Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Publication year range
1.
Mol Med Rep ; 26(2)2022 Aug.
Article in English | MEDLINE | ID: mdl-35762320

ABSTRACT

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that certain of the cell migration and invasion assay data shown in Figs. 2C and 5C were strikingly similar to data appearing in different form in other articles by different authors. Owing to the fact that the contentious data in the above article had already been published elsewhere, or were already under consideration for publication, prior to its submission to Molecular Medicine Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor apologizes to the readership for any inconvenience caused. [the original article was published in Molecular Medicine Reports 16: 9692­9700, 2017; DOI: 10.3892/mmr.2017.7814].

2.
Environ Pollut ; 284: 117213, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33933780

ABSTRACT

PM2.5 (particles matter smaller aerodynamic diameter of 2.5 µm) exposure, a major environmental risk factor for the global burden of diseases, is associated with high risks of respiratory diseases. Heme-oxygenase 1 (HMOX1) is one of the major molecular antioxidant defenses to mediate cytoprotective effects against diverse stressors, including PM2.5-induced toxicity; however, the regulatory mechanism of HMOX1 expression still needs to be elucidated. In this study, using PM2.5 as a typical stressor, we explored whether microRNAs (miRNAs) might modulate HMOX1 expression in lung cells. Systematic bioinformatics analysis showed that seven miRNAs have the potentials to target HMOX1 gene. Among these, hsa-miR-760 was identified as the most responsive miRNA to PM2.5 exposure. More importantly, we revealed a "non-conventional" miRNA function in hsa-miR-760 upregulating HMOX1 expression, by targeting the coding region and interacting with YBX1 protein. In addition, we observed that exogenous hsa-miR-760 effectively elevated HMOX1 expression, reduced the reactive oxygen agents (ROS) levels, and rescued the lung cells from PM2.5-induced apoptosis. Our results revealed that hsa-miR-760 might play an important role in protecting lung cells against PM2.5-induced toxicity, by elevating HMOX1 expression, and offered new clues to elucidate the diverse functions of miRNAs.


Subject(s)
Heme Oxygenase-1 , MicroRNAs , Apoptosis , Epithelial Cells , Heme , Heme Oxygenase-1/genetics , Humans , MicroRNAs/genetics , Particulate Matter/toxicity
3.
Onco Targets Ther ; 13: 2261-2274, 2020.
Article in English | MEDLINE | ID: mdl-32214826

ABSTRACT

PURPOSE: ASB16 antisense RNA 1 (ASB16-AS1) is a cancer-associated long non-coding RNA that contributes to tumorigenesis and tumor development. Nevertheless, to the best of our knowledge, whether and how ASB16-AS1 is implicated in osteosarcoma (OS) malignancy remains unclear and therefore warrants exploration. Our current study focused on making in-depth investigation of ASB16-AS1 in OS. In the present study, the expression pattern of ASB16-AS1 in OS tissues and cell lines was analyzed. In addition, we examined the clinical value of ASB16-AS1 for OS patients. Furthermore, we explored the impacts of ASB16-AS1 on the malignant phenotype of OS cells in vitro and in vivo as well as the underlying mechanism. METHODS: ASB16-AS1, microRNA-760 (miR-760) and hepatoma-derived growth factor (HDGF) expressions were measured using reverse transcription-quantitative PCR. Cell proliferation and apoptosis were evaluated using CCK-8 and flow cytometry analyses, respectively, and cell migration and invasion were determined via cell migration and invasion assays. RESULTS: ASB16-AS1 expression was significantly elevated in OS tissues and cell lines, and increased ASB16-AS1 expression was related to patients' tumor size, TNM stage, and distant metastasis. The overall survival rate of OS patients presenting high ASB16-AS1 expression was shorter than that of patients presenting low ASB16-AS1 expression. Reduced ASB16-AS1 expression inhibited OS cell proliferation, migration, and invasion; promoted cell apoptosis; and impaired tumor growth in vivo. Mechanistically, ASB16-AS1 served as a sponge for miR-760 and positively modulated the expression of its target HDGF. Finally, inhibiting miR-760 and restoring HDGF expression abolished the impacts of ASB16-AS1 knockdown on the malignant characteristics of OS cells. CONCLUSION: ASB16-AS1 is a novel oncogenic lncRNA in OS cells. ASB16-AS1 increased HDGF expression by sponging miR-760, thereby conferring cancer-promoting roles in OS. ASB16-AS1 is a potential early diagnostic and therapeutic target in OS.

