Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Clin Appl Thromb Hemost ; 30: 10760296241255958, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38767088

RESUMO

Venous thromboembolism (VTE) is a common complication in patients with high-grade serous ovarian cancer (HGSOC) after surgery. This study aims to establish a comprehensive risk assessment model to better identify the potential risk of postoperative VTE in HGSOC. Clinical data from 587 HGSOC patients who underwent surgical treatment were retrospectively collected. Univariate and multivariate logistic regression analyses were performed to identify independent factors influencing the occurrence of postoperative VTE in HGSOC. A nomogram model was constructed in the training set and further validated in the verification set. Logistic regression identified age (odds ratio [OR] = 1.063, P = .002), tumor size (OR = 3.815, P < .001), postoperative transfusion (OR = 5.646, P = .001), and postoperative D-dimer (OR = 1.246, P = .003) as independent risk factors for postoperative VTE in HGSOC patients. A nomogram was constructed using these factors. The receiver operating characteristic curve showed an area under the curve (AUC) of 0.840 (95% confidence interval [CI]: 0.782, 0.898) in the training set and 0.793 (95% CI: 0.704, 0.882) in the validation set. The calibration curve demonstrated a good consistency between model predictions and actual results. The decision curve analysis indicated the model benefits at a threshold probability of less than 70%. A nomogram predicting postoperative VTE in HGSOC was established and validated. This model will assist clinicians in the early identification of high-risk patients, enabling the implementation of appropriate preventive measures.


Assuntos
Nomogramas , Complicações Pós-Operatórias , Tromboembolia Venosa , Humanos , Tromboembolia Venosa/etiologia , Tromboembolia Venosa/epidemiologia , Feminino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/etiologia , Fatores de Risco , Idoso , Estudos Retrospectivos , Neoplasias Ovarianas/cirurgia , Medição de Risco/métodos , Adulto
2.
J Clin Med ; 12(3)2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36769760

RESUMO

OBJECTIVE: We aimed to compare the survival outcomes and adverse events of hyperthermic intraperitoneal chemotherapy (HIPEC), intraperitoneal chemotherapy (IP)and intravenous chemotherapy (IP)for primary advanced ovarian cancer. METHODS: PubMed, CENTRAL (Cochrane Central Registry of Controlled Trials), Embase, Web of Science and Scopus were searched using multiple terms for primary advanced ovarian cancer, including randomized controlled trials and comparative studies in both Chinese and English (up to date 15 August 2022). Outcomes include overall survival, progression-free survival and adverse events. The data were pooled and reported as hazard ratio (HRs) with 95% confidence intervals. The Newcastle-Ottawa Scales were used to assess the risk of bias in the included comparative study. The Cochrane Collaboration's Risk of Bias Tool was used for randomized controlled trials. RESULTS: In total, 32 studies, including 6347 patients and 8 different platinum-based chemotherapy regimens, were included in this network meta-analysis. Our analysis results showed that HIPEC2 (carboplatin with area under the curve 10) exhibited a statistically significant OS benefit compared to IV, weekly dose-dense chemotherapy and HIPEC1 (cisplatin with 75/100 mg/m2). Intraperitoneal plus intravenous chemotherapy was associated with a statistically significantly better likelihood of overall survival compared to IV. For progression-free survival, our statistical results only suggest a better progression-free survival in ovarian cancer patients treated with HIPEC1 compared with weekly dose-dense chemotherapy. No evidence of difference was observed between the other comparison groups. Compared with the non-HIPEC group, HIPEC may had a higher incidence of electrolyte disturbances (≥grade 3). CONCLUSION: Our statistical analysis suggests that the groups receiving HIPEC2 had a better OS than the groups receiving IV, weekly dose-dense chemotherapy and HIPEC1. For PFS, our analysis only showed HIPEC1 is better than IV. Moreover, HIPEC may lead to a higher incidence of electrolyte disturbances (≥grade 3). HIPEC therapy for advanced ovarian cancer is currently controversial.

3.
Int J Gen Med ; 14: 8073-8085, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34803394

RESUMO

BACKGROUND: Endometrial carcinoma (EC) is one of the most common malignancies. Immunotherapy has shown promising effects in the treatment against specific subtypes of EC. METHODS: The RNA and clinical information of patients with EC were acquired from The Cancer Gene Atlas (TCGA) database. Firstly, the differentially expressed pyroptosis-related lncRNAs (PRLs) were screened between the tumor and normal control tissue. Secondly, the PRLs closely related to survival were identified by univariate and multivariate regression analysis, based on which, we evaluated the risk score for each EC patient to construct a risk signature. Moreover, we assessed the prognostic value, clinical relevance immunity, and immunotherapy based on this signature. RESULTS: We screened out 9 individual PRLs (AC087491.1, AL353622.1, AL035530.2, LINC02036, AL021578.1, AL390195.2, AC009097.2, AC004585.1, and AC244517.7) closely related to the prognosis of EC. Kaplan-Meier analyses showed a poorer prognosis for the patients in the high-risk FRLs signature (P < 0.001). The area under the curve (AUC) for 1 year, 2 years, 3 years was 0.693, 0.694, 0.750, respectively. Our risk model could be considered as an independent prognostic marker for EC (P < 0.001, HR:2.172, 95% CI:1.532-3.079). Moreover, immune functions and checkpoints were generally different in the 2 groups. Simulation analysis by termed immunophenoscores hinted that immunotherapy might bring optimal therapeutic effect in the low-risk group. CONCLUSION: We successfully developed a novel signature with 9 lncRNAs related to pyroptosis, which may be used as biomarkers to evaluate the prognosis and immune treatment of EC.

4.
Aging (Albany NY) ; 13(13): 17407-17427, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34232919

RESUMO

Ovarian cancer is the third most common cancer and the second most common cause of gynecologic cancer death in women. Its routine clinical management includes surgical resection and systemic therapy with chemotherapeutics. While the first-line systemic therapy requires the combined use of platinum-based agents and paclitaxel, many ovarian cancer patients have recurrence and eventually succumb to chemoresistance. Thus, it is imperative to develop new strategies to overcome recurrence and chemoresistance of ovarian cancer. Repurposing previously-approved drugs is a cost-effective strategy for cancer drug discovery. The antiparasitic drug mebendazole (MBZ) is one of the most promising drugs with repurposing potential. Here, we investigate whether MBZ can overcome cisplatin resistance and sensitize chemoresistant ovarian cancer cells to cisplatin. We first established and characterized two stable and robust cisplatin-resistant (CR) human ovarian cancer lines and demonstrated that MBZ markedly inhibited cell proliferation, suppressed cell wounding healing/migration, and induced apoptosis in both parental and CR cells at low micromole range. Mechanistically, MBZ was revealed to inhibit multiple cancer-related signal pathways including ELK/SRF, NFKB, MYC/MAX, and E2F/DP1 in cisplatin-resistant ovarian cancer cells. We further showed that MBZ synergized with cisplatin to suppress cell proliferation, induce cell apoptosis, and blunt tumor growth in xenograft tumor model of human cisplatin-resistant ovarian cancer cells. Collectively, our findings suggest that MBZ may be repurposed as a synergistic sensitizer of cisplatin in treating chemoresistant human ovarian cancer, which warrants further clinical studies.


Assuntos
Antinematódeos/farmacologia , Antineoplásicos/farmacologia , Carcinoma Epitelial do Ovário/tratamento farmacológico , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Mebendazol/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Cisplatino/uso terapêutico , Reposicionamento de Medicamentos , Feminino , Humanos , Camundongos , Camundongos Nus , Ensaio Tumoral de Célula-Tronco , Cicatrização , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biol Reprod ; 104(6): 1347-1359, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33693648

RESUMO

Overloaded iron can deposit in the reproductive system and impair ovarian function. But few studies have identified the exact effect of overloaded iron on the endocrine function and fertility capacity in female mice. Here, we established iron-overloaded mouse models by intraperitoneal injection of iron dextran to adult female C57BL/6J mice at 0.1 g/kg (LF group), 0.5 g/kg (MF group), and 1.0 g/kg (HF group) concentrations once a week for eight consecutive weeks. We found that overloaded iron resulted in smaller ovaries, as well as accumulated oxidative damages. The endocrine function and follicle development were also impeded in the MF and HF groups. The 10-month breeding trial indicated that (1) Low concentration of iron (0.1 g/kg) wasn't detrimental to the ovary; (2) Middle concentration of iron (0.5 g/kg) impeded the childbearing process, though it could be recovered following the iron excretion; and (3) High concentration of iron (1.0 g/kg) damaged the fertility, even gave rise to sterility. Yet for those fertile mice, litter number and litter size were smaller and the ovarian reserve of their offspring was impaired. Transcriptome profiling results indicated that overloaded iron could compromise ovarian function by disrupting ovarian steroidogenesis, interfering with ovarian microenvironment, and inhibiting Wnt signaling. Taken together, we have demonstrated the effect that chronic concentration-dependent iron overload exerted on mouse ovarian function, which may act as a preliminary basis for further mechanism and intervention investigations.


Assuntos
Sobrecarga de Ferro/metabolismo , Reserva Ovariana/efeitos dos fármacos , Ovário/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Ovário/efeitos dos fármacos
6.
Hum Cell ; 33(3): 768-779, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32166565

RESUMO

Non-catalytic region of tyrosine kinase adaptor protein 1 (Nck1) is crucial for the progression of cancers. However, little is known on the role of Nck1 in the progression of ovarian carcinoma (OC). Here, we show that Nck1 expression is up-regulated in 176 OC tissues, compared with non-carcinoma ovarian tissues, and the up-regulated Nck1 expression is associated with the aggressiveness of OC and shorter overall and disease-free survival in this population. Higher Nck1 expression was an independent risk factor for poor prognosis of OC. Furthermore, Nck1 silencing by short hairpin RNA (shRNA) technology significantly inhibited the proliferation, migration and invasion of OC cells in vitro and the growth and metastasis of implanted OC tumors in vivo. Human kinase phosphorylation array indicated that Nck1 silencing significantly reduced the relative levels of 11 kinase expression and phosphorylation in OC cells, particularly for decreased levels of p70S6 kinase (p70S6K) and protein kinase B (AKT) expression in SKOV3 cells. Actually, Nck1 silencing significantly decreased PI3K and AKT expression, and reduced AKT and p70S6K phosphorylation while Nck1 over-expression had opposite effects in OC cells. Therefore, our data indicate that Nck1 promotes the progression of OC by enhancing the PI3k/AKT/p70S6K signaling in OC. Our findings suggest that Nck1 expression may be valuable for evaluating the prognosis of OC and as a target for design of new therapies for OC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Carcinoma/genética , Proteínas Oncogênicas/fisiologia , Neoplasias Ovarianas/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/fisiologia , Carcinoma/terapia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Humanos , Terapia de Alvo Molecular , Neoplasias Ovarianas/terapia , Prognóstico , Transdução de Sinais/genética
7.
Oncol Rep ; 38(2): 1035-1042, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627610

RESUMO

Cervical cancer is the second most common gynecological malignancy. The mechanisms of the genesis and progression of cervical cancer are complicated and not thoroughly understood. DEK is reported as an oncogene in various cancers, such as acute myeloid leukemia, bladder cancer, breast cancer and hepatocellular cancer. However, its role in cervical cancer has not been well studied. In our study, we confirmed the DEK protein as an oncoprotein in cervical cancer tissues which is correlated to cervical cancer FIGO staging and tumor type. Moreover, in vitro loss of DEK inhibited cervical cancer cell proliferation, migration and invasion. We proved that silencing DEK downregulated Wnt/ß-catenin and MMP-9, and silencing DEK increased GSK-3ß activity via regulating its phosphorylation instead of translation. Silencing DEK reduced p-Ser9-GSK-3ß and increased p-Tyr216-GSK-3ß, which resulted in ß-catenin degradation. Finally, the xenograft model in nude mice proved that silencing DEK impaired cervical cancer cell tumorigenicity. This research unveiled the function of DEK in tumorigenesis and metastasis via the DEK/p-Ser9-GSK-3ß/p-Tyr216-GSK-3ß/ß-catenin axis in cervical cancer and gave insights into DEK-targeting therapy for patients suffering from cervical cancer.


Assuntos
Adenocarcinoma/prevenção & controle , Carcinoma de Células Escamosas/prevenção & controle , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Proteínas Oncogênicas/antagonistas & inibidores , Proteínas de Ligação a Poli-ADP-Ribose/antagonistas & inibidores , Neoplasias do Colo do Útero/prevenção & controle , beta Catenina/antagonistas & inibidores , Adenocarcinoma/metabolismo , Adenocarcinoma/secundário , Animais , Apoptose , Biomarcadores Tumorais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundário , Estudos de Casos e Controles , Proliferação de Células , Transformação Celular Neoplásica , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Feminino , Seguimentos , Inativação Gênica , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Metástase Linfática , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Fosforilação , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Prognóstico , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/genética , beta Catenina/metabolismo
8.
Med Sci Monit ; 23: 1317-1324, 2017 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-28298621

