectively, as the luciferase activity of pGL3-control plasmid in each cell line was considered as 100%. The hTERT mRNA relative expression levels of above four cells were 1.30, 1.00, 0.63, and 0, respectively, as SKOV3 expression level was considered as 1. The telomerase activities were 0.580, 0.414, 0.386, and 0.103, respectively, as >0.2 was considered as positive. The hTERT promoter activity, hTERT mRNA expression level, and telomerase activity were specially raised in three ovarian cancer cell lines. The highest levels of hTERT promoter activity, hTERT mRNA expression, and telomerase activity were observed in OVCAR3 cell line, whereas negative in normal ovarian epithelial cells. hTERT promoter activity was closely associated with hTERT mRNA expression level and telomerase activity (P< 0.01,P< 0.02). CONCLUSION: hTERT promoter is specially activated in ovarian cancer cells which expresses telomerase. hTERT promoter activity is positively correlated with telomerase activity. Acting as a targeting promoter, hTERT promoter may be applied in gene therapy of ovarian cancer.


Assuntos
Neoplasias Ovarianas/enzimologia , Regiões Promotoras Genéticas/genética , RNA Neoplásico/metabolismo , Telomerase/genética , Telomerase/metabolismo , Proteínas de Ligação a DNA , Feminino , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , RNA Mensageiro/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...