Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros













Intervalo de año de publicación
1.
PLoS One ; 18(7): e0288672, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37432933

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0101530.].

2.
Cancer Gene Ther ; 30(4): 582-595, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36854894

RESUMEN

Cholangiocarcinoma is a highly aggressive malignant tumor disease with the increasing incidence and mortality. It's urgent to identify specific biomarkers for cholangiocarcinoma treatment and diagnosis. Recent studies have noted the importance of lncRNAs in cancer and the following downstream mechanism with miRNAs network has been a hotspot. This work aimed to discover the role of lncRNA HCG18 and its possible downstream mechanism in cholangiocarcinoma tumor progression. Initially, through bioinformatics tools, we observed abnormal expression of lncRNA HCG18 in cholangiocarcinoma. In vitro experiments like (CCK-8, EdU, colony formation, flow cytometry, transwell, wound healing assays) and animal study confirmed that lncRNA HCG18 served as a cancer-promoting gene, promoted cancer proliferation, migration and invasion abilities. Besides, we found cancer cell-secreted exosomes transitted HCG18 to surrounding tumor cells and accelerated tumor growth and metastasis. After that, we confirmed HCG18 directly interacted with miR-424-5p through FISH, RIP and dual luciferase reporter assays with negative modulation. The inhibition of miR-424-5p reversed the HCG18 knockdown induced suppression on cholangiocarcinoma cancer cells. More specific, miR-424-5p targeted to SOX9 contributed to cholangiocarcinoma growth and metastasis through mediating PI3K/AKT pathway. In conclusion, these findings provide solid evidence of lncRNAs/miRNAs regulation in cholangiocarcinoma progression.


Asunto(s)
Colangiocarcinoma , MicroARNs , ARN Largo no Codificante , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proliferación Celular/genética , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Colangiocarcinoma/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética
3.
J Gastrointest Surg ; 26(9): 1917-1929, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35689008

RESUMEN

PURPOSE: The number of neuroendocrine tumors (NETs) is gradually increasing worldwide, and those located in the small intestine (siNETs) are the most common. As some biological and clinical characteristics of tumors of the jejunum and the ileum differ, there is a need to assess the prognosis of individuals with siNETs of the jejunum and ileum separately. We generated a predictive nomogram by assessing individuals with siNETs from the Surveillance, Epidemiology, and End Results (SEER) database. METHODS: We used univariate Cox regression analysis to determine both the overall survival (OS) and the cancer-specific survival (CSS) of 2501 patients with a pathological confirmation of siNETs of the jejunum and ileum. To predict 3-, 5-, and 10-year OS of siNETs, a nomogram was generated based on a training cohort and validated with an external cohort. Accuracy and clinical practicability were evaluated separately by Harrell's C-indices, calibration plots, and decision curves. The correlation was examined between dissected lymph nodes and positive lymph nodes. RESULTS: Dissection of 7 or more lymph nodes significantly improved patient OS and was found to be a protective factor for patients with siNETs. In Cox regression analyses, age, primary site, tumor size, N stage, M stage, and regional lymph node examination were significant predictors in the nomogram. A significant positive correlation was found between dissected lymph nodes and positive lymph nodes. CONCLUSIONS: Patients with 7 or more dissected lymph nodes showed an accurate tumor stage and a better prognosis. Our nomogram accurately predicted the OS of patients with siNETs.


Asunto(s)
Neoplasias del Íleon , Neoplasias del Yeyuno , Tumores Neuroendocrinos , Humanos , Neoplasias del Íleon/mortalidad , Neoplasias del Íleon/patología , Íleon/patología , Neoplasias del Yeyuno/mortalidad , Neoplasias del Yeyuno/patología , Yeyuno/patología , Ganglios Linfáticos/patología , Estadificación de Neoplasias , Tumores Neuroendocrinos/mortalidad , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/cirugía , Nomogramas , Pronóstico , Programa de VERF
4.
Bioengineered ; 13(3): 6208-6221, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35200104

RESUMEN

Cholangiocarcinoma (abbreviated as CCA) accounts for about 3% of digestive tract tumors, which is a rare disease with relatively low incidence. Herein, we firstly discovered overexpression of microRNA-23a-3p (abbreviated as miR-23a-3p) in CCA tissues, as well as cell lines via bioinformatics prediction. Next, by conducting miR-23a-3p knockdown system in HUCCT1 cells and miR-23a-3p overexpression system in RBE cells, we investigated the biological effects of miR-23a-3p. Based on our findings, inhibition of miR-23a-3p was able to prevent cancer cell proliferation via colony formation, CCK-8, as well as EdU assays. Moreover, invasion as well as migration abilities of cells was examined by transwell assay and wound healing test. Animal study further verified that knockdown miR-23a-3p slowed down tumor growth and lung metastasis. In addition, we identified cholangiocarcinoma cells transferred miR-23a-3p through exosomes by a series of assays. Functional experiments have confirmed that exosomal miR-23a-3p could benefit for cancer cell growth and metastasis, serving as a cancer promoting gene. Furthermore, we found Dynamin3 (abbreviated as DNM3) turned out to be a target of miR-23a-3p, while DNM3 was down-regulated in cholangiocarcinoma. Knockdown DNM3 accelerated cancer cell development. Collectively, our findings firstly pointed out that exosomal miR-23a-3p was conducive to the progression of cholangiocarcinoma by interaction with DNM3, which provided potential evidence for cancer treatment.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , MicroARNs , Animales , Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Colangiocarcinoma/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo
5.
Acta Biochim Biophys Sin (Shanghai) ; 53(11): 1547-1557, 2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34596215

RESUMEN

5-Fluorouracil (5-FU) resistance has been long considered as an obstacle to the efficacy of chemotherapy in colorectal cancer (CRC). In this study, we demonstrated the role of miR-20b-5p-regulated syndecan-2 (SDC2) in 5-FU resistance of CRC cells. 5-FU-resistant SW480 CRC cells were established by treatment of SW480 cells with stepwise increase of 5-FU concentration. The results showed that SDC2 was expressed significantly higher in SW480/5-FU cells than in SW480/WT cells as revealed by quantitative real-time polymerase chain reaction and western blot analysis. MTT assay and BrdU assay showed that SDC2 overexpression led to increased cell survival rate, while SDC2 knockdown reversed the drug resistance of SW480/5-FU cells. Wound healing and transwell invasion assays revealed that knockdown of SDC2 inhibited the migratory and invasive ability of SW480/5-FU cells. Moreover, animal experiments indicated that si-SDC2 plays a suppressive role in tumor growth in vivo. We also confirmed that miR-20b-5p interacted with SDC2, which reversed the effect of SDC2 in SW480/5-FU cells via the c-Jun N-terminal kinase (JNK)/extracellular regulated protein kinases (ERK) signaling pathway. These findings showed that JNK/ERK signaling pathway is involved in miR-20b-5p/SDC2 axis-mediated 5-FU resistance in SW480/5-FU cells, indicating that the miR-20b-5p/SDC2 axis is a potential target for reversing 5-FU resistance in CRC.


Asunto(s)
Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Fluorouracilo/farmacología , MicroARNs/genética , Sindecano-2/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Emparejamiento Base , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Biología Computacional/métodos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Ratones Desnudos , MicroARNs/metabolismo , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Survivin/genética , Survivin/metabolismo , Sindecano-2/antagonistas & inhibidores , Sindecano-2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Aging (Albany NY) ; 13(12): 16786-16803, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162761

RESUMEN

Gastric cancer (GC) is one of the most common aggressive cancers. The discovery of an effective biomarker is necessary for GC diagnosis. In this study, we confirmed that Paired box gene 4 (PAX4) is up-regulated in GC tissues and cells via quantitative real time polymerase chain reaction (qRT-PCR), western blot and immunohistochemical staining. It was also identified that PAX4 contributed to GC cell proliferation, migration and invasion through Cell Counting Kit-8, BrdU, flow cytometry assay, colony formation assay, transwell assays, and wound healing assay. miR-27b-3p was confirmed with the binding site with PAX4 using ChIP assay and served as a tumor suppressor that inhibiting GC cell growth and metastasis, and reversed the effect of PAX4. Bioinformatics prediction and dual luciferase assay results demonstrated that miR-27b-3p targeted Grb2, which could alter the function of miR-27b-3p. Furthermore, the transcriptional control of PAX4-regulated miR-27b-3p activated the Ras-ERK pathway. Taken together, the PAX4/miR-27b-3p/Grb2 loop is known to be involved in GC cell promotion, and can be seen as a promising target for GC therapy.


Asunto(s)
Progresión de la Enfermedad , Proteína Adaptadora GRB2/metabolismo , Proteínas de Homeodominio/metabolismo , MicroARNs/metabolismo , Factores de Transcripción Paired Box/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Animales , Apoptosis/genética , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Factores de Transcripción Paired Box/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas ras/metabolismo
7.
Braz J Med Biol Res ; 54(9): e10390, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34076140

RESUMEN

Sorafenib (SOR) resistance is still a significant challenge for the effective treatment of hepatocellular carcinoma (HCC). The mechanism of sorafenib resistance remains unclear. Several microRNAs (miRNAs) have been identified as playing a role in impairing the sensitivity of tumor cells to treatment. We examined the mechanism behind the role of miR-92b in mediating sorafenib resistance in HCC cells. We detected that miR-92b expression was significantly upregulated in SOR-resistant HepG2/SOR cells compared to parental HepG2/WT cells. After transfection with miR-92b inhibitor, the proliferation of HepG2/SOR cells was remarkably weakened and rates of apoptosis significantly increased. PTEN was considered to be a functional target of miR-92b according to a luciferase reporter assay. Knockdown of PTEN significantly impaired the ability of miR-92b inhibitor on increasing sorafenib sensitivity of HepG2/SOR cells. Furthermore, we confirmed by western blotting and immunofluorescence that miR-92b can mediate sorafenib resistance by activating the PI3K/AKT/mTOR pathway in HCC cells by directly targeting PTEN. These findings further validate the mechanism of miR-92b in SOR resistance in HCC treatment.


Asunto(s)
Carcinoma Hepatocelular , Resistencia a Antineoplásicos , Neoplasias Hepáticas , MicroARNs , Sorafenib , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Sorafenib/farmacología , Serina-Treonina Quinasas TOR
8.
Int J Ophthalmol ; 14(5): 649-655, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34012878

RESUMEN

AIM: To investigate the protective effect of heme oxygenase-1 (HO-1) against H2O2-induced apoptosis in human ARPE-19 cells. METHODS: The lentiviral vector expressing HO-1 was prepared and transfected into apoptotic ARPE-19 cells induced by H2O2. Functional experiments including cell counting kit-8 (CCK-8) assay, flow cytometry (FCM) and mitochondrial membrane potential assay were conducted. RESULTS: The ultrastructure of ARPE-19 cells was observed using transmission electron microscope (TEM). It was found that exogenous HO-1 significantly ameliorated H2O2-induced loss of cell viability, apoptosis and intracellular levels of reactive oxygen species (ROS) in ARPE-19 cells. The overexpression of HO-1 facilitated the transfer of nuclear factor erythroid-2-related factor 2 (Nrf2) from cytoplasm to nucleus, which in turn upregualted expressions HO-1 and B-cell lymphoma-2 (Bcl-2). Furthermore, HO-1 upregulation further inhibited H2O2-induced release of cysteinyl aspartate specific proteinase-3 (caspase-3). CONCLUSION: Exogenous HO-1 protect ARPE-19 cells against H2O2-induced oxidative stress by regulating the expressions of Nrf2, HO-1, Bcl-2, and caspase-3.