RESUMO

BACKGROUND The role that nc886, a non-coding microRNA, plays in human endometrial cancer is unknown. The present study aimed to describe the functional role of nc886 in human endometrial cancer-1A (HEC-1A) cell line, which may provide another target for human endometrial cancer treatment. MATERIAL AND METHODS The expression levels of nv886 in normal human endometrial tissue and the early phase and late phase of human endometrial cancer tissues were determined and compared by fluorescence in situ hybridization (FISH). Small interference RNA (siRNA) was used to inhibit nc886, and cell proliferation was evaluated with the MTT test. mRNA levels of PKR, NF-κB, vascular endothelial growth factor (VEGF), and caspase-3 were determined against glyceraldehyde 3-phosphate dehydrogenase (GAPDH between the HEC-1A control group and the silenced group (nc886 silenced with siRNA) by real-time reverse transcription polymerase chain reaction (RT-PCR). The protein levels of PKR (total and phosphorylated form), NF-κB, VEGF, and caspase-3 were determined against GAPDH by Western blotting, and cell apoptosis was determined by flow cytometry. RESULTS Our results indicated that a higher level of nc886 was expressed in the late phase of human endometrial cancer tissue, less than in the early phase but still higher than in normal human endometrial tissue. After nc886 was silenced, protein levels of p-PKR (phosphorylated PKR) and caspase-3 were increased, whereas NF-κB and VEGF were decreased. CONCLUSIONS The rate of apoptosis in the silenced group was increased and the rate of cell proliferation was slower in comparison to the control.


Assuntos
Neoplasias do Endométrio/genética , Neoplasias do Endométrio/terapia , Tumores do Estroma Endometrial/genética , Tumores do Estroma Endometrial/terapia , MicroRNAs/genética , Apoptose/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias do Endométrio/metabolismo , Tumores do Estroma Endometrial/metabolismo , Feminino , Inativação Gênica , Células HeLa , Humanos , Hibridização in Situ Fluorescente , MicroRNAs/metabolismo , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transfecção
9.
Sci Rep ; 5: 17523, 2015 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-26639992

RESUMO

Ovarian cancer is the most lethal gynecologic malignancy with an overall cure rate of merely 30%. Most patients experience recurrence within 12-24 months of cure and die of progressively chemotherapy-resistant disease. Thus, more effective anti-ovarian cancer therapies are needed. Here, we investigate the possibility of repurposing antibiotic monensin as an anti-ovarian cancer agent. We demonstrate that monensin effectively inhibits cell proliferation, migration and cell cycle progression, and induces apoptosis of human ovarian cancer cells. Monensin suppresses multiple cancer-related pathways including Elk1/SRF, AP1, NFκB and STAT, and reduces EGFR expression in ovarian cancer cells. Monensin acts synergistically with EGFR inhibitors and oxaliplatin to inhibit cell proliferation and induce apoptosis of ovarian cancer cells. Xenograft studies confirm that monensin effectively inhibits tumor growth by suppressing cell proliferation through targeting EGFR signaling. Our results suggest monensin may be repurposed as an anti-ovarian cancer agent although further preclinical and clinical studies are needed.


Assuntos
Antibacterianos/farmacologia , Receptores ErbB/antagonistas & inibidores , Monensin/farmacologia , Compostos Organoplatínicos/farmacologia , Neoplasias Ovarianas/patologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Feminino , Células HEK293 , Humanos , Oxaliplatina , Inibidores de Proteínas Quinases/farmacologia , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Hum Pathol ; 46(9): 1331-40, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26193797

RESUMO

Abelson interactor protein 1 (Abi1) is a key regulator of actin reorganization and lamellipodia formation. Because of its role in cell migration, Abi1 has been implicated in tumor progression. In the present study, we investigated the role of Abi1 in epithelial ovarian cancer (EOC) by analyzing its expression and correlation with clinicopathological and survival data. We evaluated the expression of Abi1 in 223 paraffin-embedded EOC specimens by immunohistochemistry and 46 frozen EOC samples by Western blot and real-time reverse transcription polymerase chain reaction analysis. Results showed that Abi1 protein and mRNA expression was significantly higher in EOC tissue compared with noncancerous tumors and normal ovaries (P < .05). Moreover, high level of Abi1 expression was significantly correlated with advanced stage, high grade, elevated Ca-125 level, and suboptimal surgical debulking (P < .05). By Western blot analysis, Abi1 was expressed in highly invasive cells compared with weakly invasive cells (P < .05). Immunofluorescence was performed to demonstrate Abi1 expression in SKOV3 cells. Additionally, upregulation of Abi1 significantly correlated with shorter survival (P < .05). Most importantly, multivariate analysis showed that Abi1 overexpression is an independent prognostic factor, complementary to clinical stage and residual tumor size. In conclusion, our findings suggest that Abi1 acts as a tumor-promoting gene in EOC progression, which may lead to unfavorable prognosis. Abi1 may serve as a potential effective prognostic marker for EOC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Proteínas do Citoesqueleto/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Western Blotting , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/genética , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Análise Multivariada , Gradação de Tumores , Estadiamento de Neoplasias , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/mortalidade , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Modelos de Riscos Proporcionais , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Risco , Carga Tumoral , Regulação para Cima , Adulto Jovem
11.
Med Oncol ; 32(2): 329, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25585684

RESUMO

Ras-related C3 botulinum toxin substrate 1 (rac1) has been implicated in tumor epithelial-mesenchymal transition (EMT); however, limited information is available regarding the role of rac1 in epithelial ovarian cancer (EOC). This study aimed to evaluate the correlation of rac1 expression with EMT and EOC prognosis. Rac1 protein levels of 150 EOC specimens were evaluated by immunohistochemical staining. Survival analysis was performed to determine the correlation between rac1 expression and survival. Cellular and molecular changes were also examined after rac1 in ovarian cancer cells was silenced in vitro and in vivo. The mechanism of rac1 on EMT was investigated by Western blot analysis. Rac1 was highly expressed in EOC. Rac1 overexpression was closely associated with advanced stage based on International Federation of Gynecology and Obstetrics, poor grade, serum Ca-125, and residual tumor size. Survival analyses demonstrated that patients with high rac1 expression levels were more susceptible to early tumor recurrence with very poor prognosis. This study revealed that rac1 downregulation decreased cell EMT and proliferation capability in vitro and in vivo. Rac1 expression possibly altered cell EMT by interacting with p21-activated kinase 1 and p38 mitogen-activated protein kinase signaling pathways. The present study showed that rac1 overexpression is associated with cell EMT and poor EOC prognosis. Rac1 possibly plays an important role in predicting EOC metastasis.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Proteínas rac1 de Ligação ao GTP/biossíntese , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Carcinoma Epitelial do Ovário , Feminino , Xenoenxertos , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/mortalidade , Neoplasias Ovarianas/mortalidade , Prognóstico , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adulto Jovem
12.
Tumour Biol ; 36(3): 1429-35, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25604140

RESUMO

The significance of steroidogenic factor-1 (SF-1) in human ovarian tumor has not been fully investigated. The purposes of this study are to provide a meta-analysis for SF-1 and to determine whether SF-1 is associated with ovarian tumor progression and clinicopathological characteristics. A detailed literature search was made for related research publications written in English. Methodological quality of the studies was also evaluated. The data were extracted and assessed by two reviewers independently. Analysis of pooled data was performed, and odds ratio (OR) and corresponding confidence intervals (CIs) were calculated and summarized respectively. Final analysis from seven eligible studies was performed. Aberrant SF-1 expression was significantly lower in ovarian cancer compared to that of normal ovarian tissue (OR = 0.02, 95% CI = 0.00-0.16, p = 0.0002). However, SF-1 protein expression was not significantly different between benign and malignant ovarian tumors (p = 0.35). Interestingly, aberrant SF-1 expression was significantly higher in ovarian sex cord stromal tumors than that of ovarian cancer (OR = 0.00, 95% CI = 0.00-0.01, p < 0.00001). The results of this meta-analysis suggest that SF-1 may play an important role in ovarian cancer initiation and progression. Moreover, SF-1 expression may serve as a marker in the differential diagnosis between ovarian sex cord stromal tumors and ovarian cancer.


Assuntos
Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Tumores do Estroma Gonadal e dos Cordões Sexuais/metabolismo , Tumores do Estroma Gonadal e dos Cordões Sexuais/patologia , Fator Esteroidogênico 1/biossíntese , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Estudos de Casos e Controles , Feminino , Humanos , Neoplasias Ovarianas/genética , Tumores do Estroma Gonadal e dos Cordões Sexuais/genética , Fator Esteroidogênico 1/genética
13.
Tumour Biol ; 36(3): 1487-91, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25596080

RESUMO

Previous studies demonstrated that the loss of function of the p16INK4A gene is mainly caused by the hypermethylation of p16 gene promoter; however, whether or not it is associated with the incidence of endometrial carcinoma (EC) remains unclear. In the current study, we conducted a meta-analysis to investigate the effects of p16 gene promoter hypermethylation on the incidence of EC. Detailed research publications were searched from Embase, PubMed, and ISI Web of Knowledge for composition in English or Chinese. The pooled data were collected and analyzed by Review Manager 5.2. Odds ratios (ORs) were calculated and summarized respectively. Six eligible studies, including 261 patients were selected and analyzed. The pooled OR was 0.42, test for overall effect, Z = 10.19, P < 0.0001, indicating that p16 gene promoter hypermethylation was significantly correlated with the EC patients. The results of our study strongly suggest that p16 gene promoter hypermethylation is correlated with an increased risk of EC. P16 gene promoter hypermethylation plays a critical role in endometrial carcinogenesis.


Assuntos
Metilação de DNA , Neoplasias do Endométrio/genética , Genes p16 , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Regiões Promotoras Genéticas
14.
J Ultrasound Med ; 33(11): 1903-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25336476

RESUMO

OBJECTIVES: Focused ultrasound waves penetrate superficial tissues and are aimed toward the target tissues at specific depths to exert their biological effects. Focused ultrasound has been applied for a number of clinical indications, including vulval dystrophies and low-grade vulval disease. This study aimed to assess the efficacy and safety of focused ultrasound treatment of high-grade vulval intraepithelial neoplasia (VIN). METHODS: Eighteen women with high-grade VIN were recruited and treated with focused ultrasound. During each posttreatment follow-up, the safety of, side effects of, and clinical responses to focused ultrasound were evaluated by a standardized protocol, including symptoms, clinical appearance, and histologic findings. RESULTS: All patients completed the designed follow-ups. In most cases, superficial mild to moderate swelling and blisters were seen in the focused ultrasound-treated skin but not in adjacent normal skin. Of the 18 patients, 16 showed complete histologic regression and resolution of symptoms 6 months after treatment. Of the other 2 patients, 1 showed complete regression after a second focused ultrasound treatment. The other patient did not respond to the focused ultrasound treatment and underwent a partial vulvectomy 6 months after treatment. None of the patients developed invasive carcinoma of the vulva during the follow-up period. One patient had local pruritus that was not alleviated by anti-inflammatory medication and local care. CONCLUSIONS: The complete responses observed in women with high-grade VIN treated by focused ultrasound, together with the preservation of adjacent normal tissue, suggest that focused ultrasound may be considered for treatment of high-grade VIN.


Assuntos
Ablação por Ultrassom Focalizado de Alta Intensidade/métodos , Lesões Pré-Cancerosas/patologia , Lesões Pré-Cancerosas/terapia , Dermatopatias/patologia , Dermatopatias/terapia , Doenças da Vulva/patologia , Doenças da Vulva/terapia , Adulto , Idoso , Carcinoma in Situ , Feminino , Humanos , Pessoa de Meia-Idade , Resultado do Tratamento
15.
FEBS Lett ; 588(12): 2078-86, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24815691

RESUMO

Oncogenic activation of VEGF is found in various malignancies, including ovarian cancer. In this study, we investigate the role of microRNA (miRNA) in the regulation of VEGF in ovarian cancer. We find that miR-718 is expressed at low levels and inversely correlates with VEGF expression in ovarian cancer specimens. MiR-718 also directly targets and represses VEGF expression. In addition, miR-718 restoration inhibits ovarian cancer proliferation both in vitro and in vivo. Moreover, VEGF expression could reverse the effect of miR-718 on ovarian cancer by increasing the levels of phosphorylated AKT. These results suggest a new therapeutic strategy in ovarian cancer by restoring miR-718 expression, which is involved in VEGF regulation.


Assuntos
Progressão da Doença , MicroRNAs/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fator A de Crescimento do Endotélio Vascular/deficiência , Regiões 3' não Traduzidas/genética , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , RNA Interferente Pequeno/genética , Fator A de Crescimento do Endotélio Vascular/genética
16.
Int J Mol Med ; 33(5): 1289-97, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24626956

RESUMO

Cervical cancer is the principal cause of mortality due to cancer in women worldwide. New predictive markers may increase survival rates by improving the treatment of patients at a high risk for cancer. This study was carried out to investigate the amplification of human telomerase RNA component (hTERC) or/and c-MYC in cervical epithelial exfoliated cells for cervical carcinoma screening. We collected 171 specimens. including speciments from normal cervix, benign lesions, cervical intraepithelial neoplasia (CIN)1, CIN2 and CIN3, or carcinoma in situ, as well as invasive cervical squamous cell carcinoma. Fluorescence in situ hybridization (FISH) was performed to detect alterations in hTERC and c-MYC expression. We analyzed the area under the receiver operating characteristic (ROC) curve (AUC), as well as the sensitivity and specificity of single screening and conjoined screening. There was a trend toward an increasing amplification of 2 genes with the increasing severity of cervical lesions. ROC curve analysis demonstrated that the AUC values of the hTERC gene for the screening of different cervical lesions were >0.8. Compared with the hTERC gene, the AUC of the c-MYC gene for the screening of ≥CIN3 was >0.8 and the AUC for the screening of other cervical lesions was >0.7. For the screening of cervical lesions above the grade of benign lesions, cytological diagnosis was superior to the gene detection with significant differences. For the screening of cervical lesions >CIN1, there were no statistically significant differences (P>0.05) between the hTERC gene and cytological diagnosis, whereas the screening results of c-MYC detection and cytological diagnosis differed significantly (P<0.05). For the screening of cervical lesions >CIN2 or >CIN3, the detection of hTERC and c-MYC genes and cytological diagnosis had similar screening results with no statistically significant differences (P>0.05). In conclusion, using FISH to detect the amplification of hTERC or/and c-MYC on cervical epithelial exfoliated cells may be a useful and specific screening method for precancerous lesions.


Assuntos
Genes myc/genética , RNA/genética , Telomerase/genética , Neoplasias do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/genética , Detecção Precoce de Câncer , Feminino , Humanos , Hibridização in Situ Fluorescente , Técnicas In Vitro
17.
Hum Pathol ; 45(2): 401-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24439227

RESUMO

Neural precursor cell-expressed, developmentally down-regulated 9 (NEDD9), a scaffolding protein, has been identified as a prometastatic and poor prognostic gene in multiple malignant tumors. However, the potential role of the NEDD9 protein in epithelial ovarian cancer (EOC) remains unclear. In the present study, we investigated the expression of NEDD9 and the correlation between NEDD9 expression and prognosis in EOC. NEDD9 expression was detected in 129 archived EOC specimens by immunohistochemical staining and in 28 freshly frozen EOC specimens by Western blotting. The expression of NEDD9 was evaluated in ovarian cancer cell lines by Western blotting and immunofluorescence. The association between the expression of NEDD9 and prognosis was determined by survival analysis. Results suggested that NEDD9 was overexpressed in EOC specimens compared with noninvasive epithelial ovarian tumors and normal ovarian specimens. A high level of NEDD9 expression significantly correlated with advanced-stage tumors (International Federation of Gynecology and Obstetrics classes III-IV, P < .001), high-grade carcinoma (grades 2-3, P < .001), and suboptimal primary cytoreductive surgery (residual disease <1cm, P = .021). The expression level of NEDD9 varied in ovarian cancer cell lines. Multivariate analysis indicated that NEDD9 overexpression (P = .033), advanced stage (P < .001), and high-grade carcinoma (P = .01) were independent predictors of poor survival. In conclusion, NEDD9 is overexpressed and associated with an unfavorable prognosis in EOC. NEDD9 overexpression is an independent factor of poor prognosis and may serve as a potential biomarker in EOC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Fosfoproteínas/biossíntese , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Epitelial do Ovário , Progressão da Doença , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/mortalidade , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Prognóstico , Análise de Sobrevida
18.
J Minim Invasive Gynecol ; 21(2): 279-84, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24075837

RESUMO

STUDY OBJECTIVE: To assess the quality of life (QoL) of women at 1 and 12 months after ultrasound-guided high-intensity focused ultrasound (USgHIFU) treatment as compared with laparoscopic myomectomy for treatment of symptomatic uterine myomas. DESIGN: Nonrandomized prospective clinical trial (Canadian Task Force classification II-2). SETTING: Urban university-based hospital in China. INTERVENTIONS: One hundred thirty premenopausal women underwent USgHIFU (n = 89) or laparoscopic myomectomy (n = 41) for treatment of symptomatic uterine myomas. MEASUREMENTS AND MAIN RESULTS: Eighty-three patients in the HIFU group and 39 in the surgical group were followed up at 1 and 12 months. QoL was assessed using the Medical Outcomes Study 36-Item Short-Form General Health Survey, which showed no significant differences between groups in any of the 8 subscales at the 12-month follow-up visit. Symptom score, willingness to recommend the treatment to a friend, hospital stay, and recovery period were compared between the 2 groups. In the HIFU group, hospital stay was shorter (mean [SD] 2.9 [1.5] days vs 6.2 [2.7] days; p <.001) and patients resumed normal activities sooner (4.5 [1.5] days vs 10.9 [3.8] days; p <.001). Significant clinical complications and adverse events after each treatment were documented and compared, and HIFU yielded significantly better results. CONCLUSIONS: Compared with laparoscopic myomectomy, HIFU treatment of symptomatic uterine myomas leads to comparable QoL and symptom improvement, fewer significant clinical complications and adverse events, shorter hospital stay, and faster recovery. Randomized studies with long-term follow-up are needed to reach definitive conclusions insofar as HIFU treatment of uterine myomas.