4.
World J Gastroenterol ; 25(45): 6619-6633, 2019 Dec 07.
Article in English | MEDLINE | ID: mdl-31832002

ABSTRACT

BACKGROUND: Gastric cancer (GC) has become a serious threat to people's health. Accumulative evidence reveals that dysregulation of numerous microRNAs (miRNAs) has been found during malignant formation. So far, the role of microRNA-760 (miR-760) in the development of GC is largely unknown. AIM: To measure the expression level of miR-760 in GC and investigate its role in gastric tumorigenesis. METHODS: Real-time quantitative polymerase chain reaction and Western blot analysis were used to measure the expression of miR-760 and G-protein-coupled receptor kinase interacting protein-1 (GIT1). Cell growth was detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and cell colony formation assays. Apoptosis was assessed by flow cytometric analysis. The relationship between miR-760 and GIT1 was verified by luciferase reporter assay. RESULTS: The results showed that the expression of miR-760 was decreased in GC and associated with poor clinical outcomes in GC patients. Furthermore, miR-760 restrained cell proliferation and cell colony formation and induced apoptosis in GC cells. In addition, miR-760 directly targeted GIT1 and negatively regulated its expression in GC. GIT1 was upregulated in GC and predicted a worse prognosis in GC patients. We also found that upregulation of GIT1 weakened the inhibitory effect of miR-760 in GC. CONCLUSION: In conclusion, miR-760 targets GIT1 to inhibit cell growth and promote apoptosis in GC cells. Our data demonstrate that miR-760 may be a potential target for the treatment of GC.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , MicroRNAs/physiology , Stomach Neoplasms/metabolism , Aged , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Flow Cytometry , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Up-Regulation
5.
Front Pharmacol ; 10: 1700, 2019.
Article in English | MEDLINE | ID: mdl-32082174

ABSTRACT

Although the functions of long noncoding RNA (lncRNA) called FOXD2 adjacent opposite strand RNA 1 (FOXD2-AS1) have been well studied in multiple human cancer types, its expression status and detailed roles in cervical cancer remain unknown and merit investigation. This study was aimed at assessing FOXD2-AS1 expression in cervical cancer and at determining its effects on the aggressive behavior of cervical cancer in vitro and in vivo. Expression of FOXD2-AS1 in cervical cancer tissues and cell lines was determined via reverse-transcription quantitative PCR. The effects of FOXD2-AS1 on cervical cancer cells were examined by a 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, flow-cytometric analysis, migration and invasion assays, and an in vivo tumorigenicity assay. FOXD2-AS1 was found to be significantly upregulated in cervical cancer tissues and cell lines. High FOXD2-AS1 expression was notably linked with the Federation of Gynecology and Obstetrics (FIGO) stage, lymph node metastasis, and depth of cervical invasion in patients with cervical cancer. Kaplan-Meier survival analysis revealed significantly shorter overall survival of patients when the tumor expression of FOXD2-AS1 was higher in comparison with those in patients with lower FOXD2-AS1 expression. In vitro functional assays revealed that downregulation of FOXD2-AS1 led to suppression of proliferation, migration, and invasiveness as well as to the induction of apoptosis of cervical cancer cells. In addition, FOXD2-AS1 silencing hindered tumor growth in vivo. Mechanism investigation revealed that FOXD2-AS1 functioned as a molecular sponge of microRNA-760 (miR-760). Furthermore, hepatoma-derived growth factor (HDGF) was validated as a direct target gene of miR-760 in cervical cancer cells. Moreover, an miR-760 knockdown reversed the effects of FOXD2-AS1 silencing on cervical cancer cells. FOXD2-AS1 possesses significant oncogenic activity in cervical cancer progression; this activity is mediated by sponging of miR-760 with consequent upregulation of HDGF. The FOXD2-AS1-miR-760-HDGF axis might harbor promising targets for novel treatment strategies of cervical cancer.

6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-838017

ABSTRACT

ObjectiveTo explore the role of long non-coding RNA (lncRNA)-H19 in the proliferation and migration of non-small cell lung cancer (NSCLC) cells and the molecular mechanisms. MethodsThe expressions of lncRNA-H19 in 20 NSCLC tissues and paired non-tumor tissues, which were collected from Changhai Hospital of Naval Medical University (Second Military Medical University) from Oct. 2015 to May 2016, were detected by real-time quantitative PCR (qPCR). We also examined lncRNA-H19 expressions in NSCLC cell lines A549 and NCI-H1299 and normal lung epithelial cell line BEAS-2B by qPCR. The proliferation and migration of A549 cells overexpressing lncRNA-H19 were detected by CCK-8 assay and Transwell assay, respectively. Bioinformatics analysis and duel-luciferase reporter assay were conducted to predict and confirm the interaction between microRNA (miRNA)-760 and lncRNA-H19. Western blotting analysis and RT-qPCR were performed to observe the influence of lncRNA-H19 overexpression on the expression of miRNA-760 and target gene nanog. MiRNA-760 was overexpressed in A549 cells, and its role in lncRNA-H19 promoting proliferation and migration of NSCLC cells was observed. Results The expressions of lncRNA-H19 in NSCLC tissues and A549 and NCI-H1299 cells were significantly upregulated compared with those in normal tissues and BEAS-2B cells (all P<0.01). Overexpression of lncRNA-H19 significantly improved the proliferation ability (P<0.05) and migration ability (P<0.01) of A549 cells compared with the negative control group. The results of starBase v3.0 showed that lncRNA-H19 could specifically adsorb miRNA-760, and duel-luciferase reporter assay showed that lncRNA-H19 directly bound to miRNA-760. Compared with the negative control group, overexpression of lncRNA-H19 significantly inhibited miRNA-760 expression in A549 cells (P<0.05) and promoted the expression of the downstream gene nanog at mRNA and protein levels (all P<0.01). Overexpression of miRNA-760 significantly inhibited lncRNA-H19-induced proliferation and migration of A549 cells (all P <0.05). ConclusionLncRNA-H19 can promote the proliferation and migration of NSCLC cells through sponging miRNA-760 to regulate nanog gene expression.

7.
Oncol Lett ; 16(6): 7239-7244, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30546462

ABSTRACT

Previous studies have demonstrated that microRNA (miR)-760 serves an important role in various cancer types. However, to the best of our knowledge, its role in hepatocellular carcinoma (HCC) has not been fully elucidated. The current study investigated the prognostic role of miR-760 in HCC by using the Kaplan-Meier plotter database. The current data indicated that low expression of miR-760 was associated with higher overall survival (OS) for all patients with HCC from both the RNA-seq [hazard ratio (HR)=2.04; 95% confidence interval (CI)=1.44-2.89; P=4.9×10-5] and the non-commercial spotted microarray (HR=1.71; CI=1.05-2.76; P=0.028). In the RNA-seq platform, a lower expression of miR-760 was strongly associated with improved OS in male patients with HCC, but not in female patients with HCC. Additionally, low expression of miR-760 was associated with improved OS in patients with stage I, II and III HCC, and was associated with improved OS in Asian and Caucasian patients. The current results indicated that miR-760 serves as an oncogene for HCC and high expression of miR-760 is significantly associated with tumor progression and poor prognosis in patients with HCC.

8.
J Exp Clin Cancer Res ; 37(1): 83, 2018 Apr 16.
Article in English | MEDLINE | ID: mdl-29661228

ABSTRACT

BACKGROUND: Recent studies have reported that microRNAs (miRNAs) often function as negative post-transcriptional regulators with altered expression levels found in colorectal cancer (CRC). There have been few studies on miRNAs that regulate the oncogenic alterations in CRC. Here, we aim to explore the anti-cancer miRNA and the potential mechanisms by which miRNAs modulate CRC progression. METHODS: We performed an integrated analysis of CRC miRNA expression datasets in The Cancer Genome Atlas (TCGA). The miRNA with the lowest expression, miR-760, was validated in an independent validation sample cohort of 76 CRC tissues. Functional assays, such as CCK-8 assay, colony formation assay, and CFSE staining, were used to determine the oncogenic role of miR-760 in human CRC progression. Furthermore, western blotting and dual-luciferase reporter assay were used to determine the mechanism by which miR-760 promotes proliferation of CRC cells. Xenograft nude mouse models were used to determine the role of miR-760 in CRC tumorigenicity in vivo. Immunohistochemical assays were conducted to study the relationship between miR-760 expression and basic leucine zipper transcriptional factor ATF-like 3 (BATF3) expression in human CRC samples. RESULTS: miR-760 was markedly downregulated in CRC tissues, and low miR-760 expression was associated with poor prognosis among CRC patients. Upregulation of miR-760 suppressed CRC cell proliferation, whereas downregulation of miR-760 promoted CRC proliferation in vitro. Additionally, we identified BATF3 as a direct target of miR-760, and that the essential biological function of miR-760 during CRC progression both in vitro and in vivo is to suppress the expression of BATF3 and downstream cyclinD1 via AP-1 transcription factor. Finally, we showed a significant correlation between miR-760 and BATF3 expression in CRC tissues. CONCLUSIONS: miR-760 inhibited CRC growth by downregulating BATF3/AP-1/ cyclinD1 signaling.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , MicroRNAs/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...