9.
Aging (Albany NY) ; 13(6): 8665-8687, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33714198

RESUMEN

MicroRNAs (miRNAs) are thought to be involved in the development of cisplatin (DDP) resistance in gastric cancer (GC). Using RNA sequencing analysis (RNA-seq), we found that miR-95-3p is associated with DDP resistance in GC. We discovered that miR-95-3p is highly expressed in DDP-resistant GC tissues and cell lines (SGC7901/DDP and AGS/DDP). Furthermore, results from the BrdU and MTT assays indicated that miR-95-3p promotes GC cell proliferation. Additionally, data from transwell chamber assay, wound healing test and in vivo experiments illustrated that miR-95-3p can effectively promote invasion, migration and tumorigenic capacity, respectively, of DDP-resistant GC cells. Subsequently, results from dual luciferase assay and qRT-PCR collectively indicated that EMP1 is a target of miR-95-3p with inhibitory function through suppression of the EMT process and drug-resistance proteins. Furthermore, PI3K/AKT was identified as a downstream pathway of miR-95-3p, which promotes DDP resistance in GC. In summary, miR-95-3p helped develop DDP-resistance through down-regulation of EMP1 and increasing phosphorylation of the PI3K/Akt pathway in GC.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , MicroARNs/genética , Neoplasias Gástricas/patología , Animales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Xenoinjertos , Humanos , Ratones , Proteínas de Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Neoplasias Gástricas/metabolismo
10.
Braz. j. med. biol. res ; 54(9): e10390, 2021. graf
Artículo en Inglés | LILACS | ID: biblio-1249337

RESUMEN

Sorafenib (SOR) resistance is still a significant challenge for the effective treatment of hepatocellular carcinoma (HCC). The mechanism of sorafenib resistance remains unclear. Several microRNAs (miRNAs) have been identified as playing a role in impairing the sensitivity of tumor cells to treatment. We examined the mechanism behind the role of miR-92b in mediating sorafenib resistance in HCC cells. We detected that miR-92b expression was significantly upregulated in SOR-resistant HepG2/SOR cells compared to parental HepG2/WT cells. After transfection with miR-92b inhibitor, the proliferation of HepG2/SOR cells was remarkably weakened and rates of apoptosis significantly increased. PTEN was considered to be a functional target of miR-92b according to a luciferase reporter assay. Knockdown of PTEN significantly impaired the ability of miR-92b inhibitor on increasing sorafenib sensitivity of HepG2/SOR cells. Furthermore, we confirmed by western blotting and immunofluorescence that miR-92b can mediate sorafenib resistance by activating the PI3K/AKT/mTOR pathway in HCC cells by directly targeting PTEN. These findings further validate the mechanism of miR-92b in SOR resistance in HCC treatment.


Asunto(s)
Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/tratamiento farmacológico , Resistencia a Antineoplásicos , MicroARNs/genética , Sorafenib/farmacología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/tratamiento farmacológico , Transducción de Señal , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Fosfohidrolasa PTEN/genética , Serina-Treonina Quinasas TOR
11.
Biochem Biophys Res Commun ; 531(4): 480-489, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32807493

RESUMEN

Currently, more and more studies show that aberrantly expressed microRNAs (miRNAs) are important driving factors for the pathogenesis of hepatocellular carcinoma (HCC). Based on the TCGA and GEO databases, miR-660-5p was identified as a possible target for HCC in this study.In HCC tissues, miR-660-5pexpressionwasparticularly high, and this was confirmed inHCC cell lines. The upregulatedmiR-660-5p showed correlations with tumor size, tumor number, TNM stage and histological grade. In vitro experimental data, aswellas in vivo evidence showed that miR-660-5p has the ability to significantly enhance the cell proliferation rate, clone formation, migration, invasion, and tumorigenic capacity of HCC cells. YWHAH is validated that targeted by miR-660-5p using dual luciferase reporter assay. Knockdown of YWHAH has been shown to partially reverse the tumor suppressive function of miR-660-5p inhibitor. Furthermore, miR-660-5p/YWHAH axis could activate PI3K/AKT pathway, which promoted EMT and cell cycle processes. In conclusion, this study illustrated the function of miR-660-5p/YWHAH axis in HCC and provided potential targets for treating HCC.


Asunto(s)
Proteínas 14-3-3/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , MicroARNs/genética , Proteínas 14-3-3/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cell Mol Biol (Noisy-le-grand) ; 66(1): 95-100, 2020 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-32359391

RESUMEN

The aim of this study is to   investigate the effect of miRNA-92a on GC cell proliferation, migration and invasiveness, and the mechanism(s) involved.  Four GC cell lines (SGC-7901, BGC-823, MKN45 and HGC-27) and normal human gastric epithelial cells (GES1) were used in this study. MicroRNA-92a mimics or inhibitor were transfected into the cells. The results of transfection were assessed using real-time quantitative polymerase chain reaction (qRT-PCR). Cell proliferation, migration, invasiveness and apoptosis were determined using cell counting kit 8 (CCK-8), scratch test, Transwell invasion assay, and flow cytometric analysis, respectively. The protein target of miRNA-92a was predicted using Bioinformatics. The expression of FOXO1 protein was measured using Western blotting. The expression of miRNA-92a was significantly upregulated in GC cells, relative to normal gastric epithelial cells (p < 0.05). Overexpression of miRNA-92a significantly promoted the proliferation, migration and invasiveness of GC cells, but significantly inhibited their apoptosis (p < 0.05). MicroRNA-92a directly targeted FOXO1 gene, and significantly reduced its protein expression. Overexpression of miRNA-92a promotes the proliferation, migration and invasiveness of GC cells, and plays a role similar to that of oncogene. It directly targets FOXO1 gene by inhibiting its protein expression.


Asunto(s)
Proteína Forkhead Box O1/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Proteína Forkhead Box O1/metabolismo , Genes Reporteros , Humanos , Luciferasas/genética , MicroARNs/genética , Invasividad Neoplásica , ARN Mensajero/genética , ARN Mensajero/metabolismo
13.
J Cell Biochem ; 121(1): 142-151, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31211445

RESUMEN

BACKGROUND: Cell division cycle associated protein-3 (CDCA3) has been reported frequently upregulated in various cancers. It has been progressively realized that changed DNA methylations occur in diverse carcinomas. However, the concrete involvement of CDCA3 and DNA methylation in gastric cancer (GC) still needs to be further elucidated. METHODS: In this study, quantitative reverse-transcription polymerase chain reaction (PCR) was utilized to determine the relative expressions of CDCA3 in GC and normal tissue samples. The methylation condition of CDCA3 was determined by bisulfite-sequencing PCR (BSP) and methylation-specific PCR (MSP). A chromatin immunoprecipitation (ChIP) assay and luciferase activity assay was used for the interaction between transcription factors and promoters and binding site determination, respectively. The effects of knockdown or overexpression of specificity protein 1 (SP1) or CDCA3 on GC cells in vitro were further assessed via wound healing assay, colony formation assay, and matrigel invasion assay. RESULTS: In comparison to paired normal tissues, CDCA3 expressions were significantly increased in the GC tissues. The CDCA3 expression was regulated by DNA methylation, with the CpG island hypomethylation responsible for CDCA3 upregulation of GC. ChIP assays verified that the activity of SP1 binding to the CDCA3 promoter was dramatically increased. When the CDCA3 expression was downregulated in MKN45 cells by knockdown SP1, the proliferation ability, healing ability, and invasive ability were significantly suppressed. CONCLUSION: The process by which SP1 bound to the nearest promoter region was expedited in GC cells, by which DNA was hypomethylated and CDCA3 expression was promoted. The effect on cell proliferation and invasion by CDCA3 was under the regulation of SP1 and also affected by hypomethylation of DNA.


Asunto(s)
Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción Sp1/metabolismo , Neoplasias Gástricas/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Islas de CpG , Regulación hacia Abajo , Mucosa Gástrica/metabolismo , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estómago , Neoplasias Gástricas/patología , Activación Transcripcional , Cicatrización de Heridas
14.
Cell Prolif ; 53(1): e12630, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31713929

RESUMEN

OBJECTIVES: miR-92b has been reported to play critical roles in several carcinomas; however, our understanding of the mechanisms by which miR-92b stimulates gastric cancer (GC) is incomplete. The aim of this study was to investigate the clinical significance and functional relevance of miR-92b in GC. MATERIALS AND METHODS: Expression of miR-92b in GC and peritumoural tissues was determined using qRT-PCR, in situ hybridization and bioinformatics. CCK-8, colony formation and fluorescence-activated cell sorting assays were utilized to explore the effect of miR-92b on GC cells. A luciferase reporter assay and Western blotting were employed to verify miR-92b targeting of DAB2IP. Furthermore, Western blotting was used to evaluate the levels of DAB2IP and PI3K/Akt signalling pathway-related proteins. RESULTS: In this study, we found that miR-92b was upregulated in GC tissues compared with peritumoural tissues. Overexpression of miR-92b promoted cell proliferation, colony formation, and G0 /G1 transition and decreased apoptosis. Our results indicated that miR-92b repressed the expression of DAB2IP and that loss of DAB2IP activated the PI3K/AKT signalling pathway. Overexpression of DAB2IP rescued the effects of miR-92b in GC cells. Finally, our results demonstrated a significant correlation between miR-92b expression and DAB2IP expression in GC tissues. CONCLUSIONS: Our results suggest that miR-92b promotes GC cell proliferation by activating the DAB2IP-mediated PI3K/AKT signalling pathway. The miR-92b/DAB2IP/PI3K/AKT signalling axis may be a potential therapeutic target to prevent GC progression.


Asunto(s)
MicroARNs/metabolismo , ARN Neoplásico/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Masculino , MicroARNs/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Neoplásico/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteínas Activadoras de ras GTPasa/genética
15.
Biomed Pharmacother ; 121: 109630, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31707342

RESUMEN

PURPOSE: In this study, we aimed to elucidate the biological roles of Syndecan-2 (SDC2) in colorectal cancer (CRC), thereby further understanding its clinical role. METHODS: The expression of SDC2 was assessed by qRT-PCR and Western blot analysis. To understand the potential biological role of SDC2, we also explored the correlation between its expression level and clinicopathologic parameters. By using MTT, plate colony formation assay, Transwell invasion assays, and flow cytometry in vitro, the biological impact of SDC2 on CRC cell proliferation, migration, invasion, and apoptosis. In addition, the related signaling pathways were investigated. RESULTS: SDC2 expression was significantly upregulated in CRC tissues. The expression of SDC2 was highly associated with four parameters, i.e., stage (P < 0.01), vascular invasion (P = 0.0045), lymph node metastasis (P=0.0018), and distant metastasis (P = 0.0019). Knockdown of SDC2 significantly reduced proliferation, migration, and invasion of HCT116 and SW480 cells, and induced cell apoptosis. Moreover, SDC2 promoted epithelial-mesenchymal transition (EMT) in CRC cells, whereas the ratio of p-MEK/MEK and p-ERK/ERK markedly reduced after depleting SDC2. CONCLUSION: During CRC development, overexpression of SDC2 plays a carcinogenic role in CRC. Therapeutic solutions targeting SDC2 may provide potential insights into CRC prevention and treatment.


Asunto(s)
Neoplasias Colorrectales/etiología , Transición Epitelial-Mesenquimal , Sistema de Señalización de MAP Quinasas/fisiología , Sindecano-2/fisiología , Adulto , Anciano , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica
16.
J Cell Biochem ; 120(9): 14813-14821, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31119774

RESUMEN

Meibomian gland dysfunction (MGD) is an epidemic chronic ocular inflammation. However, little is known about its effective treatment. Here, this study identified important MGD-related genes, core regulators, and potential drugs and their targets though integrating a series of bioinformational analyses. First, there were 665 differentially expression genes (DEGs) were identified. Then, 56 coexpression modules were exacted based on the expression of DEGs and their interactors. Moreover, core transcription factors (TF) significantly regulated modules were identified, including RELA, HIF1A, SIRT1, and MYC, which related to variety of eye diseases. Finally, the prediction of potential drugs and the identification of their target were performed. The results showed that artenimol, copper, and glutathione may have the remarkable curative effect or the toxicology to MGD. Moreover, their targets module gene LDHA (lactate dehydrogenase A), ENO1 (enolase 1), ALB (albumin), and PKM (pyruvate kinase M) are play important role in eye diseases. It suggests that these potential drugs may be useful for the treatment of MGD by acting on their targets. It provides valuable references for drug redirection and new drug development for drug developers, and provides individualized treatment strategies for tarsal gland dysfunction.


Asunto(s)
Artemisininas/farmacología , Biomarcadores/análisis , Regulación de la Expresión Génica/efectos de los fármacos , Disfunción de la Glándula de Meibomio/patología , Medicina de Precisión , Biomarcadores/metabolismo , Biomarcadores de Tumor/genética , Cobre/farmacología , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Glutatión/farmacología , Humanos , L-Lactato Deshidrogenasa/genética , Disfunción de la Glándula de Meibomio/clasificación , Disfunción de la Glándula de Meibomio/tratamiento farmacológico , Disfunción de la Glándula de Meibomio/genética , Fosfopiruvato Hidratasa/genética , Piruvato Quinasa/genética , Albúmina Sérica Humana/genética , Proteínas Supresoras de Tumor/genética
17.
Oncol Rep ; 41(4): 2471-2481, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30816466

RESUMEN

Gastric cancer (GC) is an aggressive and highly lethal gastrointestinal cancer, with an exceedingly poor prognosis. In the present study, the carcinogenic mechanism of human GC and the role of cell division cycle­associated 3 (CDCA3) were investigated. The expression levels of CDCA3 were investigated in GC samples and matched, peritumoral tissues by reverse transcription­quantitative polymerase chain reaction and immunohistochemical analysis. The effects of CDCA3 on cell proliferation were explored by Cell Counting Kit­8, colony formation, flow cytometric analysis and western blotting in vitro, and in vivo tumorigenesis in nude mice. The results demonstrated that CDCA3 expression was increased in human GC tissues compared with those in adjacent non­tumor tissues. Evaluation of the clinicopathological significance indicated that CDCA3 was closely associated with features of GC and patients with unfavorable overall survival times. CDCA3 overexpression resulted in the stimulation of cell growth and colony formation in vitro and xenograft tumors in vivo. Conversely, knockdown of CDCA3 inhibited these effects. Furthermore, the ectopic expression of CDCA3 in SGC7901 cells consistently promoted the cell cycle transition from the G0/G1 phase to the S phase; whereas knockdown of CDCA3 in BGC823 cells blocked the transition from the G0/G1 phase. Additionally, the present study revealed that the Ras signaling pathway was involved in CDCA3­mediated regulation of GC cell proliferation. CDCA3 activated the Ras signaling pathway to promote cell proliferation in vitro and in vivo in GC cells. Levels of CDCA3 greatly accelerated the progression of human GC. CDCA3 served as an oncogene, and may be a significant prognostic predictor and a novel therapeutic target for patients with GC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Gástricas/patología , Animales , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Gastrectomía , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Oncogenes/genética , Pronóstico , ARN Interferente Pequeño/metabolismo , Estómago/patología , Estómago/cirugía , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/cirugía , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Oncol Lett ; 15(6): 8789-8795, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29805618

RESUMEN

Nectins are Ca2+-independent immunoglobulin-like cell adhesion molecules that belong to a family of four members that function in a number of biological cellular activities. Nectin-4 is overexpressed in several types of human cancer; however, the functional and prognostic significance of Nectin-4 in gastric cancer (GC) remains unclear. In the present study, the reverse transcription-quantitative polymerase chain reaction and tissue microarray immunohistochemical analysis were used to investigate the expression of Nectin-4 in GC as well as its function in the prognosis of patients with GC. The results indicated that mRNA and protein expression of Nectin-4 were increased in tumor tissues compared with the matched non-tumor tissues. Expression of Nectin-4 was closely associated with differentiation (P=0.004), primary tumor (P=0.001), lymph node metastasis (P<0.001) and tumor-node-metastasis (TNM) stage (P<0.001). Positive Nectin-4 expression (P=0.001) and advanced TNM stage (P<0.001) were demonstrated to be associated with overall survival time in multivariate analyses. These results suggest that Nectin-4 may serve a significant function in GC and may serve as a novel clinic pathological biomarker and therapeutic target in GC.

19.
Hum Pathol ; 72: 107-116, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29208564

RESUMEN

Nectin-4, a member of the Nectin family that includes 4 Ca+-independent immunoglobulin-like cell adhesion molecules, plays a carcinogenic role in multiple cancers. However, Nectin-4 expression and its biological role in gastric cancer (GC) remain largely unknown. In this study, quantitative real-time polymerase chain reaction, Western blotting, and immunohistochemistry were used to evaluate the expression patterns of Nectin-4 in GC specimens and cell lines. We observed that high expression of Nectin-4 in GC patients was associated with TNM stage and lymph node metastasis status, and poor prognosis. In addition, cell proliferation and cell migration assays in vitro and tumorigenicity in vivo were performed to observe the effects of up-regulation and down-regulation of Nectin-4 expression on GC cell phenotypes. In further studies, the PI3K/AKT signaling pathway was revealed to be involved in Nectin-4-mediated GC progression. These results demonstrated that Nectin-4 had a promoter effect on human GC cell growth and motility, indicating that Nectin-4 may serve as an effective therapeutic target in GC.


Asunto(s)
Moléculas de Adhesión Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Gástricas/patología , Anciano , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Metástasis Linfática/genética , Masculino , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Estómago/patología , Neoplasias Gástricas/genética
20.
Oncol Lett ; 11(2): 1081-1088, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26893696

RESUMEN

Hepatocellular carcinoma (HCC) is the most common primary cancer of the liver. MicroRNA-497 (miR-497) is known to be downregulated in several types of human cancer; however, the expression, function and underlying mechanisms of miR-497 in HCC remain unclear. Therefore, the present study investigated miR-497 expression in HCC samples and HCC-derived cell lines using reverse transcription-quantitative polymerase chain reaction. The protein expression of one of the predicted common targets of miR-497, insulin-like growth factor-1 receptor (IGF-1R), was assessed using western blot analyses and immunohistochemistry. The role of miR-497 in regulating the proliferation of HCC-derived cells was also investigated in vitro and in vivo. Of 60 paired specimens from HCC patients, miR-497 was downregulated in 42 cancer specimens compared with adjacent non-cancer tissues. Western blotting and immunohistochemical analyses revealed that IGF-1R expression was significantly increased in HCC compared to control tissues. In addition, overexpression of miR-497 was observed to inhibit colony formation and tumor growth in MHCC-97H human HCC cells. Conversely, SMMC-7721 human HCC cells transfected with a miR-497 inhibitor exhibited enhanced colony formation and tumor growth. Finally, IGF-1R protein, phosphoinositide 3-kinase/Akt signaling pathway-associated proteins and cyclin pathway-associated proteins were differentially expressed between miR-497-overexpressing cells and miR-497-silenced cells. These results indicate that miR-497 may be a potentially effective gene therapy target.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA