Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
BMC Mol Cell Biol ; 23(1): 59, 2022 Dec 17.
Article in English | MEDLINE | ID: mdl-36528556

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) is one of the most common head and neck cancers. Long non-coding RNA HOXA-AS2 (lncRNA HOXA-AS2) have been extensively studied in various cancers. However, the expression and function of HOXA-AS2 in OSCC still remain unknown. The aim of this study is to investigate the roles of HOXA-AS2 in OSCC. METHODS: OSCC tissues and adjacent normal tissues were obtained from OSCC patients. RT-qPCR and Western blot assays were used to detect the expression of target genes in OSCC tissues or cells. Cells proliferation, migration and invasion were detected by CCK-8 and transwell assays, respectively. The target gene of HOXA-AS2 was confirmed by dual-luciferase reporter gene assay. RESULTS: We found that HOXA-AS2 expression was remarkably upregulated in OSCC tissues and cell lines. The downregulation of HOXA-AS2 inhibited cells proliferation, migration and invasion. Our bioinformatics analysis found that HOXA-AS2 can target miR-520c-3p, which was confirmed by dual-luciferase reporter gene assay. The expression of HOXA-AS2 was found to be negatively associated with miR-520c-3p in OSCC tissues. Moreover, sorting nexin 5 (SNX5), a downstream target of miR-520c-3p, was inhibited by miR-520c-3p overexpression. SNX5 was also increased in OSCC tissues and cell lines. Additionally, we found that the higher expression of SNX5 was strongly associated with the tumor grade of OSCC patients in Oncomine database. Most importantly, the knockdown of HOXA-AS2 induced cells apoptosis by promoting autophagy by regulating SNX5. CONCLUSION: HOXA-AS2 served an oncogene and promoted OSCC progression via the miR-520c-3p/SNX5 axis. Thus, HOXA-AS2 may be a new biomarker for diagnosis and treatment of OSCC.


Subject(s)
Head and Neck Neoplasms , RNA, Long Noncoding , Squamous Cell Carcinoma of Head and Neck , Humans , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Sorting Nexins/genetics , Sorting Nexins/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism
2.
Cancer Cell Int ; 22(1): 252, 2022 Aug 11.
Article in English | MEDLINE | ID: mdl-35953862

ABSTRACT

PURPOSE: Nectin-4 is specifically up-regulated in various tumors, exert crucial effects on tumor occurrence and development. Nevertheless, the role and molecular mechanism of Nectin-4 in osteosarcoma (OS) are rarely studied. METHODS: The expression of Nectin-4 and its relationship with clinical characteristics of OS were investigated using OS clinical tissues, tissue microarrays, TCGA, and GEO databases. Moreover, the effect of Nectin-4 on cell growth and mobility was detected by CCK-8, colony formation, transwell, and wound-healing assays. The RT-qPCR, Western blotting, and luciferase reporter assays were performed to explore molecular mechanisms through which Nectin-4 mediates the expression of miR-520c-3p, thus modulating PI3K/AKT/NF-κB signaling. In vivo mice models constructed by subcutaneous transplantation and tail vein injection were used to validate the functional roles of Nectin-4 and miR-520c-3p. RESULTS: Nectin-4 displayed a higher expression in OS tumor tissues compared with normal tissues, and its overexpression was positively associated with tumor stage and metastasis in OS patients. Functionally, Nectin-4 enhanced OS cells growth and mobility in vitro. Mechanistically, Nectin-4 down-regulated the levels of miR-520c-3p that directly targeted AKT-1 and P65, thus leading to the stimulation of PI3K/AKT/NF-κB signaling. In addition, the expression of miR-520c-3p was apparently lower in OS tissues than in normal tissues, and its low expression was significantly related to tumor metastasis. Furthermore, ectopic expression of miR-520c-3p markedly blocked the effect of Nectin-4 on OS cell growth and mobility. Knockdown of Nectin-4 could suppress the tumorigenesis and metastasis in vivo, which could be remarkably reversed by miR-520c-3p silencing. CONCLUSIONS: Nectin-4 as an oncogene can promote OS progression and metastasis by activating PI3K/AKT/NF-κB signaling via down-regulation of miR-520c-3p, which could represent a novel avenue for identifying a potential therapeutic target for improving patient outcomes.

3.
Mol Ther Nucleic Acids ; 29: 47-63, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-35795482

ABSTRACT

Hepatitis B virus (HBV) is a major risk factor for the development and progression of hepatocellular carcinoma (HCC). It has been reported that viral infection can interfere with the expression of cellular microRNA (miRNA) to affect oncogenesis. In this study, we showed that miR-520c-3p was upregulated in liver tumor specimens, and we revealed that HBV infection enhanced the expression of miR-520c-3p through the interaction of viral protein HBV X protein (HBx) with transcription factor CREB1. We further showed that miR-520c-3p induced by HBV transfection/infection caused epithelial-mesenchymal transition (EMT). Using the miRNA target prediction database miRBase and luciferase reporter assays, we identified PTEN as a novel target gene of miR-520c-3p and miR-520c-3p directly targeted PTEN's 3'-untranslated region. Moreover, we discovered that HBV promoted EMT via the miR-520c-3p-PTEN to activate AKT-NFκB signaling pathway, leading to increased HCC migration and invasion. Importantly, miR-520c-3p antagomir significantly represses invasiveness in HBx-induced hepatocellular xenograft models. Our findings indicate that miR-520c-3p is a novel regulator of HBV and plays an important role in HCC progression. It may serve as a new biomarker and molecular therapeutic target for HBV patients.

4.
Oncotarget ; 13: 725-746, 2022.
Article in English | MEDLINE | ID: mdl-35634241

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is considered to have more than 80% of all lung cancer cases, making it the leading cause of cancer-related deaths globally. MicroRNA (miRNA) deregulation has been seen often in NSCLC and has been linked to the disease's genesis, progression, and metastasis via affecting their target genes. MATERIALS AND METHODS: Our study focused on the functionality of down-regulated miRNAs in NSCLC. For this study, we used 91 miRNAs reported to be down-regulated in NSCLC. The targets of these miRNAs were chosen from miRNA databases with functionality in NSCLC, including miRBase, miRDB, miRTV, and others. Inter-regulatory miRNA-NSCLC networks were generated. Simulated annealing was used to improve the network's resilience and understandability. GSEA was used to examine 24607 genes reported experimentally in order to gain physiologically relevant information about the target miR-520c-3p. RESULTS: The study revealed functional prominence on miR-520c-3p, down-regulated during NSCLC. The involvement of miR-520c-3p in the PI3K/AKT/mTOR signaling pathway was recognized. CONCLUSIONS: The therapeutic usage by designing a synthetic circuit of miR-520c-3p was explored, which may help in suppressing tumors in NSCLC. Our study holds promise for the successful deployment of currently proposed miRNA-based therapies for malignant disorders, which are still in the early pre-clinical stages of development.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Systems Biology , TOR Serine-Threonine Kinases/metabolism
5.
Clin Transl Med ; 12(1): e662, 2022 01.
Article in English | MEDLINE | ID: mdl-35051313

ABSTRACT

BACKGROUND: Recurrence is the major cause of hepatocellular carcinoma (HCC) death. We aimed to identify circular RNA (circRNA) with predictive and therapeutic value for recurrent HCC. METHODS: Tissue samples from recurrent and non-recurrent HCC patients were subjected to circRNA sequencing and transcriptome sequencing. circKCNN2 was identified through multi-omics analyses. The effects of circKCNN2 on HCC were evaluated in cells, animals, database of The Cancer Genome Atlas, and a cohort with 130 HCC patients. circRNA precipitation, chromatin immunoprecipitation assay, RNA pull-down, luciferase assay, and cell experiments were applied to evaluate the interaction of circKCNN2 with miRNAs and proteins. The association between circKCNN2 and the therapeutic effect of lenvatinib was investigated in HCC cell lines and HCC tissue-derived organoids. RESULTS: The expression of circKCNN2 was downregulated in HCC tissues and predicted a favorable overall survival and recurrence-free survival. The expression of circKCNN2 was positively correlated with the parental gene, potassium calcium-activated channel subfamily N member (KCNN2). Nuclear transcription factor Y subunit alpha (NFYA) was proven to inhibit the promoter activity of KCNN2, downregulate the expression of KCNN2 and circKCNN2, and predict an unfavorable recurrence-free survival. Ectopic expression of circKCNN2 inhibited HCC cell proliferation, colony formation, migration, and tumor formation in a mouse model. miR-520c-3p sponged by circKCNN2 could reverse the inhibitory effect of circKCNN2 on HCC cells and down-regulate the expression of methyl-DNA-binding domain protein 2 (MBD2). The intratumoral expression of MBD2 predicted a favorable recurrence-free survival. circKCNN2 down-regulated the expression of fibroblast growth factor receptor 4 (FGFR4), which can be reversed by miR-520c-3p and knockdown of MBD2. Lenvatinib inhibited the expression of FGFR4 and upregulated the expression of circKCNN2 and MBD2. Ectopic expression of circKCNN2 in HCC cells enhanced the therapeutic effect of lenvatinib. However, the high inherent level of circKCNN2 in HCC cells was associated with lenvatinib resistance. CONCLUSIONS: circKCNN2, transcriptionally repressed by NFYA, suppresses HCC recurrence via the miR-520c-3p/MBD2 axis. Inherent level of circKCNN2 in HCC cells predisposes anti-tumor effect of lenvatinib possibly because both circKCNN2 and lenvatinib repress the expression of FGFR4. circKCNN2 may be a promising predictive biomarker and therapeutic agent for HCC recurrence.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , DNA-Binding Proteins/drug effects , MicroRNAs/drug effects , Small-Conductance Calcium-Activated Potassium Channels/pharmacology , Animals , Carcinoma, Hepatocellular/prevention & control , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , China , Disease Models, Animal , Liver Neoplasms/drug therapy , Liver Neoplasms/prevention & control , Mice , Recurrence , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Small-Conductance Calcium-Activated Potassium Channels/therapeutic use
6.
J Orthop Surg Res ; 16(1): 347, 2021 May 29.
Article in English | MEDLINE | ID: mdl-34051811

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) have been shown to be associated with osteoarthritis (OA) progression. This study aimed to explore the role of miR-520c-3p in OA progression. METHODS: Expression levels of miR-520c-3p and Growth arrest-specific 2 (GAS2) were detected using quantitative real-time PCR. The proliferation and apoptosis of cells were measured using cell counting kit 8 (CCK8) assay and flow cytometry. Furthermore, the protein levels of apoptosis-related markers, extracellular degradation markers, inflammatory response markers, and GAS2 were tested using quantitative real-time polymerase chain reaction (RT-PCR) and western blot (WB) analysis. In addition, the interaction between miR-520c-3p and GAS2 was examined using dual luciferase reporter assay. RESULTS: GAS2 was highly expressed, and miR-520c-3p was lowly expressed in OA cartilage tissues. miR-520c-3p could promote the proliferation and inhibit the apoptosis and inflammation of OA chondrocytes. miR-520c-3p could be sponged by GAS2, and its inhibitor could reverse the regulation of GAS2 on the biological functions of OA chondrocytes. GAS2 was a target of miR-520c-3p, which was identified by bioinformatic analysis and dual-luciferase reporter assay. Overexpression of GAS2 could inhibit the proliferation and promoted the apoptosis and inflammation of OA chondrocytes. CONCLUSION: Our data showed that miR-520c-3p might regulate the GAS2 to inhibit the progression of OA.


Subject(s)
Apoptosis , Cartilage/metabolism , Chondrocytes/metabolism , MicroRNAs/physiology , Microfilament Proteins/metabolism , Osteoarthritis/metabolism , Cells, Cultured , Down-Regulation , Humans , Interleukin-1beta , Osteoarthritis/genetics , Up-Regulation
8.
J Physiol Biochem ; 77(1): 47-61, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33411212

ABSTRACT

Endothelial injury, which can cause endothelial inflammation and dysfunction, is an important mechanism for the development of atherosclerotic plaque. This study aims to investigate the functional role of miR-520c-3p in vascular endothelium during inflammatory diseases such as atherosclerosis. Quantitative real-time PCR was used to detect miR-520c-3p expression in in human umbilical vein endothelial cells (HUVECs) after treatment with platelet-derived growth factor (PDGF). Furthermore, the effects of miR-520c-3p overexpression and silencing on cell proliferation, adhesion, and apoptosis were assessed. Bioinformatics analysis and Biotin-labeled miRNA pull-down assay were used to confirm the targets of miR-520-3p. Then, the effects of miR-520c-3p on AKT and NF-κB signaling pathways were detected by western blot. Herein, we observed that the expression level of miR-520c-3p was downregulated in HUVECs under PDGF stimulation. Overexpression of miR-520c-3p not only decreased cell adhesion but also promoted proliferation and inhibited apoptosis to protect the viability of endothelial cells. It was confirmed that RELA is the target of miR-520c-3p. MiR-520c-3p inhibited the protein phosphorylation of AKT and RELA, and si-RELA reversed the promotion of AKT and RELA protein phosphorylation by anti-miR-520c-3p. In summary, our study suggested that miRNA-520c-3p targeting RELA through AKT and NF-κB signaling pathways regulated the proliferation, apoptosis, and adhesion of vascular endothelial cells. We conclude that miR-520c-3p may play an important role in the suppression of endothelial injury, which could serve as a biomarker and therapeutic target for atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , MicroRNAs/physiology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/metabolism , Apoptosis , Cell Adhesion , Cell Proliferation , Human Umbilical Vein Endothelial Cells , Humans , Signal Transduction , THP-1 Cells
9.
Anticancer Agents Med Chem ; 21(2): 237-245, 2021.
Article in English | MEDLINE | ID: mdl-32357822

ABSTRACT

BACKGROUND: Doxorubicin (DOX) is one of the most common drugs used in cancer therapy, including Hepatocellular Carcinoma (HCC). Drug resistance is one of chemotherapy's significant problems. Emerging studies have shown that microRNAs (miRNAs) could participate in regulating this mechanism. Nevertheless, the impact of miRNAs on HCC chemoresistance is still enigmatic. OBJECTIVE: Investigating the role of microRNA-520c-3p (miR-520c-3p) in the enhancement of the anti-tumor effect of DOX against HepG2 cells. METHODS: Expression profile for liver-related miRNAs (384 miRNAs) has been analyzed on HepG2 cells treated with DOX using qRT-PCR. miR-520c-3p, the most deregulated miRNA, was selected for combination treatment with DOX. The expression level for LEF1, CDK2, CDH1, VIM, Mcl-1 and p53 was evaluated in miR-520c-3p transfected cells. Cell viability, colony formation, wound healing as well as apoptosis assays have been demonstrated. Furthermore, Mcl-1 protein level was measured using the western blot technique. RESULTS: The present data indicated that miR-520c-3p overexpression could render HepG2 cells chemo-sensitive to DOX through enhancing its suppressive effects on proliferation, migration, and induction of apoptosis. The suppressive effect of miR-520c-3p involved altering the expression levels of some key regulators of cell cycle, proliferation, migration and apoptosis, including LEF1, CDK2, CDH1, VIM, Mcl-1 and p53. Interestingly, Mcl-1 was found to be one of the potential targets of miR-520c-3p, and its protein expression level was down-regulated upon miR-520c-3p overexpression. CONCLUSION: Our data referred to the tumor suppressor function of miR-520c-3p that could modulate the chemosensitivity of HepG2 cells towards DOX treatment, providing a promising therapeutic strategy in HCC.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Doxorubicin/pharmacology , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , MicroRNAs/genetics , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/pathology
10.
Int J Biochem Cell Biol ; 131: 105873, 2021 02.
Article in English | MEDLINE | ID: mdl-33166679

ABSTRACT

Atherosclerosis is a chronic inflammatory disease, and it's the leading cause of death worldwide. Dysregulation of microRNAs (miRNAs) has been found to be associated with atherosclerosis. miR-520c-3p has been implicated in several types of cancer. However, little is known about the role of miR-520c-3p in atherosclerosis. In this study, we found that miR-520c-3p agomir decreased atherosclerotic plaque size, collagen content, the quantity of PCNA-positive cell and RelA/p65 expression of vascular smooth muscle cells (VSMCs) in the aortic valve of apoE-/- mice in vivo. The possible mechanisms of the protective effects of miR-520c-3p on atherosclerotic mice were then investigated in VSMCs. in vitro experiments showed that miR-520c-3p expressions were significantly reduced in human aortic vascular smooth muscle cell (HASMCs) treated with platelet-derived growth factor (PDGF-BB). miR-520c-3p mimics repress PDGF-BB-mediated the proliferation, migration and decrease in the percentage of cells in G2/M phase, which was associated with downregulation of RelA/p65. Mechanistically, miRNA pull-down, luciferase reporter and mRNA stability assays confirmed miR-520c-3p mimics was able to directly target 3'-UTR of RelA/p65 mRNA and decreased half-life of RelA/p65 mRNA in HASMCs. Overexpression of RelA/p65 reversed the inhibition of cell proliferation induced by miR-520c-3p mimics in HASMCs. In conclusion, our findings suggest that miR-520c-3p inhibits PDGF-BB-mediated the proliferation and migration of HASMCs by targeting RelA/p65, which may provide potential therapeutic strategies in atherosclerosis treatment.


Subject(s)
Atherosclerosis/genetics , MicroRNAs/genetics , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/genetics , Transcription Factor RelA/genetics , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/therapy , Becaplermin/pharmacology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Knockout, ApoE , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/therapy , Primary Cell Culture , Signal Transduction , Transcription Factor RelA/metabolism
11.
Aging (Albany NY) ; 12(16): 16304-16325, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32857725

ABSTRACT

The purpose of this article is to explore the function and mechanism of HOXD-AS1 in cholangiocarcinoma. TCGA, StarBase and JASPAR were applied to predict the differential expression and molecular mechanism. The qRT-PCR was conducted to detect molecular expression. The effect of HOXD-AS1 on tumor proliferation, metastasis and stemness was measured through corresponding experiments. ChIP, luciferase reporter and RIP assays were implemented to explore the regulatory mechanism of HOXD-AS1 in CCA. In this study, HOXD-AS1 expression was significantly upregulated in CCA tissues and cells compared with control groups, respectively. Increased HOXD-AS1 was markedly correlated with lymph node invasion, advanced TNM stage and poor survival of CCA patients. Moreover, HOXD-AS1 was confirmed to be an unfavorable independent prognostic factor for CCA patients. Functionally, gain- and loss-of-function experiments demonstrated that HOXD-AS1 facilitated tumor proliferation, migration, invasion, EMT, stemness and drug resistance in vitro and in vivo. For the mechanism, transcription factor SP1-induced HOXD-AS1 upregulated oncogene MYCN through competitively binding to miR-520c-3p. Furthermore, HOXD-AS1-induced malignant phenotypes were rescued by interfering miR-520c-3p and MYCN, respectively. SP1/HOXD-AS1/miR-520c-3p/MYCN plays a vital role in initiation and progression of CCA, and HOXD-AS1 is expected to be an efficient biomarker and therapeutic target.


Subject(s)
Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , MicroRNAs/metabolism , N-Myc Proto-Oncogene Protein/metabolism , RNA, Long Noncoding/metabolism , Sp1 Transcription Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/secondary , Disease Progression , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Invasiveness , RNA, Long Noncoding/genetics , Signal Transduction , Sp1 Transcription Factor/genetics , Xenograft Model Antitumor Assays
12.
Cell Physiol Biochem ; 51(2): 886-896, 2018.
Article in English | MEDLINE | ID: mdl-30466095

ABSTRACT

BACKGROUND/AIMS: Among different molecular candidates, there is growing data to support that long noncoding RNAs (lncRNAs) play a significant role in acute myeloid leukemia (AML). HOXA-AS2 is significantly overexpressed in a variety of tumors and associated with anti-cancer drug resistance, however, little is known regarding the expression and function of HOXA-AS2 in the chemoresistance of AML. In this study, we aimed to determine the role and molecular mechanism of HOXA-AS2 in adriamycin-based chemotherapy resistance in AML cells. METHODS: Quantitative real-time PCR was used to detect HOXA-AS2 expression in the BM samples and ADR cell lines, U/A and T/A cells. Furthermore, the effects of HOXA-AS2 silencing on cell proliferation and apoptosis were assessed in vitro by CCK8 and flow cytometry, and on tumor growth in vivo. Furthermore, bioinformatics online programs predicted and luciferase reporter assay were used to validate the association of HOXA-AS2 and miR-520c-3p in AML. RESULTS: In this study, we showed that HOXA-AS2 is significantly upregulated in BM samples from AML patients after treatment with adriamycin-based chemotherapy and in U/A and T/A cells. Knockdown of HOXA-AS2 inhibited ADR cell proliferation in vitro and in vivo and promoted apoptosis. Bioinformatics online programs predicted that HOXA-AS2 sponge miR-520c-3p at 3'-UTR with complementary binding sites, which was validated using luciferase reporter assay and anti-Ago2 RIP assay. HOXA-AS2 could negatively regulate the expression of miR-520c-3p in ADR cells. S100A4 was predicted as a downstream target of miR-520c-3p, which was confirmed by luciferase reporter assay. CONCLUSION: Our results suggest that HOXA-AS2 plays an important role in the resistance of AML cells to adriamycin. Thus, HOXA-AS2 may represent a therapeutic target for overcoming resistance to adriamycin-based chemotherapy in AML.


Subject(s)
Leukemia, Myeloid, Acute/pathology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , S100 Calcium-Binding Protein A4/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , S100 Calcium-Binding Protein A4/chemistry , S100 Calcium-Binding Protein A4/genetics
13.
Cell Physiol Biochem ; 50(6): 2124-2138, 2018.
Article in English | MEDLINE | ID: mdl-30415263

ABSTRACT

BACKGROUND/AIMS: Previous studies have demonstrated that long non-coding RNAs (lncRNAs) may play critical roles in cancer biology, including Hepatocellular carcinoma (HCC). The HOXA cluster antisense RNA2 (HOXA-AS2) lncRNA plays an important role in carcinogenesis, however, the underlying role of HOXA-AS2 in HCC remains unknown. The present study examined the effects of HOXA-AS2 on the progression of HCC, and explored the underlying molecular mechanisms. METHODS: Quantitative real-time PCR was used to detect HOXA-AS2 expression in HCC tissues and cell lines. Furthermore, the effects of HOXA-AS2 silencing and overexpression on cell proliferation, cell cycle, apoptosis, migration, and invasion were assessed in HCC in vitro and in vivo. Furthermore, bioinformatics online programs predicted and luciferase reporter assay were used to validate the association of HOXA-AS2 and miR-520c-3p in HCC cells. RESULTS: We observed that HOXA-AS2 was up-regulated in HCC tissues and cell lines. In vitro experiments revealed that HOXA-AS2 knockdown significantly inhibited HCC cells proliferation by causing G1 arrest and promoting apoptosis, whereas HOXA-AS2 overexpression promoted cell growth. Further functional assays indicated that HOXA-AS2 significantly promoted HCC cell migration and invasion by promoting EMT. Bioinformatics online programs predicted that HOXA-AS2 sponge miR-520c-3p at 3'-UTR with complementary binding sites, which was validated using luciferase reporter assay. HOXA-AS2 could negatively regulate the expression of miR-520c-3p in HCC cells. MiR-520c-3p was down-regulated and inversely correlated with HOXA-AS2 expression in HCC tissues. miR-520c-3p suppressed cell proliferation, invasion and migration in HCC cells, and enforced expression of miR-520c-3p attenuated the oncogenic effects of HOXA-AS2 in HCC cells. By bioinformatic analysis and dual-luciferase reporter assay, we found that miR-223-3p directly targeted the 3'-untranslated region (UTR) of Glypican-3 (GPC3), one of the key players in HCC. GPC3 was up-regulated in HCC tissues, and was negatively correlated with miR-520c-3p expression and positively correlated with HOXA-AS2 expression. CONCLUSION: In summary, our results suggested that the HOXA-AS2/miR-520c-3p/GPC3 axis may play an important role in the regulation of PTC progression, which could serve as a biomarker and therapeutic target for HCC.


Subject(s)
Cell Proliferation , Epithelial-Mesenchymal Transition , Glypicans/metabolism , RNA, Long Noncoding/metabolism , 3' Untranslated Regions , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , G1 Phase Cell Cycle Checkpoints , Glypicans/chemistry , Glypicans/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use
14.
Cell Physiol Biochem ; 50(5): 1659-1672, 2018.
Article in English | MEDLINE | ID: mdl-30384358

ABSTRACT

BACKGROUND/AIMS: Thyroid cancer is one of the most prevalent endocrine tumors. The present study examined the effects of lncRNA HOXA cluster antisense RNA2 (HOXA-AS2) on the progression of papillary thyroid cancer (PTC), and explored the underlying molecular mechanisms. METHODS: Quantitative real-time PCR was used to detect HOXA-AS2, miR-520c-3p and S100 calcium-binding protein A4 (S100A4) expression. Furthermore, the effects of HOXA-AS2 silencing and overexpression on cell proliferation, migration, and invasion were assessed in PTC in vitro by CCK8 and transwell assay. Furthermore, bioinformatics online programs predicted and luciferase reporter assay were used to validate the association of HOXA-AS2 and miR-520c-3p in PTC. RESULTS: We observed that HOXA-AS2 was up-regulated in PTC tissues. In vitro experiments revealed that HOXA-AS2 knockdown significantly inhibited cell growth in PTC in vitro and in vivo. Further functional assays indicated that HOXA-AS2 significantly promoted PTC cell migration and invasion by promoting EMT. Bioinformatics online programs predicted that HOXA-AS2 sponge miR-520c-3p at 3'-UTR with complementary binding sites, which was validated using luciferase reporter assay. HOXA-AS2 could negatively regulate the expression of miR-520c-3p in PTC cells. MiR-520c-3p was down-regulated in PTC tissues, and S100A4 was predicted as a downstream target of miR-520c-3p, which was confirmed by luciferase reporter assay. CONCLUSION: In summary, our results suggested that the HOXA-AS2/miR-520c-3p/S100A4 axis may play an important role in the regulation of PTC progression, which provides us with new insights into understanding the PTC.


Subject(s)
Carcinoma, Papillary/pathology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , S100 Calcium-Binding Protein A4/metabolism , Thyroid Neoplasms/pathology , 3' Untranslated Regions , Adult , Animals , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , S100 Calcium-Binding Protein A4/chemistry , S100 Calcium-Binding Protein A4/genetics , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Vimentin/metabolism
15.
Cell Cycle ; 17(13): 1637-1648, 2018.
Article in English | MEDLINE | ID: mdl-30081707

ABSTRACT

Osteosarcoma (OS) is the commonest primary malignant tumour originating from bone. Previous studies demonstrated that long non-coding RNAs (lncRNAs) could participate in both oncogenic and tumor suppressing pathways in various cancer, including OS. The HOXA cluster antisense RNA2 (HOXA-AS2) plays an important role in carcinogenesis, however, the underlying role of HOXA-AS2 in OS progression remains unknown. The aim of the present study was to evaluate the expression and function of HOXA-AS2 in OS. The qRT-PCR analysis was to investigate the expression pattern of HOXA-AS2 in OS tissues. Then, the effects of HOXA-AS2 on cell proliferation, cell cycle, apoptosis, migration, and invasion were assessed in OS in vitro. Furthermore, bioinformatics online programs predicted and luciferase reporter assay were used to validate the association of HOXA-AS2 and miR-520c-3p in OS cells. We observed that HOXA-AS2 was up-regulated in OS tissues. In vitro experiments revealed that HOXA-AS2 knockdown significantly inhibited OS cells proliferation by promoting apoptosis and causing G1 arrest, whereas HOXA-AS2 overexpression promoted cell proliferation. Further functional assays indicated that HOXA-AS2 significantly promoted OS cell migration and invasion by promoting epithelial-mesenchymal transition (EMT). Bioinformatics online programs predicted that HOXA-AS2 sponge miR-520c-3p at 3'-UTR with complementary binding sites, which was validated using luciferase reporter assay. HOXA-AS2 could negatively regulate the expression of miR-520c-3p in OS cells. In conclusion, our study suggests that HOXA-AS2 acts as a functional oncogene in OS.


Subject(s)
Cell Movement/genetics , MicroRNAs/metabolism , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA, Long Noncoding/metabolism , Adult , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/genetics , Neoplasm Invasiveness , RNA, Long Noncoding/genetics , Up-Regulation/genetics
16.
Oncotarget ; 8(28): 46090-46103, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28545023

ABSTRACT

The long non-coding RNA (lncRNA) HOXA cluster antisense RNA2 (HOXA-AS2) has recently been shown to be dysregulated and involved in the progression of several cancers. However, the biological role and clinical significance of HOXA-AS2 in the carcinogenesis of breast cancer are still unclear. In the present study, we found that HOXA-AS2 was up-regulated in human breast cancer tissues and cell lines and associated with clinicopathological characteristics. Silencing of HOXA-AS2 inhibited the progression of breast cancer cells in vitro and in vivo. Furthermore, microarray profiling indicated that HOXA-AS2 serves as an endogenous sponge by directly binding to miR-520c-3p and down-regulating miR-520c-3p expression. We demonstrated that HOXA-AS2 controls the expression of miR-520c-3p target genes, TGFBR2 and RELA, in breast cancer cells. Therefore, our study may provide a better understanding of the pathogenesis of breast cancer and suggests that HOXA-AS2 may be a potential prognostic and therapeutic target in breast cancer.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Aged , Breast Neoplasms/pathology , Carcinogenesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
17.
Asian Pac J Trop Med ; 9(9): 898-902, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27633306

ABSTRACT

OBJECTIVE: To investigate the effects of co-transfection of miR-520c-3p and miR-132 on proliferation and apoptosis of hepatocellular carcinoma Huh7. METHODS: Hepatocellular carcinoma Huh7 was cultured in vitro and lipidosome was used to transfect miR-520c-3p and miR-132, respectively or together. The effects of transfection of miR-520c-3p and miR-132 on proliferation and apoptosis of Huh7 were detected by CCK8 and Annexin V staining and flow cytometry, and the expression level of the targeted gene of over-expressed miR-520c-3p and miR-132 was determined by Western blot and realtime PCR. RESULTS: Compared with the control group, the proliferation ability of Huh7 of the single transfected and co-transfected miR-520c-3p and miR-132 decreased significantly, and the apoptosis ratio increased distinctly (P < 0.05). Besides, the effect of the co-transfection group was better than that of the single transfection group. The protein levels of GPC3 (Glypican-3) and YAP (Yes-associated protein), the target genes transfected only by miR-520c-3p and miR-132, respectively, reduced obviously (P < 0.05), which was similar with the co-infected cells, but cells transfected by miR-132 only showed a decrease of YAP. CONCLUSIONS: The co-transfection of miR-520c-3p and miR-132 can target-regulate the expression of GPC3 and YAP, enhance the exhibition effect on proliferation of hepatocellular carcinoma Huh7 and induce cell apoptosis synergistically.

18.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-951333

ABSTRACT

Objective To investigate the effects of co-transfection of miR-520c-3p and miR-132 on proliferation and apoptosis of hepatocellular carcinoma Huh7. Methods Hepatocellular carcinoma Huh7 was cultured in vitro and lipidosome was used to transfect miR-520c-3p and miR-132, respectively or together. The effects of transfection of miR-520c-3p and miR-132 on proliferation and apoptosis of Huh7 were detected by CCK8 and Annexin V staining and flow cytometry, and the expression level of the targeted gene of over-expressed miR-520c-3p and miR-132 was determined by Western blot and realtime PCR. Results Compared with the control group, the proliferation ability of Huh7 of the single transfected and co-transfected miR-520c-3p and miR-132 decreased significantly, and the apoptosis ratio increased distinctly (P < 0.05). Besides, the effect of the co-transfection group was better than that of the single transfection group. The protein levels of GPC3 (Glypican-3) and YAP (Yes-associated protein), the target genes transfected only by miR-520c-3p and miR-132, respectively, reduced obviously (P < 0.05), which was similar with the co-infected cells, but cells transfected by miR-132 only showed a decrease of YAP. Conclusions The co-transfection of miR-520c-3p and miR-132 can target-regulate the expression of GPC3 and YAP, enhance the exhibition effect on proliferation of hepatocellular carcinoma Huh7 and induce cell apoptosis synergistically.

19.
Article in English | WPRIM (Western Pacific) | ID: wpr-819895

ABSTRACT

OBJECTIVE@#To investigate the effects of co-transfection of miR-520c-3p and miR-132 on proliferation and apoptosis of hepatocellular carcinoma Huh7.@*METHODS@#Hepatocellular carcinoma Huh7 was cultured in vitro and lipidosome was used to transfect miR-520c-3p and miR-132, respectively or together. The effects of transfection of miR-520c-3p and miR-132 on proliferation and apoptosis of Huh7 were detected by CCK8 and Annexin V staining and flow cytometry, and the expression level of the targeted gene of over-expressed miR-520c-3p and miR-132 was determined by Western blot and realtime PCR.@*RESULTS@#Compared with the control group, the proliferation ability of Huh7 of the single transfected and co-transfected miR-520c-3p and miR-132 decreased significantly, and the apoptosis ratio increased distinctly (P < 0.05). Besides, the effect of the co-transfection group was better than that of the single transfection group. The protein levels of GPC3 (Glypican-3) and YAP (Yes-associated protein), the target genes transfected only by miR-520c-3p and miR-132, respectively, reduced obviously (P < 0.05), which was similar with the co-infected cells, but cells transfected by miR-132 only showed a decrease of YAP.@*CONCLUSIONS@#The co-transfection of miR-520c-3p and miR-132 can target-regulate the expression of GPC3 and YAP, enhance the exhibition effect on proliferation of hepatocellular carcinoma Huh7 and induce cell apoptosis synergistically.

20.
Hepatol Res ; 44(3): 338-48, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23607462

ABSTRACT

AIM: Glypican-3 (GPC3) is a membrane-associated heparan sulfate proteoglycan involved in regulation of cell proliferation, cell survival, cell migration and differentiation process. MicroRNAs (miRNAs) are single-stranded, non-coding functional RNAs that are important in many biological processes. GPC3 and miRNAs have been found to play essential roles in the development and progression of hepatocellular carcinoma (HCC). However, little information about the relationship between GPC3 and miRNAs is available nowadays. Therefore, this study aims to examine the relationship between GPC3 and miRNAs. METHODS: Dual-luciferase reporter assay was used to validate the direct target of GPC3. Fluorescence quantitative PCR and Western blotting were used to examined the gene expression at mRNA and protein levels. Cell apoptosis was evaluated by flow cytometric analysis and Annexin V-FITC staining. Invasion of cells was evaluated by Transwell matrigel assay. RESULTS: The results showed that miR-520c-3p could specifically target GPC3 in HCC cells. GPC3 protein levels decreased with unchanged transcription efficiency after miRNA transfection, and there was negative correlation of miR-520c-3p expression in HCC in relate to GPC3 protein levels. Moreover, miR-520c-3p not only induced HCC cell apoptosis, but also inhibited the growth and invasion of the cells. Interestingly, overexpression of GPC3 could effectively reverse apoptosis induced by miR-520c-3p transfection in HCC. CONCLUSIONS: Taken together, these results supported that miR-520c-3p may decrease GPC3 protein levels to inhibit proliferation of HCC cells. Therefore, GPC3 could be a new target for genetic diagnosis and treatment of HCC.

SELECTION OF CITATIONS
SEARCH DETAIL
...