Assuntos
Ablação por Ultrassom Focalizado de Alta Intensidade/métodos , Laparoscopia/métodos , Leiomioma/cirurgia , Qualidade de Vida , Neoplasias Uterinas/cirurgia , Adulto , China , Feminino , Hospitais Universitários , Humanos , Estudos Prospectivos , Inquéritos e Questionários , Resultado do Tratamento , Miomectomia Uterina/métodos , Neoplasias Uterinas/psicologia
19.
Gynecol Oncol ; 131(1): 69-76, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23820113

RESUMO

OBJECTIVES: Abelson tyrosine kinase (c-Abl) has been shown to promote solid tumor invasion and metastasis. However, little is known regarding whether c-Abl contributes to the development or progression of epithelial ovarian cancer (EOC). The aims of this study are to determine the expression of c-Abl and investigate a possible relationship between c-Abl and prognosis in EOC. METHODS: c-Abl protein level was evaluated in 137 EOC specimens by immunohistochemical staining and 32 EOC specimens by Western blot analysis. Expression of c-Abl in ovarian cancer cell lines was measured by Western blot analysis and immunofluorescence. Survival analysis was performed to assess the correlation between c-Abl expression and survival. RESULTS: Immunohistochemical staining and Western blot analysis revealed that c-Abl was overexpressed in EOC compared with samples from a non-invasive ovarian tumor and normal ovaries (P<0.05). Furthermore, expression of c-Abl was significantly associated with advanced FIGO stage, poor grade, serum Ca-125 and residual tumor size (P<0.05). By Western blot analysis, c-Abl expression was examined in four ovarian cancer cell lines. Meanwhile, immunofluorescence was performed to show c-Abl expression in SKOV3 and 3AO cell lines. Survival analysis demonstrated that patients with low c-Abl staining had a significantly better survival compared to patients with high c-Abl staining (P<0.05). In multivariate analysis, c-Abl overexpression, poor grade, advanced stage and suboptimal surgical debulking were independent prognostic factors of poor survival. CONCLUSIONS: Our present study finds that c-Abl overexpression is associated with an unfavorable outcome. c-Abl may be a crucial predictor for EOC metastasis.


Assuntos
Neoplasias Epiteliais e Glandulares/química , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/química , Neoplasias Ovarianas/patologia , Proteínas Proto-Oncogênicas c-abl/análise , Adulto , Idoso , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Feminino , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Ovário/química , Prognóstico , Modelos de Riscos Proporcionais
20.
Gynecol Oncol ; 130(2): 354-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23680521

RESUMO

OBJECTIVE: Wiskott-Aldrich syndrome protein family verprolin-homologous protein 1 (WAVE1) has been implicated in cancer cell migration and invasion. We have previously shown that the overexpression of WAVE1 in epithelial ovarian cancer (EOC) tissues is associated with a poor prognosis. However, the mechanism of WAVE1 regulating the malignant behaviors in EOC remains unclear. METHODS: In the present study, we knocked down WAVE1 expression in SKOV3 and OVCAR-3 cells through RNA interference to detect the cell biology and molecular biology changes. Moreover, western-blot was used to investigate the underlying mechanism of WAVE1 regulating the proliferative and invasive malignant behaviors in ovarian cancer cells. RESULTS: The down-regulation of WAVE1 had a significant effect on cell morphological changes. WAVE1 silencing decreased cell migration, cell invasion, cell adhesion, colony formation and cell proliferation in vitro. In addition, we found that down-regulation of WAVE1 inhibited malignant behaviors in vivo. Furthermore, our study also indicated that the PI3K/AKT and p38MAPK signaling pathways might contribute to WAVE1 promotion of ovarian cancer cell proliferation, migration, and invasion. CONCLUSIONS: WAVE1 might promote the proliferative and invasive malignant behaviors through the activation of the PI3K/AKT and p38MAPK signaling pathways in EOC.


Assuntos
Movimento Celular , Proliferação de Células , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Família de Proteínas da Síndrome de Wiskott-Aldrich/fisiologia , Animais , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Feminino , Inativação Gênica , Humanos , Camundongos , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Interferência de RNA , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA