Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Mol Med ; 28(1): 62, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35658829

RESUMEN

BACKGROUND: Ovarian cancer is one of the important factors that seriously threaten women's health and its morbidity and mortality ranks eighth among female cancers in the world. It is critical to identify potential and promising biomarkers for prognostic evaluation and molecular therapy of OV. Ubiquitin-conjugating enzyme E2S (UBE2S), a potential oncogene, regulates the malignant progression of various tumors; however, its role in OV is still unclear. METHODS: The expression and prognostic significance of UBE2S at the pan-cancer level were investigated through high-throughput gene expression analysis and clinical prognostic data from TCGA, GEPIA, and GEO databases. 181 patients with OV were included in this study. Cell culture and cell transfection were performed on OV cell lines (SKOV3 and A2780) and a normal ovarian cell line (IOSE80). The expression level and prognostic significance of UBE2S in OV were verified by western blot, immunohistochemistry, and Kaplan-Meier survival analysis. Through cell transfection, CCK-8, Ki-67 immunofluorescence, wound healing, Transwell, clonogenic, and flow cytometry assays, the effect and detailed mechanism of UBE2S knockdown on the malignant biological behavior of OV cells were explored. RESULTS: UBE2S exhibited abnormally high expression at the pan-cancer level. The results of RT-qPCR and Western blotting indicated that UBE2S was significantly overexpressed in ovarian cancer cell lines compared with normal cell lines (P < 0.05). Kaplan-Meier survival analysis and Immunohistochemistry indicated that overexpression of UBE2S was related to poor prognosis of OV (HR > 1, P < 0.05). Results of in vitro experiments indicated that UBE2S gene knockdown might inhibit the proliferation, invasion, and prognosis of OV cells by inhibiting the PI3K/AKT/mTOR signaling pathway, thereby blocking the cell cycle and promoting apoptosis (P < 0.05). CONCLUSION: UBE2S is a potential oncogene strongly associated with a poor prognosis of OV patients. Knockdown of UBE2S could block the cell cycle and promote apoptosis by inhibiting the PI3K/AKT/mTOR pathway and ultimately inhibit the proliferation, migration and prognosis of ovarian cancer, which suggested that UBE2S might be used for molecular therapy and prognostic evaluation of ovarian cancer.


Asunto(s)
Neoplasias Ováricas , Proteínas Proto-Oncogénicas c-akt , Apoptosis , Carcinoma Epitelial de Ovario/genética , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Femenino , Humanos , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
2.
Cancer Cell Int ; 20: 477, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33024415

RESUMEN

BACKGROUND: Exploring novel and sensitive targets is urgent due to the high morbidity of endometrial cancer (EC). The purpose of our study was to explore the transcription factors and immune-related genes in EC and further identify immune-based lncRNA signature as biomarker for predicting survival prognosis. METHODS: Transcription factors, aberrantly expressed immune-related genes and immune-related lncRNAs were explored through bioinformatics analysis. Cox regression and the least absolute shrinkage and selection operator (LASSO) analysis were conducted to identify the immune and overall survival (OS) related lncRNAs. The accuracy of model was evaluated by Kaplan-Meier method and receiver operating characteristic (ROC) analysis, and the independent prognostic indicator was identified with Cox analysis. Quantitative real-time polymerase chain reaction (qRT-PCR) were conducted to detect the accuracy of our results. RESULTS: A network of 29 transcription factors and 17 immune-related genes was constructed. Furthermore, four immune-prognosis-related lncRNAs were screened out. Kaplan-Meier survival analysis and time-dependent ROC analysis revealed a satisfactory predictive potential of the 4-lncRNA model. Consistency was achieved among the results from the training set, testing set and entire cohort. The distributed patterns between the high- and low-risk groups could be distinguished in principal component analysis. Comparisons of the risk score and clinical factors confirmed the four-lncRNA-based signature as an independent prognostic indicator. Last, the reliability of the results was verified by qRT-PCR in 29 cases of endometrial carcinoma and in cells. CONCLUSIONS: Overall, our study constructed a network of transcription factors and immune-related genes and explored a four immune-related lncRNA signature that could serve as a novel potential biomarker of EC.

3.
Zhongguo Zhong Yao Za Zhi ; 45(23): 5762-5769, 2020 Dec.
Artículo en Zh | MEDLINE | ID: mdl-33496117

RESUMEN

This paper discussed the synergistic anti-tumor effect of Shuangdan Capsules combined with 5-fluorouracil(5-FU) on human liver cancer cell line Huh-7 and tumor bearing mice. The effects of Shuangdan Capsules combined with 5-FU on the activity and vascular endothelial growth factor(VEGF) receptor protein expression of Huh-7 cells were investigated, and the effects of drug combination on tube formation of HUVEC cell were also verified. In addition, the mice model of Huh-7 was established to observe the anti-tumor effect of drug combination and the distribution of tumor blood flow in tumor bearing mice by using molecular imaging. HPLC analysis showed that Shuangdan Capsules mainly consisted of danshensusodium, protocatechuic aldehyde, paeoniflorin, rosmarinic acid, alkannic acid, salvianolic acid B, and paeonol. In MTT experiment, the inhibition rate of Shuangdan Capsules(20 mg·L~(-1)) and 5-FU(1 µmol·L~(-1)) on Huh-7 cells was 60%, and the CI value was 0.59, suggesting that these two drugs had synergistic anti-hepatoma cells effect. The expression of VEGF receptor in Huh-7 cells was inhibited by the combination of these two drugs. In addition, the process of HUVEC was slow, and the number, length and area of the lumen branches decreased significantly. In vivo, Shuangdan Capsules combined with 5-FU inhibited the growth and prolongation of survival of Huh-7 cells in subcutaneous transplanted tumor nude mice; serum expression of CD31 and VEGF in nude mice were decreased, while caspase-3 was increased. Meanwhile, the drug combination significantly inhibited the expressions of MMP2 and VEGF in tumor tissues. Ultrasound showed that Shuangdan Capsules combined with 5-FU also inhibited tumor angiogenesis and reduced blood flow of tumor tissue. The results showed that Shuangdan Capsules combined with 5-FU may inhibit tumor angiogenesis by inhibiting VEGF and MMP2 expressions, thereby blocking tumor growth.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Cápsulas , Línea Celular Tumoral , Proliferación Celular , Medicamentos Herbarios Chinos , Fluorouracilo , Xenoinjertos , Ratones , Ratones Desnudos , Factor A de Crecimiento Endotelial Vascular , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Cell Int ; 19: 235, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31516392

RESUMEN

BACKGROUND: Hyperoside (Hy) is a plant-derived quercetin 3-d-galactoside that exhibits inhibitory activities on various tumor types. The objective of the current study was to explore Hy effects on cervical cancer cell proliferation, and to perform a transcriptome analysis of differentially expressed genes. METHODS: Cervical cancer HeLa and C-33A cells were cultured and the effect of Hy treatment was determined using the Cell Counting Kit-8 (CCK-8) assay. After calculating the IC50 of Hy in HeLa and C-33A cells, the more sensitive to Hy treatment cell type was selected for RNA-Seq. Differentially expressed genes (DEGs) were identified by comparing gene expression between the Hy and control groups. Candidate genes were determined through DEG analysis, protein interaction network (PPI) construction, PPI module analysis, transcription factor (TF) prediction, TF-target network construction, and survival analysis. Finally, the key candidate genes were verified by RT-qPCR and western blot. RESULTS: Hy inhibited HeLa and C33A cell proliferation in a dose- and time-dependent manner, as determined by the CCK-8 assay. Treatment of C-33A cells with 2 mM Hy was selected for the subsequent experiments. Compared with the control group, 754 upregulated and 509 downregulated genes were identified after RNA-Seq. After functional enrichment, 74 gene ontology biological processes and 43 Kyoto Encyclopedia of Genes and Genomes pathways were obtained. According to the protein interaction network (PPI), PPI module analysis, TF-target network construction, and survival analysis, the key genes MYC, CNKN1A, PAX2, TFRC, ACOX2, UNC5B, APBA1, PRKACA, PEAR1, COL12A1, CACNA1G, PEAR1, and CCNA2 were detected. RT-qPCR was performed on the key genes, and Western blot was used to verify C-MYC and TFRC. C-MYC and TFRC expressions were lower and higher than the corresponding values in the control group, respectively, in accordance with the results from the RNA-Seq analysis. CONCLUSION: Hy inhibited HeLa and C-33A cell proliferation through C-MYC gene expression reduction in C-33A cells and TFRC regulation. The results of the current study provide a theoretical basis for Hy treatment of cervical cancer.

5.
Cell Physiol Biochem ; 44(4): 1325-1336, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29183027

RESUMEN

BACKGROUND/AIMS: Ovarian cancer (OC) causes more death and serious conditions than any other female reproductive cancers, and many expression signatures have been identified for OC prognoses. However, no significant overlap is found among signatures from different studies, indicating the necessity of signature identifications at the functional level. METHODS: We performed an integrated analyses of miRNA and gene expressions to identify OC prognostic subpathways (pathway regions). Using The Cancer Genome Atlas data set, we identified core prognostic subpathways, and calculated subpathway risk scores using both miRNA and gene components. Finally, we performed global risk impact analyses to optimize core subpathways using the random walk algorithm. RESULTS: Subpathway-level analyses displayed more robust results than the gene- and miRNA-level analyses. Moreover, we verified the advantage of core subpathways over the entire pathway-based results and their prognostic performance in two independent validation data sets. Based on the global impact score, 13 subpathway signatures were selected and a combined subpathway-based risk score was further calculated for OC patient prognoses. CONCLUSIONS: Overall, it was possible to systematically perform integrated analyses of the expression levels of miRNAs and genes to identify prognostic subpathways and infer subpathway risk scores for use in OC clinical applications.


Asunto(s)
MicroARNs/metabolismo , Neoplasias Ováricas/patología , ARN Mensajero/metabolismo , Algoritmos , Bases de Datos Genéticas , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Ováricas/genética , Neoplasias Ováricas/mortalidad , Pronóstico , Modelos de Riesgos Proporcionales
6.
Int J Gynecol Cancer ; 26(1): 114-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26512788

RESUMEN

OBJECTIVES: Seven in absentia homolog 2 (Siah2) is an E3 ubiquitin ligase that is expressed in mammals and is homologous to seven in absentia in Drosophila. Siah2 is involved in the progression of many malignancies. However, the role of Siah2 in ovarian cancer remains unclear. This study aims to evaluate the prognostic value of Siah2 expression for epithelial ovarian carcinoma (EOC) patients. MATERIALS AND METHODS: Immunohistochemical analysis was conducted using 32 normal ovarian specimens and 122 ovarian carcinoma specimens, respectively. We analyzed the correlations of Siah2 expression with the clinicopathological factors and prognosis of ovarian cancer patients. χ Analysis, Kaplan-Meier method, and multivariate Cox proportional hazard analysis were conducted for statistical analyses. RESULTS: Immunohistochemical staining demonstrated that the expression of Siah2 was higher in the EOC tissues than in the normal tissues. High Siah2 expression positively correlated with histological grade and lymph node metastasis but not with age, histologic type, International Federation of Gynecology and Obstetrics staging, and CA125. Patients with positive Siah2 expression showed lower overall survival and disease-free survival rates than those with negative Siah2 expression (P < 0.05 for both). Multivariate Cox analysis indicated that Siah2 was an independent parameter for overall survival (hazards ratio, 2.166; 95% confidence interval, 1.182-3.970; P = 0.012) and disease-free survival (hazards ratio, 1.819; 95% confidence interval, 1.030-3.216; P = 0.039). CONCLUSIONS: Siah2 is possibly involved in tumor development and progression in EOC. Thus, Siah2 is a promising biomarker for predicting the prognosis of ovarian cancer patients and may serve as a novel target for treating ovarian carcinoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Ubiquitina-Proteína Ligasas/metabolismo , Adulto , Anciano , Carcinoma Epitelial de Ovario , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Histerectomía , Técnicas para Inmunoenzimas , Escisión del Ganglio Linfático , Metástasis Linfática , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/cirugía , Neoplasias Ováricas/cirugía , Ovariectomía , Pronóstico , Salpingostomía , Tasa de Supervivencia
7.
Cell Physiol Biochem ; 37(3): 890-900, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26383850

RESUMEN

BACKGROUND/AIMS: LAPTM4B (lysosome-associated protein transmembrane 4 beta) is a novel oncogene with important functions in aggressive human carcinomas, including cervical cancer. However, the specific functions and internal molecular mechanisms associated with this gene in the context of cervical cancer remain unclear. METHODS: In this study, we explored the effects and mechanisms of LAPTM4B on tumor growth, metastasis and angiogenesis in vitro by depletion of LAPTM4B in Hela cell. RNA interference was used to induce down regulation of LAPTM4B gene expression in Hela cells. The motility, migration potential, and proliferation of the Hela cells were measured by flow cytometry, Transwell migration assays, wound healing assays, and Cell Counting Kit-8 assays. In addition, the cell cycle analysis utilized fluorescence-activated cell sorting. RESULTS: In this study, RNAi-mediated LAPTM4B knockdown inhibited cell growth and angiogenesis. In vitro, HeLa cells with down regulated LAPTM4B also exhibited decreased migration and invasion activity as well as significantly reduced CDK12, HIF-1α, MMP-2, MMP-9 and VEGF expression. LAPTM4B blockade significantly decreased cord lengths and branch points in a tube formation assay. CONCLUSIONS: These results suggested that LAPTM4B inactivation could be a novel therapeutic target for cervical cancer.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Técnicas In Vitro , Interferencia de ARN , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Tumour Biol ; 36(6): 4479-85, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25934333

RESUMEN

This study aimed to explore the clinical significance of AAA+ (ATPases associated with various cellular activities) nuclear coregulator cancer-associated (ANCCA) protein expression in endometrial carcinoma (EC). Correlations of ANCCA expression with clinicopathological factors and prognosis of EC patients were analyzed. Expression of ANCCA was detected in EC from 207 patients along with corresponding normal endometrium specimens by immunohistochemistry. ANCCA immunoreactivity was overexpressed in EC cases compared with that in normal endometrium (P < 0.001). High ANCCA expression was positively correlated with the International Federation of Gynecology and Obstetrics (FIGO) stage, histological grade, depth of myometrial invasion, lymph node metastasis, lymph vascular space involvement, and recurrence but not with age and histological type. Patients with high ANCCA expression exhibited significantly poorer overall survival (OS) and disease-free survival (DFS) than patients with low ANCCA expression (P = 0.001 and 0.002, respectively). Cox multivariate analysis showed that high ANCCA expression was an independent prognostic factor for both OS (hazard ratio (HR) = 4.954, 95 % confidence interval (CI) = 1.537-15.966; P = 0.007) and DFS of patients with EC (HR = 4.237, 95 % CI = 1.295-13.859; P = 0.017). We identified ANCCA protein expression as a novel independent poor prognostic indicator in EC.


Asunto(s)
Adenosina Trifosfatasas/biosíntesis , Biomarcadores de Tumor/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Neoplasias Endometriales/genética , Recurrencia Local de Neoplasia/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Adulto , Anciano , Biomarcadores de Tumor/genética , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Neoplasias Endometriales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica/métodos , Metástasis Linfática , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Pronóstico
9.
Jpn J Clin Oncol ; 45(9): 812-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26117495

RESUMEN

OBJECTIVE: Special AT-rich sequence-binding protein 1 is aberrantly expressed in various malignant tumors. However, the expression and function of special AT-rich sequence-binding protein 1 in cervical squamous cell carcinoma have not been reported. The objective of this study was to investigate the clinical significance of special AT-rich sequence-binding protein 1 in cervical squamous cell carcinoma. METHODS: In this study, we investigated the expression of special AT-rich sequence-binding protein 1 through immunohistochemistry in 25 normal cervix specimens and 167 cervical squamous cell carcinomas and analyzed its association with various clinicopathologic parameters, including patient outcome. RESULTS: Special AT-rich sequence-binding protein 1 protein was detected in 58 (34.7%) out of 167 patients and was highly related to International Federation of Gynecology and Obstetrics stage, histologic grade, lymph node metastasis, vascular-lymphatic invasion and recurrence of cervical squamous cell carcinoma. Patients with positive special AT-rich sequence-binding protein 1 expression had significantly lower overall survival and disease-free survival compared with patients with negative expression of special AT-rich sequence-binding protein 1 (P = 0.001 and P < 0.001, respectively). A multivariate Cox regression analysis revealed that special AT-rich sequence-binding protein 1 was an independent prognostic marker for both disease-free survival and overall survival of cervical squamous cell carcinoma patients (P = 0.038 and P = 0.010, respectively). A multivariate logistic regression analysis showed that special AT-rich sequence-binding protein 1 expression was strongly associated with lymph node metastasis (odds ratio = 2.497; P = 0.032). Sensitivity and specificity of special AT-rich sequence-binding protein 1 for lymph node metastasis were 61.0 and 73.8%, respectively. CONCLUSIONS: These results showed that special AT-rich sequence-binding protein 1 expression was associated with tumor progression, metastasis and poor prognosis in cervical squamous cell carcinoma. It may serve as a new prognostic biomarker or a target for improving the treatment efficiency of patients with cervical squamous cell carcinoma.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Neoplasias del Cuello Uterino/diagnóstico , Adulto , Anciano , Biomarcadores/metabolismo , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Análisis Multivariante , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Neoplasias del Cuello Uterino/mortalidad , Neoplasias del Cuello Uterino/patología
10.
Int J Gynecol Cancer ; 25(1): 4-11, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25347096

RESUMEN

OBJECTIVE: Special AT-rich sequence-binding protein 1 (SATB1), as a genome organizer, serves important functions in tumor progression and metastasis. The SATB1 is overexpressed in various malignant tumors. However, the expression and prognostic value of SATB1 in endometrial cancer remain unknown. The aim of this study was to explore the prognostic values of SATB1 expression in endometrial cancer. METHODS/MATERIALS: We investigated the expression of SATB1 in 172 untreated endometrial cancer tissues and 25 normal endometrial tissues through immunohistochemical staining. We also analyzed the association of SATB1 level with clinicopathologic parameters and determined its prognostic significance. RESULT: Special AT-rich sequence-binding protein 1 was expressed in 78 (45.3%) of the 172 endometrial cancer samples, but not in the normal endometrial samples. The positive expression of SATB1 was associated with clinicopathologic factors, such as International Federation of Gynecology and Obstetrics stage, histological grade, myometrial invasion depth, lymph node metastasis, vascular/lymphatic invasion, and recurrence. The patients with positive SATB1 expression had worse overall survival and disease-free survival rates than the patients with negative SATB1 expression (P < 0.001 for both). Multivariate Cox analysis indicated that SATB1 was an independent parameter for overall survival (hazards ratio, 2.928; 95% confidence interval, 1.072-7.994; P = 0.036) and disease-free survival (hazards ratio, 2.825; 95% confidence interval, 1.111-7.181; P = 0.029). CONCLUSIONS: Results showed that SATB1 may be involved in tumor development and progression in endometrial cancer, may serve as a promising biomarker for predicting the prognosis of endometrial cancer patients, and thus may act as a novel target for treating endometrial carcinoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Adulto , Anciano , Neoplasias Endometriales/mortalidad , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Recurrencia Local de Neoplasia/mortalidad , Estadificación de Neoplasias , Pronóstico , Tasa de Supervivencia , Adulto Joven
11.
Int J Gynecol Cancer ; 25(8): 1453-60, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26166558

RESUMEN

OBJECTIVE: Lysine-specific demethylase 1 (LSD1) is a kind of flavin adenine dinucleotide-dependent amine oxidase that regulates normal cellular differentiation, gene activation, tumorigenesis, and progression. This study aims to detect the expression level of LSD1 in endometrial cancer and to explore its role in the progression and prognosis of endometrioid endometrial adenocarcinoma (EEA). METHODS: Immunohistochemistry was used to examine the expression of LSD1 in 206 EEA specimens, 50 benign endometrial lesion specimens, and 45 normal endometrium specimens. χ Analysis, Kaplan-Meier method, and multivariate Cox proportional hazard analysis were applied for the statistical analysis. RESULTS: Compared with normal endometrium and benign endometrial lesion (both P < 0.001), LSD1 was overexpressed in EEA. LSD1 expression was correlated with histological grade, International Federation of Gynecology and Obstetrics (FIGO) stage, vascular/lymphatic invasion, depth of myometrial invasion, and lymph node metastasis. Results of the Kaplan-Meier analysis indicated that LSD1 expression was associated with overall survival (OS) and disease-free survival (DFS) of EEA. The negative expression LSD1 group had longer OS and DFS than did the positive expression group. The difference was significant (both P < 0.001, log-rank test). Multivariate Cox regression analysis revealed that the LSD1 expression status was an independent prognostic factor for both OS (P = 0.027) and DFS (P = 0.016) of patients with EEA. CONCLUSIONS: Overexpression of LSD1 may contribute to the progression of EEA and may thus serve as a new biomarker to predict the prognosis of EEA.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Endometrioide/enzimología , Neoplasias Endometriales/enzimología , Histona Demetilasas/metabolismo , Adulto , Anciano , Pueblo Asiatico/etnología , Carcinoma Endometrioide/diagnóstico , Carcinoma Endometrioide/etnología , Estudios de Casos y Controles , Progresión de la Enfermedad , Neoplasias Endometriales/diagnóstico , Neoplasias Endometriales/etnología , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Tasa de Supervivencia
12.
Int J Gynecol Cancer ; 24(7): 1319-25, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25033256

RESUMEN

OBJECTIVE: Several inflammatory parameters are applied to predict the survival of patients with various cancers. Neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) are 2 nonspecific markers of systemic inflammation. This study aimed to evaluate the clinicopathologic and prognostic values of NLR and PLR in patients with cervical cancer undergoing primary radical hysterectomy with pelvic lymphadenectomy. METHODS: A total of 460 cervical cancer patients were enrolled in this study. These patients were histologically confirmed with cervical cancer from February 2005 to June 2008, at the Department of Gynecology, the Third Affiliated Hospital of Harbin Medical University, China. Their clinical and histopathological markers and complete blood counts were collected and analyzed. Prognostic factors were assessed by univariate and multivariate analyses. RESULTS: The median NLR and PLR were 2.213 and 150.9, respectively. The clinicopathologic analysis showed that NLR was highly associated with depth of stromal infiltration (P = 0.007) and lymph node metastasis (P = 0.003), and PLR was significantly related to tumor size (P = 0.020) and lymph node metastasis (P = 0.027).Univariate analysis identified high NLR as a statistically significant poor predictive factor for the progression-free survival (PFS) (P = 0.008) and overall survival (OS) (P = 0.014), and PLR exhibited no significance on PFS (P = 0.075) and OS (P = 0.110).Multivariable analysis showed that the NLR was an independent prognostic marker for PFS (hazard ratio, 1.799; 95% confidence interval, 1.069-3.028; P = 0.027), but not for OS (hazard ratio, 1.631; 95% confidence interval, 0.968-2.750; P = 0.066). CONCLUSIONS: Preoperative NLR and PLR were found to be correlated to unfavorable histopathologic features of cervical cancer. The preoperative NLR, but not PLR, may be used as a potential and easy biomarker for survival prognosis in patients with cervical cancer receiving initial radical hysterectomy with pelvic lymphadenectomy.


Asunto(s)
Adenocarcinoma/sangre , Adenocarcinoma/cirugía , Plaquetas/patología , Carcinoma de Células Escamosas/sangre , Carcinoma de Células Escamosas/cirugía , Linfocitos/patología , Neutrófilos/patología , Adenocarcinoma/diagnóstico , Adulto , Anciano , Recuento de Células Sanguíneas , Carcinoma de Células Escamosas/diagnóstico , Femenino , Humanos , Histerectomía , Persona de Mediana Edad , Terapia Neoadyuvante , Periodo Preoperatorio , Pronóstico , Estudios Retrospectivos , Resultado del Tratamiento , Neoplasias del Cuello Uterino , Adulto Joven
13.
Discov Oncol ; 15(1): 243, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38916621

RESUMEN

BACKGROUND: miR-200a-3p is involved in the progression of malignant behavior in various tumors, and its mechanism of action in endometrial cancer is speculated to be related to epithelial-mesenchymal transition (EMT). Therefore, this study explored the metastatic mechanism of miR-200a-3p and EMT in endometrial cancer, with the aim of identifying potential therapeutic targets. METHODS: qRT-PCR was used to analyze miR-200a-3p expression in HEC-1B and Ishikawa cell lines. The cell proliferation assay, transwell assay, and cell scratch test were used to assess changes in the malignant phenotypes of cells after regulating miR-200a-3p expression. Changes in EMT-related protein zinc finger E-box binding homeobox 1 (ZEB1) were detected after regulating miR-200a-3p expression. An endometrial carcinoma transplantation mouse tumor model was constructed, and multiple EMT-related proteins were examined. RESULTS: The expression of miR-200a-3p and ZEB1 in the endometrial cancer cell lines was higher than in normal endometrial epithelial cell lines (P < 0.05). After silencing miR-200a-3p, the expression of EMT-related protein ZEB1 increased, indicating a negative correlation. Simultaneously, the proliferation, invasion, and metastasis of endometrial cancer cells were significantly enhanced. After miR-200a-3p overexpression, the corresponding malignant phenotype was reversed (P < 0.05). In in vivo experiments, the degree of tumor malignancy and the expression level of EMT-related proteins were significantly reduced in the miR-200a-3p mimic group (P < 0.05). CONCLUSION: This study found that miR-200a-3p is a promising target, regulating the EMT process and promoting endometrial cancer progression.

14.
Int J Gynecol Cancer ; 23(2): 304-11, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23321718

RESUMEN

BACKGROUND: Wnt7a is a secreted glycoprotein that regulates normal cellular proliferation and differentiation, as well as tumorigenesis and progression. The aim of the present study was to detect the level of expression of Wnt7a in endometrial cancer and explore its role in progression and prognosis of endometrial cancer. METHODS: Immunohistochemistry was used to examine the expression of Wnt7a in 244 endometrial cancer specimens, in 48 benign endometrial lesion specimens, and 43 normal endometrium specimens. χ(2) Analysis, Kaplan-Meier analysis and log-rank test, and multivariate Cox proportional hazards analysis were applied for statistical analysis. RESULTS: Wnt7a was overexpressed in endometrial cancer compared with normal endometrium and benign endometrial lesion (both P < 0.001). Wnt7a expression was correlated with histological grade, International Federation of Gynecology and Obstetrics stage, depth of myometrial invasion, vascular/lymphatic invasion, and lymph node metastasis. The results of Kaplan-Meier analysis indicated that Wnt7a expression was associated with overall survival (OS) and disease-free survival (DFS) of endometrial cancer. The survival of negative expression Wnt7a group had longer OS and DFS compared with the group with positive expression. The difference was significant (both P < 0.001, log-rank test). Multivariate Cox regression analysis revealed that Wnt7a expression status was an independent prognostic factor for both OS and DFS (P = 0.034 and P = 0.009, respectively) of patients with endometrial cancer. CONCLUSIONS: Overexpression of Wnt7a may contribute to the progression of endometrial cancer and thus may serve as a new biomarker to predict the prognosis of endometrial cancer.


Asunto(s)
Carcinoma Endometrioide/diagnóstico , Neoplasias Endometriales/diagnóstico , Proteínas Wnt/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/mortalidad , Carcinoma Endometrioide/patología , Progresión de la Enfermedad , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/mortalidad , Neoplasias Endometriales/patología , Endometrio/metabolismo , Endometrio/patología , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia , Regulación hacia Arriba
15.
Pathol Res Pract ; 251: 154887, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37871443

RESUMEN

Long-stranded non-coding RNAs (lncRNAs) are RNA molecules that are longer than 200 nucleotides and do not code for proteins. They play a significant role in various biological processes, including epigenetics, cell cycle, and cell differentiation. Many studies have shown that the occurrence of human cancer is closely related to the abnormal expression of lncRNA. In recent years, lncRNAs have been a hot topic in cancer research. TRPM2-AS, a novel lncRNA, is aberrantly expressed in many human cancers, and its overexpression is strongly linked to poor clinical outcomes in patients. It has been demonstrated that TRPM2-AS acts as a ceRNA, participates in signaling pathways, and interacts with biological proteins and other molecular mechanisms to regulate gene expression. In addition, it can regulate the proliferation, migration, invasion, apoptosis, and treatment resistance of cancer cells. As a result, TRPM2-AS may be a potential target for cancer treatment and a possible biomarker for cancer prognosis. This review outlined the expression, biological processes, and molecular mechanisms of TRPM2-AS in various malignancies, and discussed potential therapeutic uses.


Asunto(s)
Neoplasias , ARN Largo no Codificante , Canales Catiónicos TRPM , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo , Neoplasias/genética , Transducción de Señal , Regulación Neoplásica de la Expresión Génica/genética
16.
Cell Signal ; 109: 110730, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37244634

RESUMEN

This study clarified the possible molecular mechanisms by which the miR-139-5p/SOX4/TMEM2 axis affected angiogenesis and tumorigenesis of ovarian cancer (OC) based on GEO microarray datasets and experimental support. The expression of miR-139-5p and SOX4 was examined in clinical OC samples. Human umbilical vein endothelial cells (HUVECs) and human OC cell lines were included in vitro experiments. Tube formation assay was conducted in HUVECs. The expression of SOX4, SOX4, and VEGF in OC cells was identified using Western blot and immunohistochemistry. Luciferase assays were conducted to validate the targeting relationship between miR-139-5p and SOX4 and between SOX4 and TMEM2. A RIP assay assessed the binding of SOX4 and miR-139-5p. The impact of miR-139-5p and SOX4 on OC tumorigenesis in vivo was evaluated in nude mice. SOX4 was up-regulated, while miR-139-5p was down-regulated in OC tissues and cells. Ectopic miR-139-5p expression or SOX4 knockdown inhibited angiogenesis and tumorigenicity of OC. By targeting SOX4 in OC, miR-139-5p lowered VEGF expression, angiogenesis, and TMEM2 expression. The miR-139-5p/SOX4/TMEM2 axis also reduced VEGF expression and angiogenesis, which might curtail OC growth in vivo. Collectively, miR-139-5p represses VEGF expression and angiogenesis by targeting the transcription factor SOX4 and down-regulating TMEM2 expression, thereby impeding OC tumorigenesis.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Ratones , Animales , Humanos , Femenino , MicroARNs/metabolismo , Ratones Desnudos , Factor A de Crecimiento Endotelial Vascular/genética , Proliferación Celular/fisiología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Carcinogénesis/genética , Transformación Celular Neoplásica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Análisis por Micromatrices , Línea Celular Tumoral , Factores de Transcripción SOXC/genética
17.
ACS Appl Mater Interfaces ; 15(22): 26484-26495, 2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37218712

RESUMEN

Cisplatin (CDDP) is a widely used chemotherapeutic drug with proven efficacy for treating tumors. However, its use has been associated with severe side effects and eventually leads to drug resistance, thus limiting its clinical application in patients with ovarian cancer (OC). Herein, we aimed to investigate the success rate of reversing cisplatin resistance using a synthetic, multitargeted nanodrug delivery system comprising a Mn-based metal-organic framework (Mn-MOF) containing niraparib (Nira) and CDDP alongside transferrin (Tf) conjugated to the surface (Tf-Mn-MOF@Nira@CDDP; MNCT). Our results revealed that MNCT can target the tumor site, consume glutathione (GSH), which is highly expressed in drug-resistant cells, and then decompose to release the encapsulated Nira and CDDP. Nira and CDDP play a synergistic role in increasing DNA damage and apoptosis, exhibiting excellent antiproliferation, migration, and invasion activities. In addition, MNCT significantly inhibited tumor growth in tumor-bearing mice and exhibited excellent biocompatibility without side effects. Furthermore, it depleted GSH, downregulated multidrug-resistant transporter protein (MDR) expression, and upregulated tumor suppressor protein phosphatase and tensin homolog (PTEN) expression, consequently reducing DNA damage repair and reversing cisplatin resistance. These results indicate that multitargeted nanodrug delivery systems can provide a promising clinical approach to overcoming cisplatin resistance. This study provides an experimental basis for further investigation into multitargeted nanodrug delivery systems to reverse cisplatin resistance in patients with OC.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias Ováricas , Humanos , Femenino , Animales , Ratones , Cisplatino/farmacología , Cisplatino/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Transferrina/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Nanopartículas/uso terapéutico , Línea Celular Tumoral , Resistencia a Antineoplásicos
18.
Adv Sci (Weinh) ; 10(36): e2303812, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37973560

RESUMEN

Protein arginine methyltransferase (PRMT) plays essential roles in tumor initiation and progression, but its underlying mechanisms in the treatment sensitivity of endometrial cancer (EC) remain unclear and warrant further investigation. Here, a comprehensive analysis of the Cancer Genome Atlas database and Clinical Proteomic Tumor Analysis Consortium database identifies that PRMT3 plays an important role in EC. Specifically, further experiments show that PRMT3 inhibition enhances the susceptibility of EC cells to ferroptosis. Mechanistically, PRMT3 interacts with Methyltransferase 14 (METTL14) and is involved in its arginine methylation. In addition, PRMT3 inhibition-mediated METTL14 overexpression promotes methylation modification via an m6 A-YTHDF2-dependent mechanism, reducing Glutathione peroxidase 4 (GPX4) mRNA stability, increasing lipid peroxidation levels, and accelerating ferroptosis. Notably, combined PRMT3 blockade and anti-PD-1 therapy display more potent antitumor effects by accelerating ferroptosis in cell-derived xenograft models. The specific PRMT3 inhibitor SGC707 exerts the same immunotherapeutic sensitizing effect in a patient-derived xenograft model. Notably, blocking PRMT3 improves tumor suppression in response to cisplatin and radiation therapy. Altogether, this work demonstrates that PRMT3 depletion is a promising target for EC.


Asunto(s)
Arginina , Neoplasias Endometriales , Humanos , Femenino , Arginina/metabolismo , Proteómica , Metilación , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Metiltransferasas/metabolismo
19.
J Biol Chem ; 286(49): 42371-42380, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22020941

RESUMEN

Aberrant growth of vascular smooth muscle cells (VSMCs) is a major cellular event in the pathogenesis of many proliferative vascular diseases. Recently, microRNA-31 (miR-31) has been found to play a critical role in cancer cell proliferation. However, the biological role of miR-31 in VSMC growth and the mechanisms involved are currently unknown. In the present study, the expression of rat mature miR-31 (rno-miR-31) was determined in cultured VSMCs and in rat carotid arteries. We identified that rno-miR-31 is an abundant miRNA in VSMCs, and its expression was significantly increased in proliferative VSMCs and in vascular walls with neointimal growth. The up-regulation of rno-miR-31 in proliferative VSMCs was inhibited by the inhibitor of mitogen-activated protein kinase/extracellular regulated kinase (MAPK/ERK). By both gain-of-function and loss-of-function approaches, we demonstrated that rno-miR-31 had a proproliferative effect on VSMCs. We further identified that LATS2 (large tumor suppressor homolog 2) is a downstream target gene product of rno-miR-31 that is involved in rno-miR-31-mediated effect on VSMC proliferation. The LATS2 as a target gene protein of rno-miR-31 is verified in vivo in balloon-injured rat carotid arteries. The results suggest that MAPK/ERK/miR-31/LATS2 may represent a novel signaling pathway in VSMC growth. miR-31 is able to enhance VSMC proliferation via its downstream target gene product, LATS2.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica , MicroARNs/biosíntesis , Músculo Liso Vascular/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Arterias Carótidas/patología , Línea Celular , Proliferación Celular , Humanos , Masculino , MicroARNs/genética , Modelos Biológicos , Ratas , Ratas Sprague-Dawley , Transducción de Señal
20.
J Surg Oncol ; 105(2): 206-11, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21815150

RESUMEN

BACKGROUND: The use of neoadjuvant chemotherapy (NACT) for the treatment of locally advanced cervical cancer (LACC) remains controversial. In current clinical practice, platinum-based chemotherapy is the major option for patients with LACC. However, serious adverse events have been reported after platinum-based chemotherapy treatment for LACC patients. In this study, the authors evaluated whether nedaplatin and paclitaxel (NP), as a new NACT regimen, offers less toxicity and better long-term efficacy for LACC (stages IB2-IIB) treatment. Comparisons between NP and paclitaxel and cisplatin (PC) in terms of toxicity and long-term efficacy are also presented. METHODS: The authors retrospectively reviewed 252 consecutive patients with LACC, of whom 104 received NP; the others received PC. Toxicity was assessed according to the International WHO (1979) criteria for chemotherapy side effects, and the chi-squared test was used to identify whether there was a statistically significant difference in toxicity between the NP regimen and the PC regimen. A univariate and a Cox regression model were used to assess whether the patients who were administered NP were statistically significantly different from those who were administered PC with respect to the disease-free survival rate (DFS) and the overall survival rate (OS). RESULTS: The overall response rate for NP and PC were 80.77% and 68.24%, respectively (P = 0.0267). The incidences of toxic reactions for NP and PC were 32.69% and 85.14%, respectively (P < 0.0001). The DFS for patients who were given the NP and the PC regimens were 81.41% and 67.28%, respectively (P = 0.014). The OS was 81.54% for patients who received the PC program and 93.89% for those who received the NP program (P = 0.0084). The NP program participants experienced a significant increase in the survival rate when compared to the group that received the PC program (DFS hazard ratio = 0.539, P = 0.0144 and OS hazard ratio = 0.354, P = 0.0077). CONCLUSIONS: NP NACT followed by radical hysterectomy offers a higher response rate, lower incidence of toxic reactions and better long-term DFS and OS for patients with LACC compared with the chemotherapy regimen of PC followed by radical hysterectomy.


Asunto(s)
Adenocarcinoma de Células Claras/tratamiento farmacológico , Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Terapia Neoadyuvante , Neoplasias del Cuello Uterino/tratamiento farmacológico , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adenocarcinoma/cirugía , Adenocarcinoma de Células Claras/mortalidad , Adenocarcinoma de Células Claras/secundario , Adenocarcinoma de Células Claras/cirugía , Adulto , Anciano , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Carcinoma de Células Escamosas/cirugía , Quimioterapia Adyuvante , Cisplatino/administración & dosificación , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Histerectomía , Persona de Mediana Edad , Estadificación de Neoplasias , Compuestos Organoplatinos/administración & dosificación , Paclitaxel/administración & dosificación , Estudios Retrospectivos , Tasa de Supervivencia , Factores de Tiempo , Resultado del Tratamiento , Neoplasias del Cuello Uterino/mortalidad , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/cirugía
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda