1
|
Xu L, Liu S, Yang Y, Shu L, Sun Y. LINC00313 suppresses autophagy and promotes stemness of nasopharyngeal carcinoma cells through PTBP1/STIM1 axis. Radiother Oncol 2024; 196:110310. [PMID: 38677328 DOI: 10.1016/j.radonc.2024.110310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a kind of malignant head and neck tumor with high mortality. lncRNAs are valuable diagnostic biomarkers and therapeutic targets for various tumors. This study investigated the effects and mechanism of LINC00313 in nasopharyngeal carcinoma. METHODS Cell Counting Kit-8 (CCK-8) and immunohistochemistry were used for assessing cell proliferation. The levels of autophagy-related proteins, and stem cell markers were detected. Immunofluorescence assay was used for LC3 detection. Methylated RNA Immunoprecipitation (meRIP) of LINC00313 in NPC cells was assessed. The localization of LINC00313 was verified by luorescence in situ hybridization (FIHS). The interaction between LINC00313 and the downstream targets were analyzed and confirmed by immunoprecipitation (RIP). Besides, the tumorigenesis roles of LINC00313 were confirmed in tumor growth mice model. RESULTS LINC00313 was increased in NPC tissues and cells. LINC00313 knockdown enhanced autophagy, and decreased stemness and cell viability of NPC cells through regulating STIM1. METTL3/IGF2BP1-mediated m6A modification promoted the stabilization and up-regulation of LINC00313. LINC00313 activated AKT/mTOR pathway in NPC cells through PTBP1/STIM1 axis. Moreover, LINC00313 promoted tumor growth and metastasis in xenograft model. CONCLUSION Upregulation of LINC00313 suppressed autophagy and promoted stemness of NPC cells through PTBP1/STIM1 axis.
Collapse
Affiliation(s)
- Lina Xu
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, PR China
| | - Sile Liu
- Department of Laboratory Medicine, Hunan Provincial People's Hospital, Changsha 410005, Hunan Province, PR China
| | - Yang Yang
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, PR China
| | - Lu Shu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, PR China
| | - Yi Sun
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, PR China.
| |
Collapse
|
2
|
Liu J, Shang G. The Roles of Noncoding RNAs in the Development of Osteosarcoma Stem Cells and Potential Therapeutic Targets. Front Cell Dev Biol 2022; 10:773038. [PMID: 35252166 PMCID: PMC8888953 DOI: 10.3389/fcell.2022.773038] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma (OS) is the common bone tumor in children and adolescents. Because of chemotherapy resistance, the OS patients have a poor prognosis. The one reason of chemotherapeutic resistance is the development of cancer stem cells (CSCs). CSCs represent a small portion of tumor cells with the capacity of self-renewal and multipotency, which are associated with tumor initiation, metastasis, recurrence and drug resistance. Recently, noncoding RNAs (ncRNAs) have been reported to critically regulate CSCs. Therefore, in this review article, we described the role of ncRNAs, especially miRNAs, lncRNAs and circRNAs, in regulating CSCs development and potential mechanisms. Specifically, we discussed the role of multiple miRNAs in targeting CSCs, including miR-26a, miR-29b, miR-34a, miR-133a, miR-143, miR-335, miR-382, miR-499a, miR-1247, and let-7days. Moreover, we highlighted the functions of lncRNAs in regulating CSCs in OS, such as B4GALT1-AS1, DANCR, DLX6-AS1, FER1L4, HIF2PUT, LINK-A, MALAT1, SOX2-OT, and THOR. Due to the critical roles of ncRNAs in regulation of OS CSCs, targeting ncRNAs might be a novel strategy for eliminating CSCs for OS therapy.
Collapse
Affiliation(s)
- Jinxin Liu
- Department of Orthopedic Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Guanning Shang
- Department of Orthopedic Surgery, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
3
|
Almeida TC, Seibert JB, Amparo TR, de Souza GHB, da Silva GN, Dos Santos DH. Modulation of Long Non-Coding RNAs by Different Classes of Secondary Metabolites from Plants: A Mini-Review on Antitumor Effects. Mini Rev Med Chem 2021; 22:1232-1255. [PMID: 34720079 DOI: 10.2174/1389557521666211101161548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/07/2021] [Accepted: 09/10/2021] [Indexed: 11/22/2022]
Abstract
The broad pharmacological spectrum of plants is related to their secondary metabolism, which is responsible for the synthesis of different compounds that have multiple effects on cellular physiology. Among the biological effects presented by phytochemicals, their use for the prevention and treatment of cancer can be highlighted. This occurs due to several mechanisms of antitumor action demonstrated by these compounds, including regulation of the cell signaling pathways and inhibition of tumor growth. In this way, long non-coding RNAs (lncRNAs) appear to be promising targets for the treatment of cancer. Their deregulation has already been related to a variety of clinical-pathological parameters. However, the effects of secondary metabolites on lncRNAs are still restricted. For this reason, the present review aimed to gather data on phytochemicals with action on lncRNAs in order to confirm their possible antitumor potential. According to the literature, terpenoid and flavonoid are the main examples of secondary metabolites involved with lncRNAs activity. In addition, the lncRNAs H19, CASC2, HOTAIR, NKILA, CCAT1, MALAT1, AFAP1-AS1, MEG3, and CDKN2B-AS1 can be highlighted as important targets in the search for new anti-tumor agents since they act as modulating pathways related to cell proliferation, cell cycle, apoptosis, cell migration and invasion. Finally, challenges for the use of natural products as a commercial drug were also discussed. The low yield, selectivity index and undesirable pharmacokinetic parameters were emphasized as a difficulty for obtaining these compounds on a large scale and for improving the potency of its biological effect. However, the synthesis and/or development of formulations were suggested as a possible approach to solve these problems. All of these data together confirm the potential of secondary metabolites as a source of new anti-tumor agents acting on lncRNAs.
Collapse
Affiliation(s)
- Tamires Cunha Almeida
- Department of Pharmacy, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto. Brazil
| | | | - Tatiane Roquete Amparo
- Department of Pharmacy, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto. Brazil
| | | | - Glenda Nicioli da Silva
- Department of Clinical Analysis, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto. Brazil
| | | |
Collapse
|
4
|
Chen X, Liu H. Alisol A Inhibited the Proliferation, Migration, and Invasion of Nasopharyngeal Carcinoma Cells by Inhibiting the Hippo Signaling Pathway. Yonsei Med J 2021; 62:895-902. [PMID: 34558868 PMCID: PMC8470560 DOI: 10.3349/ymj.2021.62.10.895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 12/09/2022] Open
Abstract
PURPOSE Alisol A is a bioactive triterpenoid isolated from the Rhizoma Alismatis. Previous studies have shown that alisol A has anticancer potential. In this study, we explored the effect of alisol A on the growth of nasopharyngeal carcinoma (NPC) cells. MATERIALS AND METHODS MTT assay, colony formation assay, flow cytometry, transwell assay, wound healing assay, and western blotting were used to assess cell viability, proliferation, cell cycle, migration, invasion, and protein expression, respectively, in vitro. AutoDock Vina and Discovery Studio software were used for molecular docking. RESULTS Alisol A inhibited the viability, proliferation, migration, and invasion of NPC cells. The molecular docking simulation assay confirmed that alisol A bound to YAP protein. In addition, alisol A promoted the phosphorylation of YAP and suppressed the expression of YAP in NPC cells. CONCLUSION Alisol A inhibited the proliferation, migration, and invasion of NPC cells by inhibiting the Hippo signaling pathway. Alisol A may be a candidate drug for NPC.
Collapse
Affiliation(s)
- Xianghong Chen
- Department of Otolaryngology, Affiliated Hospital of Hebei University, Hebei, China
| | - Huiqing Liu
- Department of Otolaryngology, Affiliated Hospital of Hebei University, Hebei, China.
| |
Collapse
|
5
|
Dong W, Dong Q, Ding H. Ophiopogonin B induces reactive oxygen species‑dependent apoptosis through the Hippo pathway in nasopharyngeal carcinoma. Mol Med Rep 2021; 24:534. [PMID: 34080657 PMCID: PMC8170196 DOI: 10.3892/mmr.2021.12173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/15/2021] [Indexed: 12/15/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common malignant tumor in South China and is characterized by a high death rate. Ophiopogonin B (OP‑B) is a bioactive component of Radix Ophiopogon japonicus, which is frequently used in traditional Chinese medicine to treat cancer. The present study aimed to examine the anti‑cancer properties of OP‑B on NPC cells. Cell viability and cell proliferation were measured using MTT and EdU assays. Flow cytometry was used to measure cell apoptosis, reactive oxygen species and mitochondrial membrane potential. Western blotting was used to investigate the expression of apoptosis and Hippo signaling pathway proteins. OP‑B inhibited the proliferation of NPC cells by inducing apoptosis and disturbing the mitochondrial integrity. OP‑B enhanced ROS accumulation. In addition, OP‑B promoted the expression of mammalian STE20‑like kinase 1, large tumor suppressor 1 and phosphorylated yes‑associated protein (YAP) and suppressed the expression of YAP and transcriptional enhanced associate domain in NPC cells. OP‑B increased the expression of forkhead box transcription factor O1 in the nuclear fraction. In conclusion, OP‑B has therapeutic potential and feasibility in the development of novel YAP inhibitors for NPC.
Collapse
Affiliation(s)
- Wenhui Dong
- Department of Otorhinolaryngology, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Qing Dong
- Department of Operating Room, Weifang Yidu Central Hospital, Weifang, Shandong 262500, P.R. China
| | - Hairui Ding
- Department of Emergency, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
6
|
Wu X, Liu Y, Ji Y. Carboxymethylated chitosan alleviated oxidative stress injury in retinal ganglion cells via IncRNA-THOR/IGF2BP1 axis. Genes Genomics 2021; 43:643-651. [PMID: 33811613 DOI: 10.1007/s13258-021-01085-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Glaucoma is an advanced nerve disorder described by the deterioration of axon and RGCs. CMCS has been previously used as an anti-apoptotic and anti-oxidant agent. OBJECTIVE The current study aimed to explore the protective impact of CMCS against H2O2-induced injury in glaucoma in vitro. METHODS The relative expression of lncRNA THOR and the protein expression of IGF2BP1 in H2O2-induced RGC-5 cells were detected by RT-PCR and western blot methods respectively. The cell viability was measured using MTT assay while apoptosis rate was measured by flow cytometry. Moreover, ROS level was measured using ROS assay kit. Furthermore, the relations between THOR and IGF2BP1 were determined by using RNA pull-down. RESULTS The expression of THOR was reduced in H2O2-induced RGCs. Also, RGCs viability was inhibited while the level of ROS and cell apoptosis were enhanced. CMCS treatment considerably enhanced the expression of THOR and IGF2BP1 protein and cell viability but reduced ROS level and cell apoptosis. Moreover, IGF2BP1 protein was positively regulated by lncRNA THOR. CMCS protected the RGCs from oxidative stress via regulating lncRNA THOR/IGF2BP1. CONCLUSION CMCS enhanced the cell viability and reduced the cell apoptosis and ROS level and protected RGCs from oxidative stress via lncRNATHOR/IGF2BP1 pathway, potentially suggesting a new therapeutic strategy for the treatment of glaucoma.
Collapse
Affiliation(s)
- Xiaoli Wu
- Ophthalmology Department, Shandong Rongjun General Hospital, Jinan, Shandong Province, China
| | - Yingying Liu
- Neurology Department, Shandong Rongjun General Hospital, Jinan, Shandong Province, China
| | - Yun Ji
- Yantai Laiyang Central Hospital, Yantai, Shandong, China.
| |
Collapse
|
7
|
Tang Y, He X. Long non-coding RNAs in nasopharyngeal carcinoma: biological functions and clinical applications. Mol Cell Biochem 2021; 476:3537-3550. [PMID: 33999333 DOI: 10.1007/s11010-021-04176-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common head and neck malignancies. It has obvious ethnic and regional specificity. Long non-coding RNAs (LncRNAs) are a class of non-protein coding RNA molecules. Emerging research shows that lncRNAs play a key role in tumor development, prognosis, and treatment. With the deepening of sequence analysis, a large number of functional LncRNAs have been found in NPC, which interact with coding genes, miRNAs, and proteins to form a complex regulatory network. However, the specific role and mechanism of abnormally expressed lncRNAs in the pathogenesis of NPC is not fully understood. This article briefly introduced the concept, classification, and functional mechanism of lncRNAs and reviewed their biological functions and their clinical applications in NPC. Specifically, we described lncRNAs related to the occurrence, growth, invasion, metastasis, angiogenesis, and cancer stem cells of NPC; discussed lncRNAs related to Epstein-Barr virus infection; and summarized the role of lncRNAs in NPC treatment resistance. We have also sorted out lncRNAs related to Chinese medicine treatment. We believe that with the deepening of lncRNAs research, tumor-specific lncRNAs may become a new target for the treatment and a biomarker for predicting prognosis.
Collapse
Affiliation(s)
- Yao Tang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology (2016TP1015), Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, 421001, Hunan Province, China
| | - Xiusheng He
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology (2016TP1015), Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|
8
|
Yuan F, Lou Z, Zhou Z, Yan X. Long non‑coding RNA KCNQ1OT1 promotes nasopharyngeal carcinoma cell cisplatin resistance via the miR‑454/USP47 axis. Int J Mol Med 2021; 47:54. [PMID: 33576460 PMCID: PMC7895519 DOI: 10.3892/ijmm.2021.4887] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/11/2021] [Indexed: 12/17/2022] Open
Abstract
Long non‑coding RNAs serve an essential role in drug resistance in various types of cancer, including lung, breast and bladder cancer. The present study aimed to investigate whether KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) was associated with cisplatin (DDP) resistance in nasopharyngeal carcinoma (NPC). KCNQ1OT1, microRNA (miR)‑454 and ubiquitin specific peptidase 47 (USP47) expression levels were measured via reverse transcription‑quantitative PCR. 5‑8F/DDP and SUNE‑1/DDP cell viability and chemosensitivity were assessed by performing Cell Counting Kit‑8 assays. Colony forming and Transwell assays were conducted to assess the effect of the KCNQ1OT1/miR‑454/USP47 axis on DDP resistance in NPC cells. The association between miR‑454 and KCNQ1OT1 or USP47 was verified via bioinformatics analysis, dual‑luciferase reporter assays and RIP assays. KCNQ1OT1 and USP47 expression levels were significantly upregulated, whereas miR‑454 expression levels were significantly downregulated in DDP‑resistant NPC cells compared with parental NPC cells. KCNQ1OT1 knockdown promoted chemosensitivity in DDP‑resistant NPC cells (5‑8F/DDP and SUNE‑1/DDP), as indicated by significantly decreased cell proliferation, migration and invasion in the short hairpin RNA (sh)KCNQ1OT1 group compared with the sh‑negative control (NC) group. Moreover, miR‑454 was identified as a target of KCNQ1OT1. KCNQ1OT1 overexpression significantly reversed miR‑454 overexpression‑mediated effects on NPC cell viability and DDP resistance. Furthermore, the results indicated that miR‑454 directly targeted USP47. Compared with the shNC group, USP47 knockdown significantly suppressed NPC cell viability and DDP resistance, which was significantly reversed by co‑transfection with miR‑454 inhibitor. Furthermore, compared with the shNC group, KCNQ1OT1 knockdown significantly downregulated USP47 expression, which was significantly counteracted by miR‑454 knockdown. Collectively, the results of the present study indicated that KCNQ1OT1 enhanced DDP resistance in NPC cells via the miR‑454/USP47 axis, suggesting a potential therapeutic target for patients with DDP‑resistant NPC.
Collapse
Affiliation(s)
- Feng Yuan
- Department of Otolaryngology, Zhuji Hospital Affiliated to Shaoxing College of Arts and Sciences, Zhuji, Zhejiang 311800, P.R. China
| | - Zhiping Lou
- Department of Otolaryngology, Zhuji Hospital Affiliated to Shaoxing College of Arts and Sciences, Zhuji, Zhejiang 311800, P.R. China
| | - Zhifeng Zhou
- Department of Otolaryngology, Zhuji Central Hospital, Zhuji, Zhejiang 311800, P.R. China
| | - Xiaojun Yan
- Department of Otorhinolaryngology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
9
|
Liao B, Yi Y, Zeng L, Wang Z, Zhu X, Liu J, Xie B, Liu Y. LINC00667 Sponges miR-4319 to Promote the Development of Nasopharyngeal Carcinoma by Increasing FOXQ1 Expression. Front Oncol 2021; 10:632813. [PMID: 33569351 PMCID: PMC7868543 DOI: 10.3389/fonc.2020.632813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/07/2020] [Indexed: 01/08/2023] Open
Abstract
Accumulating evidence has indicated that lncRNAs regulate various biological and pathological processes in diverse malignant tumors. The roles of LINC00667 in cancer development have been explored in glioma, hepatocellular carcinoma and non-small cell lung cancer, but not in nasopharyngeal carcinoma (NPC). In the present study, we characterize the role and molecular mechanism of LINC00667 in NPC progression. It was found that LINC00667 was overexpressed in NPC cells compared to normal cells. Silencing LINC00667 suppressed the proliferation, migration, invasion and epithelial mesenchymal transition (EMT) in NPC cells. In addition, bioinformatics analysis revealed that LINC00667 acted as a ceRNA to absorb miR-4319. Further investigations illustrated that miR-4319 had low expression in NPC cells and functioned as a tumor suppressor in the progression of NPC. Mechanistic study identified forkhead box Q1 (FOXQ1) as a functional target of miR-4319. The effect of LINC00667 in NPC development was mediated by the miR-4319/FOXQ1 axis. Analysis on tumorxenograft mouse model demonstrated that knockdown of LINC00667 repressed NPC tumor growth in vivo and confirmed the in vitro results. Our present study suggested that LINC00667 promoted the malignant phenotypes of NPC cells by competitively binding to miR-4319 to up-regulate FOXQ1 expression. Our results reveled that LINC00667 could be a diagnostic and therapeutic target for NPC patients.
Collapse
Affiliation(s)
- Bing Liao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yun Yi
- Department of Gynaecological Oncology, Jiangxi Cancer Hospital, Nanchang, China
| | - Lei Zeng
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinhua Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianguo Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bingbin Xie
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuehui Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
10
|
Yang M, Zhao L, Hu X, Feng H, Kang X. Identification of Key mRNAs and lncRNAs Associated with the Effects of Anti-TWEAK on Osteosarcoma. Curr Bioinform 2021. [DOI: 10.2174/1574893615999200626191405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background:
Osteosarcoma (OS) is one of the most common primary malignant bone tumors in teenagers.
Emerging studies demonstrated TWEAK and Fn14 were involved in regulating cancer cell differentiation, proliferation,
apoptosis, migration and invasion.
Objective:
The present study identified differently expressed mRNAs and lncRNAs after
anti-TWEAK treatment in OS cells using GSE41828.
Methods:
We identified 922 up-regulated mRNAs, 863 downregulated mRNAs, 29 up-regulated lncRNAs, and 58 down-regulated lncRNAs after anti-TWEAK treatment in OS cells.
By constructing PPI networks, we identified several key proteins involved in anti-TWEAK treatment in OS cells, including
MYC, IL6, CD44, ITGAM, STAT1, CCL5, FN1, PTEN, SPP1, TOP2A, and NCAM1. By constructing lncRNAs coexpression networks, we identified several key lncRNAs, including LINC00623, LINC00944, PSMB8-AS1,
LOC101929787.
Result:
Bioinformatics analysis revealed DEGs after anti-TWEAK treatment in OS were involved in
regulating type I interferon signaling pathway, immune response related pathways, telomere organization, chromatin
silencing at rDNA, and DNA replication. Bioinformatics analysis revealed differently expressed lncRNAs after antiTWEAK treatment in OS were related to telomere organization, protein heterotetramerization, DNA replication, response
to hypoxia, TNF signaling pathway, PI3K-Akt signaling pathway, Focal adhesion, Apoptosis, NF-kappa B signaling
pathway, MAPK signaling pathway, FoxO signaling pathway.
Conclusion: :
This study provided useful information for
understanding the mechanisms of TWEAK underlying OS progression and identifying novel therapeutic markers for OS.
Collapse
Affiliation(s)
- Mingxuan Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou City, Gansu Province 730030,China
| | - Liangtao Zhao
- Tsing Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou City, Gansu Province 730030,China
| | - Xuchang Hu
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou City, Gansu Province 730030,China
| | - Haijun Feng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou City, Gansu Province 730030,China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou City, Gansu Province 730030,China
| |
Collapse
|
11
|
Zhao Q, Sun P, Qin S, Liu J. Acylglycerol kinase promotes the stemness of nasopharyngeal carcinoma cells by promoting β-catenin translocation to the nucleus through activating PI3K/Akt pathway. ENVIRONMENTAL TOXICOLOGY 2020; 35:1299-1307. [PMID: 32652857 DOI: 10.1002/tox.22994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/25/2020] [Accepted: 06/14/2020] [Indexed: 06/11/2023]
Abstract
Recent evidences show that acylglycerol kinase (AGK) expression is related to the occurrence and development of various human cancers. However, its roles in nasopharyngeal carcinoma (NPC) progression are still unclear. This work aims to explore the roles of AGK in NPC cell stemness. It was shown that AGK expression was higher in NPC tissues compared to the adjacent tissues. Online dataset analysis revealed that AGK expression was negatively correlated with the overall survival of NPC patients. Gain and loss of functional experiments demonstrated that AGK positively regulated the stemness of NPC cells, as evident by the change of the tumor sphere-formation ability, ALDH1 activity and expression of stemness critical regulators. KEGG analysis were performed to determine the potential pathways of AGK involved in NPC cell stemness and showed that the PI3K/Akt pathway exhibited the most correlation with AGK expression. Further mechanistic studies confirmed that AGK promoted the stemness of NPC cells through activating the PI3K/Akt pathway, and thus enhancing β-catenin accumulation in nucleus. This study demonstrates a novel AGK/PI3K/Akt/β-catenin axis involving in NPC cell stemness.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| | - Peng Sun
- Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| | - Songbing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| | - Jisheng Liu
- Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| |
Collapse
|
12
|
Fang M, Zhang M, Wang Y, Wei F, Wu J, Mou X, Zhang Y, Liang X, Tang J. Long Noncoding RNA AFAP1-AS1 Is a Critical Regulator of Nasopharyngeal Carcinoma Tumorigenicity. Front Oncol 2020; 10:601055. [PMID: 33330099 PMCID: PMC7719841 DOI: 10.3389/fonc.2020.601055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background The long noncoding RNA actin filament associated protein 1 antisense RNA1 (AFAP1-AS1) is a critical player in various cancers. However, the clinical value and functional mechanisms of AFAP1-AS1 during the tumorigenicity of nasopharyngeal carcinoma (NPC) remain unclear. Here, we investigated the clinical application and potential molecular mechanisms of AFAP1-AS1 in NPC tumorigenesis and progression. Methods The expression level of AFAP1-AS1 was determined by qRT-PCR in 10 paired fresh human NPC tissues and adjacent normal tissues. RNAscope was performed on 100 paired paraffin-embedded NPC and adjacent nontumor specimens. The biological functions of AFAP1-AS1 were assessed by in vitro and in vivo functional experiments. RNA-protein pull-down assays were performed to detect and identify the AFAP1-AS1-interacting protein KAT2B. Protein-RNA immunoprecipitation (RIP) assays were conducted to examine the interaction of AFAP1-AS1 and KAT2B. Chromatin immunoprecipitation (ChIP) and luciferase analyses were utilized to identify the binding site of transcription intermediary factor 1 alpha (TIF1α) and H3K14ac on the RBM3 promoter. Results AFAP1-AS1 is upregulated in NPC and is a poor prognostic indicator for survival in NPC patients. AFAP1-AS1 was required for NPC proliferation in vitro and tumorigenicity in vivo. Mechanistic investigations suggested that AFAP1-AS1 binds to KAT2B and promotes acetyltransferase activation at two residues (E570/D610). KAT2B further promotes H3K14 acetylation and protein binding to the bromo domain of TIF1α. Consequently, TIF1α acts as a nuclear transcriptional coactivator of RBM3 transcription, leading to YAP mRNA stabilization and enhanced NPC tumorigenicity. Conclusions Our findings suggest that AFAP1-AS1 functions as an oncogenic biomarker and promotes NPC tumorigenicity through enhanced KAT2B acetyltransferase activation and YAP mRNA stabilization.
Collapse
Affiliation(s)
- Min Fang
- Department of Radiation Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Minjun Zhang
- Department of Radiation Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Graduate Department, Bengbu Medical College, Bengbu, China
| | - Yiqing Wang
- The Reproductive Medicine Special Hospital of the First Hospital of Lanzhou University, Key Laboratory for Reproductive Medicine and Embryo, Lanzhou, China
| | - Fangqiang Wei
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jianhui Wu
- Department of the Otolaryngology, Zhongshan City People's Hospital, Zhongshan Affiliated Hospital of Sun Yan-sen University, Zhongshan, China
| | - Xiaozhou Mou
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yigan Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Xiaodong Liang
- Department of Radiation Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Graduate Department, Bengbu Medical College, Bengbu, China
| | - Jianming Tang
- Department of Radiation Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
13
|
Garofoli M, Volpicella M, Guida M, Porcelli L, Azzariti A. The Role of Non-Coding RNAs as Prognostic Factor, Predictor of Drug Response or Resistance and Pharmacological Targets, in the Cutaneous Squamous Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12092552. [PMID: 32911687 PMCID: PMC7565940 DOI: 10.3390/cancers12092552] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is the most common keratinocyte-derived skin cancer in the Caucasian population. Exposure to UV radiations (UVRs) represents the main risk carcinogenesis, causing a considerable accumulation of DNA damage in epidermal keratinocytes with an uncontrolled hyperproliferation and tumor development. The limited and rarely durable response of CSCC to the current therapeutic options has led researchers to look for new therapeutic strategies. Recently, the multi-omics approaches have contributed to the identification and prediction of the key role of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), circularRNAs (circRNAs) and long non-coding RNAs (lncRNAs) in the regulation of several cellular processes in different tumor types, including CSCC. ncRNAs can modulate transcriptional and post-transcriptional events by interacting either with each other or with DNA and proteins, such as transcription factors and RNA-binding proteins. In this review, the implication of ncRNAs in tumorigenesis and their potential role as diagnostic biomarkers and therapeutic targets in human CSCC are reported.
Collapse
Affiliation(s)
- Marianna Garofoli
- Experimental Pharmacology Laboratory, IRCCS IstitutoTumori Giovanni Paolo II, 70124 Bari, Italy; (M.G.); (L.P.)
| | - Mariateresa Volpicella
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy;
| | - Michele Guida
- Rare Tumors and Melanoma Unit, IRCCS IstitutoTumori Giovanni Paolo II, 70124 Bari, Italy;
| | - Letizia Porcelli
- Experimental Pharmacology Laboratory, IRCCS IstitutoTumori Giovanni Paolo II, 70124 Bari, Italy; (M.G.); (L.P.)
| | - Amalia Azzariti
- Experimental Pharmacology Laboratory, IRCCS IstitutoTumori Giovanni Paolo II, 70124 Bari, Italy; (M.G.); (L.P.)
- Correspondence: ; Tel.: +39-080-555-5986
| |
Collapse
|
14
|
Huang D, Zhu X, Wang Y, Yu H, Pu Y. Long non-coding RNA FAM133B-2 represses the radio-resistance of nasopharyngeal cancer cells by targeting miR-34a-5p/CDK6 axis. Aging (Albany NY) 2020; 12:16936-16950. [PMID: 32889799 PMCID: PMC7521541 DOI: 10.18632/aging.103600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/13/2020] [Indexed: 01/24/2023]
Abstract
Long non-coding RNAs (lncRNAs) were found to play roles in various cancers, including nasopharyngeal carcinoma. In this study, we focused on the biological function of the lncRNA FAM133B-2 in the radio-resistance of nasopharyngeal carcinoma. The RNA-seq and qRT-PCR analysis showed that FAM133B-2 is highly expressed in the radio-resistant nasopharyngeal carcinoma cells. The following biochemical assays showed that FAM133B-2 represses the nasopharyngeal carcinoma radio-resistance and also affects the apoptosis and proliferation of nasopharyngeal carcinoma cells. Further investigations suggested that miR-34a-5p targets FAM133B-2 and also regulates the cyclin-dependent kinase 6 (CDK6). All these results suggested that the lncRNA FAM133B-2 might function as a competitive endogenous RNA (ceRNA) for miR-34a-5p in nasopharyngeal carcinoma radio-resistance, thus it may be regarded as a novel prognostic biomarker and therapeutic target in nasopharyngeal carcinoma diagnosis and treatment.
Collapse
Affiliation(s)
- Dabing Huang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, P.R. China
| | - Xianhai Zhu
- Department of Interventional Oncology, Anhui Provincial Cancer Hospital, West Branch of the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, P.R. China
| | - Yong Wang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, P.R. China
| | - Haobin Yu
- Department of Cancer Nutrition and Metabolic Therapy, No.3 Ward of Oncology, Anhui Provincial Cancer Hospital, West Branch of the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, P.R. China
| | - Youguang Pu
- Department of Cancer Epigenetics Program, Anhui Provincial Cancer Hospital, West Branch of the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, P.R. China
| |
Collapse
|
15
|
Liu C, Wu Y, Ma J. Interaction of non-coding RNAs and Hippo signaling: Implications for tumorigenesis. Cancer Lett 2020; 493:207-216. [PMID: 32822816 DOI: 10.1016/j.canlet.2020.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Hippo signaling is an evolutionarily conserved pathway that controls organ size by regulating cell proliferation, apoptosis, and stem cell self-renewal by "turning off" or "turning on" the kinase cascade chain reaction to manipulate the expression of downstream genes. Dysregulation of the Hippo pathway contributes to cancer development and metastasis. Emerging evidence has revealed new insights into tumorigenesis through the interplay between the Hippo pathway and non-coding RNAs (ncRNAs), especially microRNA, long non-coding RNA and circular RNA. Here, we reviewed the interactions between the Hippo pathway and ncRNAs and their implication for a variety of tumor-promoting or tumor-repressing effects. These interactions have the potential to serve as cancer biomarkers and therapeutic targets in clinical applications.
Collapse
Affiliation(s)
- Can Liu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yangge Wu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
16
|
Tu C, Yang K, Wan L, He J, Qi L, Wang W, Lu Q, Li Z. The crosstalk between lncRNAs and the Hippo signalling pathway in cancer progression. Cell Prolif 2020; 53:e12887. [PMID: 32779318 PMCID: PMC7507458 DOI: 10.1111/cpr.12887] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/23/2020] [Accepted: 07/18/2020] [Indexed: 12/11/2022] Open
Abstract
LncRNAs play a pivotal role in the regulation of epigenetic modification, cell cycle, differentiation, proliferation, migration and other physiological activities. In particular, considerable studies have shown that the aberrant expression and dysregulation of lncRNAs are widely implicated in cancer initiation and progression by acting as tumour promoters or suppressors. Hippo signalling pathway has attracted researchers’ attention as one of the critical cancer‐related pathways in recent years. Increasing evidences have demonstrated that lncRNAs could interact with Hippo cascade and thereby contribute to acquisition of multiple malignant hallmarks, including proliferation, metastasis, relapse and resistance to anti‐cancer treatment. Specifically, Hippo signalling pathway is reported to modulate or be regulated by widespread lncRNAs. Intriguingly, certain lncRNAs could form a reciprocal feedback loop with Hippo signalling. More speculatively, lncRNAs related to Hippo pathway have been poised to become important putative biomarkers and therapeutic targets in human cancers. Herein, this review focuses on the crosstalk between lncRNAs and Hippo pathway in carcinogenesis, summarizes the comprehensive role of Hippo‐related lncRNAs in tumour progression and depicts their clinical diagnostic, prognostic or therapeutic potentials in tumours.
Collapse
Affiliation(s)
- Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Kexin Yang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Lu Wan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jieyu He
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wanchun Wang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiong Lu
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Li F, Song X, Li X, Zhang X, Feng X, Wang L, Xu L, Luo J, Zhu B, Ren W, Yu H, Yu Y. Lgr5 maintains stemness and regulates cell property in nasopharyngeal carcinoma through Wnt/β-catenin signaling pathway. Stem Cell Res 2020; 47:101916. [PMID: 32721896 DOI: 10.1016/j.scr.2020.101916] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/28/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common malignant tumor in Southern China and Southeast Asia. In this study, we found that Leucine rich repeat containing G protein-coupled receptor 5 (Lgr5) was highly expressed in NPC tissues and marked NPC stem cells. Lgr5high tumors showed differential transcriptional landscape compared to Lgr5not high tumors. Lgr5 expression was associated with the clinicopathologic features in NPC and was able to regulate the stemness and viability of NPC cell line CNE1 and HNE1. Meanwhile, the migration, invasion and epithelial-mesenchymal transition (EMT) was modulated by Lgr5 via Wnt/β-catenin signaling pathway. Furthermore, Lgr5 could regulate the sensitivity of NPC cells to chemotherapy drugs. Xenografted tumors from Lgr5-overexpressed CNE1 cells showed stronger tumor forming capacity and higher expression level of stem cell markers. Thus, we characterized previously unidentified role of Lgr5 in NPC cells, potential serving as a NPC stem cell biomarker and a therapeutic target against NPC.
Collapse
Affiliation(s)
- Fangqi Li
- School of Life Sciences, Shanghai University, Shanghai 200444 China
| | - Xiaole Song
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical, Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031 China
| | - Xuewen Li
- School of Life Sciences, Shanghai University, Shanghai 200444 China
| | - Xiujuan Zhang
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical, Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031 China
| | - Xiaoyu Feng
- School of Life Sciences, Shanghai University, Shanghai 200444 China
| | - Li Wang
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical, Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031 China
| | - Lun Xu
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical, Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031 China
| | - Jiqin Luo
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical, Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031 China
| | - Bijun Zhu
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical, Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031 China
| | - Wenwen Ren
- Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical, Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031 China.
| | - Hongmeng Yu
- School of Life Sciences, Shanghai University, Shanghai 200444 China.
| | - Yiqun Yu
- School of Life Sciences, Shanghai University, Shanghai 200444 China; Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical, Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031 China.
| |
Collapse
|
18
|
Chu F, Cui Y, Li K, Xiao X, Zhang L, Zhang L, Wang L, Gao L, Yin N, Wu H. Long noncoding RNA THOR is highly expressed in colorectal cancer and predicts a poor prognosis. Future Oncol 2020; 16:1911-1920. [PMID: 32615044 DOI: 10.2217/fon-2020-0393] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aim: Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide. This study aimed to investigate the role of long noncoding RNA THOR in CRC. Materials & methods: The expression of THOR in 103 cases of CRC tissues and four CRC cell lines was examined by quantitative real-time PCR. Cell counting kit-8 and colony formation assays were applied to detect cell proliferation, and flow cytometry was used for testing cell cycle and apoptosis of CRC. Results: We found that THOR was highly expressed in CRC and correlated with tumor node metastasis stage, histological subtype, tumor size and differentiation and survival in CRC patients. Meanwhile, knockdown of THOR significantly suppressed cell proliferation and cell cycle of CRC, whereas promoted cell apoptosis. Conclusion: Our findings suggest that THOR is an oncogenic long noncoding RNA in CRC and a potential prognostic biomarker for this cancer.
Collapse
Affiliation(s)
- Feifei Chu
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Yuanbo Cui
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Kunkun Li
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Xingguo Xiao
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Li Zhang
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Lei Zhang
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Lihong Wang
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Lei Gao
- Department of Gastrointestinal Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Ningwei Yin
- Department of Science & Education, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| | - Huili Wu
- Department of Gastroenterology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, PR China
| |
Collapse
|
19
|
Wu X, Wang Y, Zhong W, Cheng H, Tian Z. The Long Non-Coding RNA MALAT1 Enhances Ovarian Cancer Cell Stemness by Inhibiting YAP Translocation from Nucleus to Cytoplasm. Med Sci Monit 2020; 26:e922012. [PMID: 32433460 PMCID: PMC7254939 DOI: 10.12659/msm.922012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The purpose of this work was to unearth the effects and underlying mechanism of long non-coding RNA (lncRNA) MALAT1 in ovarian cancer cell stemness. MATERIAL AND METHODS Western blot, quantitative polymerase chain reaction (qPCR) and sphere forming analysis were performed to evaluate the stem-like traits of cells and MALAT1-induced effects on ovarian cancer cell stemness. Cell viability was performed to evaluate MALAT1 role in the chemoresistance of ovarian cancer cells. RNA immunoprecipitation (RIP) and luciferase reporter analysis were constructed to investigate the underlying mechanisms. RESULTS Here, qPCR assay showed that MALAT1 level was remarkably higher in non-adherent spheres formed by adherent ovarian cancer cells, as well as cisplatin-resistant ovarian cancer cells. Additionally, MALAT1 knockdown reduced ovarian cancer cell stemness, characterized as the decrease of sphere forming ability, expression of stemness regulatory masters, and attenuation of cisplatin resistance. Moreover, MALAT1 interacted with yes-associated protein (YAP), inhibited its nuclear-cytoplasm translocation, promoted YAP protein stability and expression and thus increased its activity. Notably, rescuing expression of YAP attenuated the inhibition of MALAT1 knockdown on ovarian cancer cell stemness. CONCLUSIONS In conclusion, these results demonstrate a MALAT1/YAP axis responsible for ovarian cancer cell stemness.
Collapse
Affiliation(s)
- XingMei Wu
- Department of Gynecology, The People's Hospital of Lishui, Lishui, Zhejiang, China (mainland)
| | - YongHui Wang
- Department of Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| | - WeiJuan Zhong
- Department of Gynecology, The People's Hospital of Lishui, Lishui, Zhejiang, China (mainland)
| | - HuiFei Cheng
- Department of Radiation Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| | - Zhifeng Tian
- Department of Radiation Oncology, Lishui Municipal Central Hospital, Lishui, Zhejiang, China (mainland)
| |
Collapse
|
20
|
Zhan Y, Fan S. Multiple Mechanisms Involving in Radioresistance of Nasopharyngeal Carcinoma. J Cancer 2020; 11:4193-4204. [PMID: 32368302 PMCID: PMC7196263 DOI: 10.7150/jca.39354] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 02/04/2020] [Indexed: 02/07/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is the malignant tumor with ethnic and geographical distribution preference. Although intensity-modulated radiotherapy (IMRT)-based radiotherapy combined with chemotherapy and targeted therapy has dramatically improved the overall survival of NPC patients, there are still some patients suffering from recurrent tumors and the prognosis is poor. Multiple mechanisms may be responsible for radioresistance of NPC, such as cancer stem cells (CSCs) existence, gene mutation or aberrant expression of genes, epigenetic modification of genes, abnormal activation of certain signaling pathways, alteration of tumor microenvironment, stress granules (SGs) formation, etc. We conduct a comprehensive review of the published literatures focusing on the causes of radioresistance, retrospect the regulation mechanisms following radiation, and discuss future directions of overcoming the resistance to radiation.
Collapse
Affiliation(s)
- Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
Wang Y, Sun B, Wen X, Hao D, Du D, He G, Jiang X. The Roles of lncRNA in Cutaneous Squamous Cell Carcinoma. Front Oncol 2020; 10:158. [PMID: 32185124 PMCID: PMC7059100 DOI: 10.3389/fonc.2020.00158] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/29/2020] [Indexed: 02/05/2023] Open
Abstract
Cutaneous squamous cell carcinoma derives from keratinocytes and is the second most common cause of non-melanoma skin cancer. Cutaneous squamous cell carcinoma (cSCC) develops rapidly and is also the leading cause of death in non-melanoma cancers. Lymph node metastasis occurs in 5% of cSCC patients, and some patients may even metastasize to the viscera. Patients with regional lymphatic metastasis or distant metastases have a <20% 10-year survival rate, indicating the substantial challenge in treating advanced and metastatic cSCC. Some lncRNAs have been found to be abnormally overexpressed in many tumor tissues, so that they can be considered as potential new biomarkers or targets that can be used in the diagnosis and treatment of cSCC in the future. In this review, we summarize the role of lncRNA in cutaneous squamous cell carcinoma to make a better understanding of mutations in cSCC and lay the foundation for effective target therapy of cSCC.
Collapse
Affiliation(s)
- Yujia Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Bensen Sun
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Wen
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Hao
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Du
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Gu He
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Lnc-THOR silencing inhibits human glioma cell survival by activating MAGEA6-AMPK signaling. Cell Death Dis 2019; 10:866. [PMID: 31727877 PMCID: PMC6856358 DOI: 10.1038/s41419-019-2093-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Long non-coding RNA THOR (Lnc-THOR) binds to IGF2BP1, essential for its function. We here show that Lnc-THOR is expressed in human glioma tissues and cells. Its expression is extremely low or even undetected in normal brain tissues, as well as in human neuronal cells and astrocytes. We show that Lnc-THOR directly binds to IGF2BP1 in established and primary human glioma cells. shRNA-mediated Lnc-THOR knockdown or CRISPR/Cas9-induced Lnc-THOR knockout potently inhibited cell survival and proliferation, while provoking glioma cell apoptosis. Contrarily, forced overexpression of Lnc-THOR promoted glioma cell growth and migration. Importantly, Lnc-THOR shRNA or knockout activated MAGEA6-AMPK signaling in glioma cells. AMPK inactivation, by AMPKα1 shRNA, knockout, or dominant-negative mutation (T172A), attenuated Lnc-THOR shRNA-induced A172 glioma cell apoptosis. Moreover, CRISPR/Cas9-induced IGF2BP1 knockout activated MAGEA6-AMPK signaling as well, causing A172 glioma cell apoptosis. Significantly, Lnc-THOR shRNA was ineffective in IGF2BP1 KO A172 cells. In vivo, Lnc-THOR silencing or knockout potently inhibited subcutaneous A172 xenograft tumor growth in mice. MAGEA6 downregulation and AMPK activation were detected in Lnc-THOR-silenced/-KO A172 tumor tissues. Taken together, Lnc-THOR depletion inhibits human glioma cell survival possibly by activating MAGEA6-AMPK signaling.
Collapse
|
23
|
Gao L, Sang JZ, Cao H. Limonin enhances the radiosensitivity of nasopharyngeal carcinoma cells via attenuating Stat3-induced cell stemness. Biomed Pharmacother 2019; 118:109366. [PMID: 31545261 DOI: 10.1016/j.biopha.2019.109366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 12/17/2022] Open
Abstract
The inhibitory effects of limonin have been disclosed in various tumors, however, its roles in nasopharyngeal carcinoma (NPC) progression are never been revealed. In the current work, we collected NPC cells with a higher stemness compared with bulk cells through isolating the side population (SP) cells. It was found that limonin exhibited a stronger inhibitory effect on SP cells than that in bulk cells, which was evident by a lower IC50 value. Additionally, limonin attenuated the stemness and migration ability of SP cells with the higher stemness, characterized as decreasing the spheroid formation ability, expression of stemness markers and migration ability. Moreover, the proportion of SP cells in G0 phase was remarkably higher than that in bulk cells. Notably, upon limonin treatment, the proportion of SP cells in G0 was decreased and S/G2/M increased. Furthermore, limonin enhanced the radiosensitivity of NPC cells. The mechanistic studies based on RNA-sequencing analysis revealed that limonin inhibited the gene transcription driven by Stat3 (signal transducer and activator of transcription 3) and an activator of Stat3 (Colivelin or IL-6) rescued the inhibitory effects of limonin. Therefore, these results demonstrate that limonin could reduce the stemness of NPC cells and thus the radiosensitivity through suppressing Stat3 transcriptional activity.
Collapse
Affiliation(s)
- Ling Gao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road No.1, Zhengzhou, 450052, China
| | - Jian-Zhong Sang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road No.1, Zhengzhou, 450052, China
| | - Hua Cao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road No.1, Zhengzhou, 450052, China.
| |
Collapse
|
24
|
Lv Y, Yang X, Wang L. Effect of lncRNA THOR on proliferation and migration of colon cancer cells. Oncol Lett 2019; 18:2518-2522. [PMID: 31452739 PMCID: PMC6676674 DOI: 10.3892/ol.2019.10585] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
The effect of long non-coding RNA (lncRNA) THOR on proliferation and migration of colon cancer cells was investigated. Lentiviral vector expressing lncRNA THOR shRNA was used to establish colon cancer SW620 lncRNA THOR knockdown cell line (experimental group), and at the same time, a control vector cell line (control group) was established by empty vector virus. Proliferation ability of the two groups was analyzed by CCK8 and EdU methods. Migration ability of the cells was analyzed by Transwell method. Xenograft tumor method was used to analyze the in vivo proliferation ability of the two groups of cells. mRNA levels of lncRNA THOR target genes were analyzed by reverse transcription-quantitative PCR (RT-qPCR). Compared with control cells, the cell proliferation ability of the experimental group was significantly decreased (P<0.05). Compared with the control group, the cell migration ability of the experimental group was significantly decreased (P<0.05). The tumor growth rate of the experimental group in the mice was significantly lower than that of the control group (P<0.05). Compared with the control group, mRNA levels of lncRNA THOR target genes IGF2BP1, SOX9 and c-myc in the experimental group were significantly downregulated (P<0.05). The results indicated that lncRNA THOR knockdown can significantly downregulate the expression of genes involved in tumor proliferation and migration, promote tumor cell proliferation and migration, indicating that lncRNA THOR plays an important role in colon cancer.
Collapse
Affiliation(s)
- Ying Lv
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Xiuhua Yang
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Lei Wang
- Department of Gastrointestinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
25
|
Yang H, Fu G, Liu F, Hu C, Lin J, Tan Z, Fu Y, Ji F, Cao M. LncRNA THOR promotes tongue squamous cell carcinomas by stabilizing IGF2BP1 downstream targets. Biochimie 2019; 165:9-18. [PMID: 31220513 DOI: 10.1016/j.biochi.2019.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 06/14/2019] [Indexed: 02/07/2023]
Abstract
THOR, a highly conserved lncRNA, is potentially involved in various cancer development. However, its involvement in tongue squamous cell carcinoma (TSCC) remains unclear. The present study aims to explore the biological function and molecular mechanism of THOR in TSCC progression. The expressions of THOR and IGF2BP1 in TSCC tissues and adjacent non-cancerous tongue tissues (ANT) were examined through qRT-PCR. THOR levels were manipulated in TSCC cells to explore its function in cancer progression in vitro and in vivo, which were subsequently evaluated by CCK8, colony formation assay, flow cytometry, xenograft tumor assays. In situ hybridization, RIP and Western blot assay were performed to explore the underlying molecular mechanisms. We discovered that THOR and IGF2BP1 were dramatically upregulated in TSCC tissues. The expression of THOR is positively correlated with IGF2BP1 mRNA level. THOR mediated IGF2 expression via interacting with IGF2BP1, and affected the downstream MEK-ERK signaling pathway to regulate TSCC cells proliferation. THOR/IGF2BP1/IGF2-MEK-ERK axis regulated the proliferation of TSCC cells, implying that THOR would be a promising therapeutic target for TSCC patients.
Collapse
Affiliation(s)
- Haojie Yang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ganglan Fu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Funing Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chuwen Hu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Junjie Lin
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zicong Tan
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yanni Fu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fengtao Ji
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Minghui Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
26
|
Jia X, Niu P, Xie C, Liu H. Long noncoding RNA PXN-AS1-L promotes the malignancy of nasopharyngeal carcinoma cells via upregulation of SAPCD2. Cancer Med 2019; 8:4278-4291. [PMID: 31173488 PMCID: PMC6675719 DOI: 10.1002/cam4.2227] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 04/02/2019] [Accepted: 04/23/2019] [Indexed: 01/19/2023] Open
Abstract
Accumulating evidences highlight the critical roles of long noncoding RNAs (lncRNAs) in a variety of cancers. LncRNA PXN‐AS1‐L was previously shown to exert oncogenic roles in hepatocellular carcinoma. However, the expression, role, and molecular mechanism of PXN‐AS1‐L in nasopharyngeal carcinoma (NPC) malignancy remain unknown. Here, we determined that PXN‐AS1‐L is upregulated in NPC tissues and cell lines. Increased expression of PXN‐AS1‐L predicts worse prognosis of NPC patients. PXN‐AS1‐L overexpression promotes NPC cell proliferation, migration, and invasion in vitro, and NPC tumor growth in vivo. PXN‐AS1‐L silencing suppresses NPC cell proliferation, migration, and invasion in vitro. Mechanistically, PXN‐AS1‐L directly interacts with SAPCD2 mRNA 3′‐untranslated region, prevents the binding of microRNAs‐AGO silencing complex to SAPCD2 mRNA, and upregulates the mRNA and protein level of SAPCD2. SAPCD2 is also increased in NPC tissues. The expression of SAPCD2 is significantly positively associated with that of PXN‐AS1‐L in NPC tissues. Gain‐of‐function and loss‐of‐function experiments demonstrated that SAPCD2 also promotes NPC cell proliferation, migration, and invasion. Furthermore, depletion of SAPCD2 significantly reverses the roles of PXN‐AS1‐L in promoting NPC cell proliferation, migration, and invasion in vitro, and NPC tumor growth in vivo. In conclusion, lncRNA PXN‐AS1‐L is upregulated in NPC and promoted NPC malignancy by upregulating SAPCD2 via direct RNA‐RNA interaction.
Collapse
Affiliation(s)
- Xiaodong Jia
- Department of Otolaryngology, Henan Province People's Hospital of Henan University, Zhengzhou, China
| | - Po Niu
- Department of Radiotherapy, Henan Province People's Hospital of Henan University, Zhengzhou, China
| | - Cuncun Xie
- Department of Otolaryngology, Henan Province People's Hospital of Henan University, Zhengzhou, China
| | - Hongjian Liu
- Department of Otolaryngology, Henan Province People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
27
|
Zhang E, Li X. LncRNA SOX2-OT regulates proliferation and metastasis of nasopharyngeal carcinoma cells through miR-146b-5p/HNRNPA2B1 pathway. J Cell Biochem 2019; 120:16575-16588. [PMID: 31099048 DOI: 10.1002/jcb.28917] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is an aggressive malignancy with a high mortality on account of its frequent metastasis and poor prognosis. An extensive body of investigations has proven that long noncoding RNAs are implicated in a variety of biological processes. Although SOX2-OT has been reported to play an oncogenic role in osteosarcoma, the mechanism of SOX2-OT-driven NPC progression is still obscure. The aim of this study was to elucidate the biological function of SOX2-OT and the related possible mechanism in NPC. In our study, SOX2-OT was notably elevated in NPC samples and cells. Further, a high expression level of SOX2-OT was correlated with poor clinical outcomes of NPC. Results from loss-of-function experiments suggested that knockdown of SOX2-OT repressed cell proliferation, arrested cell cycle, facilitated cell apoptosis, and inhibited cell metastasis of NPC. To further investigate the molecular mechanism of SOX2-OT, miR-146b-5p was found to directly bind to SOX2-OT, which mediated the role of SOX2-OT in NPC tumorigenesis. In addition, HNRNPA2B1 was a target of miR-146b-5p and SOX2-OT modulated the expression of HNRNPA2B1 through competitively binding to miR-146b-5p. At last, we discovered that SOX2-OT regulated NPC progression by targeting miR-146b-5p/HNRNPA2B1 pathway, which may provide more innovative targets for the treatment of patients with NPC.
Collapse
Affiliation(s)
- Enqin Zhang
- Department of Otorhinolaryngology, Ankang in Shaanxi Province Chinese Traditional Medicine Hospital, Ankang, Shaanxi, China
| | - Xueping Li
- Department of Otorhinolaryngology, Ankang in Shaanxi Province Chinese Central Medicine Hospital, Ankang, Shaanxi, China
| |
Collapse
|
28
|
Song H, Xu Y, Shi L, Xu T, Fan R, Cao M, Xu W, Song J. LncRNA THOR increases the stemness of gastric cancer cells via enhancing SOX9 mRNA stability. Biomed Pharmacother 2018; 108:338-346. [PMID: 30227327 DOI: 10.1016/j.biopha.2018.09.057] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 08/31/2018] [Accepted: 09/11/2018] [Indexed: 12/31/2022] Open
Abstract
This work aims to explore the roles and mechanisms of long non coding RNA (lncRNA) THOR in regulating the stemness of gastric cancer cells. RNA-sequencing combined with quantitative real-time PCR (qRT-PCR) indicated that lncRNA THOR level was significantly upregulated in gastric cancer tissues compared with that in normal adjacent tissues. Knockdown of THOR attenuated the stemnness of gastric cancer cells, evident by the decrease of stemness markers expression and capacity of cells spheroid formation. Further RNA-sequencing combined with qRT-PCR and western blot analysis demonstrated that expression of transcriptional factor SOX9 was remarkably decreased in gastric cancer cells with THOR stable knockdown. Additionally, RNA immunoprecipitation (RIP) combined with luciferase reporter assay revealed that THOR directly bound to SOX9 3' untranslated region (3'UTR), but not its 5'UTR or coding area. Notably, overexpression of SOX9 rescued THOR knockdown-mediated inhibition on the stemness of gastric cancer cells. Thus, our results suggest that THOR could potentiate the stemness of gastric cancer cells via directly binding to SOX9 3'UTR.
Collapse
Affiliation(s)
- Hu Song
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China
| | - Yixin Xu
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China
| | - Linseng Shi
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China
| | - Teng Xu
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China
| | - Ruizhi Fan
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China
| | - Meng Cao
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China
| | - Wei Xu
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China
| | - Jun Song
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu, 221002, PR China.
| |
Collapse
|