1
|
Rahman MS, Ghorai S, Panda K, Santiago MJ, Aggarwal S, Wang T, Rahman I, Chinnapaiyan S, Unwalla HJ. Dr. Jekyll or Mr. Hyde: The multifaceted roles of miR-145-5p in human health and disease. Noncoding RNA Res 2025; 11:22-37. [PMID: 39736851 PMCID: PMC11683234 DOI: 10.1016/j.ncrna.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/14/2024] [Accepted: 11/09/2024] [Indexed: 01/01/2025] Open
Abstract
MicroRNAs (miRNAs) are classified as small, non-coding RNAs that play crucial roles in diverse biological processes, including cellular development, differentiation, growth, and metabolism. MiRNAs regulate gene expression by recognizing complementary sequences within messenger RNA (mRNA) molecules. Recent studies have revealed that miR-145-5p functions as a tumor suppressor in several cancers, including lung, liver, and breast cancers. Notably, miR-145-5p plays a vital role in the pathophysiology underlying HIV and chronic obstructive pulmonary diseases associated with cigarette smoke. This miRNA is abundant in biofluids and shows potential as a biomarker for the diagnosis and prognosis of several infectious diseases, such as hepatitis B, tuberculosis, and influenza. Additionally, numerous studies have indicated that other non-coding RNAs, including long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), can regulate miR-145-5p. Given the significance of miR-145-5p, a comprehensive overview focusing on its roles in health and disease is essential. This review discusses the dual role of miR-145-5p as a protagonist and antagonist in important human diseases, with particular emphasis on disorders of the respiratory, digestive, nervous, reproductive, endocrine, and urinary systems.
Collapse
Affiliation(s)
- Md. Sohanur Rahman
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Suvankar Ghorai
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Kingshuk Panda
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Maria J. Santiago
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Saurabh Aggarwal
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Ting Wang
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Srinivasan Chinnapaiyan
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Hoshang J. Unwalla
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| |
Collapse
|
2
|
Durbas M. Expanding on roles of pleckstrin homology-like domain family A member 1 protein. Cell Tissue Res 2025; 399:9-25. [PMID: 39630301 PMCID: PMC11742907 DOI: 10.1007/s00441-024-03942-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/27/2024] [Indexed: 01/19/2025]
Abstract
Pleckstrin homology-like domain, family A, member 1 (PHLDA1), one of the three members of PHLDA (1-3) family, has been reported to be expressed in mammalian cells and tissues and play diverse roles in various biological processes such as apoptosis, pyroptosis, and differentiation. Nevertheless, new roles and mechanisms of PHLDA1 action have come to light, with some needing further clarification. The major aim of the publication is to review proapoptotic or antiapoptotic roles of PHLDA1 in cancer, including ample evidence on PHLDA1 role as a tumor suppressor gene or oncogene and its influence on tumor progression. The role of PHLDA1 as a prognostic marker of cancer emerges, as well as its role in drug response and resistance. PHLDA1 involvement in autophagy, endoplasmic reticulum stress, pyroptosis, or differentiation is also scrutinized. It is also important to note that the association of PHLDA1 with miRNA regulation is described. Additionally, the emerging functions of PHLDA1 are indicated, specifically in inflammation and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Małgorzata Durbas
- Laboratory of Molecular Genetics and Virology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
3
|
Xu W, Hu J, Ma Z, Feng W, Gong W, Fu S, Chen X. Decreased BIRC5-206 promotes epithelial-mesenchymal transition in nasopharyngeal carcinoma through sponging miR-145-5p. Open Med (Wars) 2024; 19:20241007. [PMID: 39308922 PMCID: PMC11416051 DOI: 10.1515/med-2024-1007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 09/25/2024] Open
Abstract
Metastasis significantly contributes to the poor prognosis of advanced nasopharyngeal carcinoma (NPC). Our prior studies have demonstrated a decrease in BIRC5-206 expression in NPC, which promotes disease progression. However, the role of BIRC5-206 in the invasion and metastasis of NPC has not been fully elucidated. In this study, our objective was to explore the biological function and underlying mechanisms of BIRC5-206 in NPC. Additionally, we established an NPC mouse model of lung invasiveness using C666 cells to assess the impact of BIRC5-206 on NPC metastasis. Our results revealed that silencing BIRC5-206 inhibited apoptosis and enhanced the invasion of NPC cells, whereas its overexpression reversed these effects. Moreover, decreased BIRC5-206 expression significantly increased N-cadherin and Vimentin expression while reducing E-cadherin and occludin levels, both in vivo and in vitro. Additionally, silencing BIRC5-206 markedly augmented the formation of invasive foci in lung tissues. Rescue experiments further confirmed that decreased BIRC5-206 expression facilitates NPC metastasis via modulation of the miR-145-5p/CD40 signaling pathway. In summary, our study suggests that BIRC5-206 may serve as a potential prognostic biomarker and therapeutic target in the diagnosis and treatment of NPC.
Collapse
Affiliation(s)
- Weihua Xu
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Junjie Hu
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Zhichao Ma
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, Haikou, Hainan, 570312, China
| | - Wanyi Feng
- Hainan Lvtou Medical Laboratory Center, Haikou, Hainan, 570206, China
- School of Life Sciences, Hainan University, Haikou, Hainan, 570228, China
| | - Wei Gong
- Hainan Lvtou Medical Laboratory Center, Haikou, Hainan, 570206, China
- School of Life Sciences, Hainan University, Haikou, Hainan, 570228, China
| | - Shengmiao Fu
- Central Laboratory, Hainan General Hospital, Hainan Hospital Affiliated to the Hainan Medical College, No. 19 Xiuhua Road, Xiuying District, Haikou, Hainan, 570311, China
- Hainan Lvtou Medical Laboratory Center, No. 16 Jinyu East Road, Longhua District, Haikou, Hainan, 570206, China
| | - Xinping Chen
- Department of Medical Laboratory, Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical University, Hainan Tropical Cancer Research Institute, No. 6, Changbin West 4th Street, Xiuying District, Haikou, Hainan, 570312, China
| |
Collapse
|
4
|
Salamanca E, Wu YF, Aung LM, Chiu BR, Chen MK, Chang WJ, Sun YS. Allylamine coating on zirconia dental implant surface promotes osteogenic differentiation in vitro and accelerates osseointegration in vivo. Clin Oral Implants Res 2024; 35:1101-1113. [PMID: 38804531 DOI: 10.1111/clr.14300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/19/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVES The glow discharge plasma (GDP) procedure has proven efficacy in grafting allylamine onto zirconia dental implant surfaces to enhance osseointegration. This study explored the enhancement of zirconia dental implant properties using GDP at different energy settings (25, 50, 75, 100, and 200 W) both in vitro and in vivo. MATERIALS AND METHODS In vitro analyses included scanning electron microscopy, wettability assessment, energy-dispersive X-ray spectroscopy, and more. In vivo experiments involved implanting zirconia dental implants into rabbit femurs and later evaluation through impact stability test, micro-CT, and histomorphometric measurements. RESULTS The results demonstrated that 25 and 50 W GDP allylamine grafting positively impacted MG-63 cell proliferation and increased alkaline phosphatase activity. Gene expression analysis revealed upregulation of OCN, OPG, and COL-I. Both 25 and 50 W GDP allylamine grafting significantly improved zirconia's surface properties (p < .05, p < .01, p < .001). However, only 25 W allylamine grafting with optimal energy settings promoted in vivo osseointegration and new bone formation while preventing bone level loss around the dental implant (p < .05, p < .01, p < .001). CONCLUSIONS This study presents a promising method for enhancing Zr dental implant surface's bioactivity.
Collapse
Affiliation(s)
- Eisner Salamanca
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Fan Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biomedical Engineering, Ming-Chuan University, Taoyuan, Taiwan
| | - Lwin Moe Aung
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Bor Rong Chiu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei Kuang Chen
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jen Chang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Dental Department, Taipei Medical University, Shuang-Ho Hospital, Taipei, Taiwan
| | - Ying Sui Sun
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
5
|
Zhang X, Liu J, Ji M, Qi G, Qiao R. Long Noncoding RNA GUSBP11 Knockdown Alleviates Nasopharyngeal Carcinoma via Regulating miR-1226-3p/ TM9SF4 Axis. Cancer Biother Radiopharm 2024; 39:133-143. [PMID: 35675666 DOI: 10.1089/cbr.2021.0391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose: Long noncoding RNAs (lncRNAs) have been confirmed related to the occurrence and progress of multiple cancers, including cervical cancer nasopharyngeal carcinoma (NPC). This study focused on assessing GUSBP11 effects on NPC progression and exploring possible mechanisms. Materials and Methods: RT-qPCR was conducted for assessing GUSBP11 levels within NPC tissues and cells. CCK-8, colony formation, and Transwell were adopted for examining GUSBP11 impacts on NPC cell proliferation and cell metastasis. RT-qPCR analysis and dual-luciferase reporter assay were conducted for judging the expression interrelation of GUSBP11 and its potential target miR-1226-3p. The same methods were carried out for verifying the inhibiting influences of miR-1226-3p upregulation and its potential target TM9SF4. Results: GUSBP11 levels were upregulated within NPC tissues and cells. GUSBP11 downregulation repressed NPC cell proliferation and cell metastasis. In addition, GUSBP11 targeted and negatively regulated miR-1226-3p. Furthermore, miR-1226-3p targeted TM9SF4 and mediated GUSBP11's impacts on TM9SF4 levels. At last, the authors proved the critical role of the GUSBP11/miR-1226-3p/TM9SF4 axis in regulating NPC progression. Conclusion: These findings indicate that downregulation of GUSBP11 alleviates NPC development by regulating the miR-1226-3p/TM9SF4 axis.
Collapse
Affiliation(s)
- Xiaofeng Zhang
- Department of Otolaryngology, Head and Neck Surgery, Weihai Maternal and Child Health Hospital, Affiliated Weihai Hospital of Qingdao University, Weihai, China
| | - Jinzhi Liu
- Department of the First Internal Medicine, Dongying District People's Hospital, Dongying, China
| | - MengMeng Ji
- Blood Purifying Center, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - GuiQin Qi
- Department of Outpatient, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Renling Qiao
- Department of Otolaryngology, Laiyang Central Hospital, Yantai, China
| |
Collapse
|
6
|
Wang J, Yang Y, Xu Y, Xu Z, Zhao X, Jia R, Dai Y. Long noncoding RNA XIST inhibition promotes Leydig cell apoptosis by acting as a competing endogenous RNA for microRNA-145a-5p that targets SIRT1 in late-onset hypogonadism. Cell Tissue Res 2024; 395:285-297. [PMID: 38353742 DOI: 10.1007/s00441-024-03860-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/04/2024] [Indexed: 03/01/2024]
Abstract
Leydig cell (LCs) apoptosis is responsible for decreased serum testosterone levels during late-onset hypogonadism (LOH). Our study was designed to illustrate the regulatory effect of lncRNA XIST on LCs and to clarify its molecular mechanism of action in LOH. The Leydig cells (TM3) was treated by 300 μM H2O2 for 8 h to establish Leydig cell oxidative stress model in vitro. The expression levels of lncRNA XIST in the testicular tissues of patients with LOH were measured using fluorescence in situ hybridization (FISH). The interaction between lncRNA XIST/SIRT1 and miR-145a-5p was assessed using starBase and dual-luciferase reporter gene assays. Apoptotic cells and Caspase3 activity were determined by flow cytometry (FCM) assay. Testosterone concentration was determined by ELISA. Moreover, histological assessment of testicles in mice was performed by using HE staining and the TUNEL assay was used to determine apoptosis. We found that the lncRNA XIST was downregulated in the testicular tissues of LOH patients and mice and in H2O2-induced TM3 cells. XIST siRNA significantly promoted apoptosis, enhanced Caspase3 activity and reduced testosterone levels in H2O2-stimulated TM3 cells. Further studies showed that the miR-145a-5p inhibitor reversed the effect of XIST-siRNA on H2O2-induced Leydig cell apoptosis. MiR-145a-5p negatively regulated SIRT1 expression, and SIRT1-siRNA reversed the effects of the miR-145a-5p inhibitor on H2O2 stimulated TM3 cells. The in vivo experiments indicated that silencing of the lncRNA XIST aggravated LOH symptoms in mice. Inhibition of lncRNA XIST induces Leydig cell apoptosis through the miR-145a-5p/SIRT1 axis in the progression of LOH.
Collapse
Affiliation(s)
- Jing Wang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210006, China
- Department of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing, 210008, China
| | - Yiqiong Yang
- Department of Emergency, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yang Xu
- Department of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing, 210008, China
| | - Zhipeng Xu
- Department of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing, 210008, China
| | - Xiaozhi Zhao
- Department of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing, 210008, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Qinhuai District, Nanjing, 210006, China.
| | - Yutian Dai
- Department of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing, 210008, China.
| |
Collapse
|
7
|
Yang H, Wei Z, Song Y, Du K, Yin N, Lu H, Li B, Hou L, Xing P, Chen L, Wang C, Xie S. NUAK1 promotes tumor metastasis through upregulating slug transcription in esophageal squamous cell carcinoma. Cancer Cell Int 2023; 23:258. [PMID: 37919754 PMCID: PMC10621130 DOI: 10.1186/s12935-023-03101-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/16/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Metastasis is still a major cause of poor pathological outcome and prognosis in esophageal squamous cell carcinoma (ESCC) patients. NUAK1 has been reported highly expressed in many human cancers and is associated with the poor prognosis of cancer patients. However, the role of NUAK1 and its underlying signaling mechanism in ESCC metastasis remain unclear. METHODS Expression of NUAK1 in ESCC was detected by real-time quantitative RT-PCR (qRT-PCR), Western blotting and immunohistochemical staining. MTT, colony formation, wound-healing and transwell assays were used to determine the role NUAK1 in vitro. Metastasis was evaluated by use of an experimental pulmonary metastasis model in BALB/c-nu/nu mice. The mechanisms were assessed by using coimmunoprecipitation, immunofluorescence and dual-luciferase reporter gene experiments. RESULTS NUAK1 was highly expressed in ESCC tissues compared with the adjacent normal esophageal epithelial tissues. Moreover, the elevated expression of NUAK1 positively correlated with tumor invasion depth, lymph node metastasis, pathological TNM stage, and poor survival in ESCC patients. Further experiments showed that NUAK1 overexpression did not change the cell viability and colony formation of ESCC cells, while remarkably promoted the migration and invasion in vitro and experimental pulmonary metastasis in vivo. Mechanistically, NUAK1 enhanced the transcription level of Slug, which enhanced the migratory and invasive capability of ESCC cells. Consistently, silencing Slug almost completely diminished the migration and invasion of NUAK1-overexpressing ESCC cells. Further studies demonstrated that NUAK1 upregulated the transcription activity of Slug through activating the JNK/c-Jun pathway. CONCLUSION These results demonstrated that NUAK1 promoted the metastasis of ESCC cells through activating JNK/c-Jun/Slug signaling, indicating NUAK1 is a promising therapeutic target for metastatic ESCC.
Collapse
Affiliation(s)
- Huiru Yang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China
| | - Zhen Wei
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China
| | - Yifan Song
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China
| | - Kexin Du
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China
| | - Nannan Yin
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China
| | - Hong Lu
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, 475004, Henan, China
| | - Bingbing Li
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, 475004, Henan, China
| | - Lili Hou
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China
| | - Panfei Xing
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China
| | - Liang Chen
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China.
| | - Chaojie Wang
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China.
| | - Songqiang Xie
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China.
- The Academy for Advanced Interdisciplinary Studies, Henan University, N. Jinming Ave., Kaifeng, 475004, Henan, China.
| |
Collapse
|
8
|
Zhou S, Cao C, Hu J. Long Non-Coding RNA Small Nucleolar RNA Host Gene 4 Induced by Transcription Factor SP1 Promoted the Progression of Nasopharyngeal Carcinoma Through Modulating microRNA-510-5p/Centromere Protein F Axis. Biochem Genet 2023; 61:1967-1986. [PMID: 36899270 DOI: 10.1007/s10528-023-10351-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/15/2023] [Indexed: 03/12/2023]
Abstract
Long non-coding RNAs (LncRNAs) are implicated with tumorigenesis and the development of nasopharyngeal carcinoma (NPC). Previous studies suggested that long non-coding RNA small nucleolar RNA host gene 4 (SNHG4) exerted oncogenic roles in various cancers. However, the function and molecular mechanism of SNHG4 in NPC have not been investigated. In our study, it was confirmed that the SNHG4 level was enriched in NPC tissues and cells. Functional assays indicated that SNHG4 depletion inhibited the proliferation and metastasis but promoted apoptosis of NPC cells. Furthermore, we identified miR-510-5p as a downstream gene of SNHG4 in NPC cells and SNHG4 upregulated CENPF expression by binding to miR-510-5p. Moreover, there was a positive (or negative) association between CENPF and SNHG4 (or miR-510-5p) expression in NPC. In addition, rescue experiments verified that CENPF overexpression or miR-510-5p silencing abrogated inhibitory effects on NPC tumorigenesis caused by SNHG4 deficiency. The study demonstrated that SNHG4 promoted NPC progression via miR-510-5p/CENPF axis, providing a novel potential therapeutic target for NPC treatments.
Collapse
Affiliation(s)
- Shao Zhou
- Department of Otorhinolaryngology, The Affiliated People's Hospital of Ningbo University, No. 251 East Baizhang Road, Ningbo, 315000, Zhejiang, China.
| | - Cheng Cao
- Department of Otorhinolaryngology, The Affiliated People's Hospital of Ningbo University, No. 251 East Baizhang Road, Ningbo, 315000, Zhejiang, China
| | - Jiandao Hu
- Department of Otorhinolaryngology, The Affiliated People's Hospital of Ningbo University, No. 251 East Baizhang Road, Ningbo, 315000, Zhejiang, China
| |
Collapse
|
9
|
Zeng H, Zhou S, Cai W, Kang M, Zhang P. LncRNA SNHG1: role in tumorigenesis of multiple human cancers. Cancer Cell Int 2023; 23:198. [PMID: 37684619 PMCID: PMC10492323 DOI: 10.1186/s12935-023-03018-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/06/2023] [Indexed: 09/10/2023] Open
Abstract
Small nucleolar RNA host gene 1 (SNHG1) is an important member of the SNHG family. This family is composed of a group of host genes that can be processed into small nucleolar RNAs and play important biological functions. In an oncogenic role, the SNHG1 expression is increased in various cancers, which has immense application prospects in the diagnosis, treatment, and prognosis of malignant tumors. In this review, we have summarized the role and molecular mechanism of SNHG1 in the development of various cancers. In addition, we have emphasized the clinical significance of SNHG1 in cancers in our article. This molecule is expected to be a new marker for potential usage in the diagnosis, prognosis, and treatment of cancer.
Collapse
Affiliation(s)
- Huang Zeng
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shouang Zhou
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Weiqiang Cai
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China.
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China.
| |
Collapse
|
10
|
Davoodvandi A, Rafiyan M, Mansournia MA, Rajabpoor Nikoo N, Saati M, Samimi M, Asemi Z. MicroRNA and gynecological cancers: Focus on miR-195. Pathol Res Pract 2023; 249:154784. [PMID: 37639954 DOI: 10.1016/j.prp.2023.154784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Different cancer types have been shown to have down-regulated expression levels of miR-195 as an anti-tumor agent. MiR-195 family members can inhibit cancer cell proliferation, angiogenesis, epithelial-mesenchymal transition and metastases, immunosuppression, glycolysis, drug resistance, and cancer stem cell development by targeting the 3'-UTR of the mRNA of different pro-tumor genes. MiR-195 identified as a tumor suppressor miR in a variety of cancers, most notably gynecological malignancies such as cervical, endometrial, and ovarian carcinoma. As a result, restoring miR-195 expression should be regarded as a potential therapy for either prevention or treatment of gynecological cancers. This review will present the most recent data about miR-195-mediated anti-tumor effects in gynecological malignancies, emphasizing its downstream signaling pathways and target genes, as well as prospective treatment techniques.
Collapse
Affiliation(s)
- Amirhossein Davoodvandi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Islamic Republic of Iran; Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Mahdi Rafiyan
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Nesa Rajabpoor Nikoo
- Department of Gynecology and Obstetrics, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Maryam Saati
- Department of Nursing, Semnan Branch, Islamic Azad University, Semnan, Islamic Republic of Iran
| | - Mansooreh Samimi
- Department of Gynecology and Obstetrics, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
11
|
Cui X, Wang J, Fan C, Jiang H, Li W. Astragalosides inhibit proliferation of fibroblast-like synoviocytes in experimental arthritis by modulating LncRNA S56464.1/miR-152-3p/Wnt1 signaling axis. Int J Rheum Dis 2023; 26:1547-1556. [PMID: 37317788 DOI: 10.1111/1756-185x.14782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
AIM Astragalus membranaceus (Fisch.) Bunge., the dried root of the plant A. membranaceus, is widely used in the treatment of rheumatoid arthritis (RA) in many Chinese herbal remedies. Astragalosides (AST) is the primary medicinal ingredient of A. membranaceus and has a therapeutic effect on RA, but the specific mechanism of this effect has yet to be elucidated. METHODS In this study, MTT and flow cytometry were used to determine the effects of AST on fibroblast-like synoviocyte (FLS) proliferation and cell cycle progression. Additionally, real-time quantitative polymerase chain reaction and Western blotting were used to determine the effects of AST on the LncRNA S56464.1/miR-152-3p/Wnt1 signaling axis and on critical genes that are essential to the Wnt pathway. RESULTS The data showed that after the administration of AST, FLS proliferation and LncRNA S56464.1, β-catenin, C-myc, Cyclin D1, and p-GSK-3β(Ser9)/GSK-3β expression were significantly reduced, and miR-152 and SFRP4 expression was notably increased. CONCLUSION These results suggest that AST can inhibit FLS proliferation by modulating the LncRNA S56464.1/miR-152-3p/Wnt1 signaling axis and that AST may be a potential therapeutic drug for RA.
Collapse
Affiliation(s)
- Xiaoya Cui
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jing Wang
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Chang Fan
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hui Jiang
- Clinical Research Experiment Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- College of Basic Medical, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Modern Chinese Medicine Department of Internal Medicine Application Foundation Research and Development, Hefei, Anhui, China
| | - Weiping Li
- College of Basic Medical, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
12
|
Cheng J, Huang A, Cheng J, Pei X, Yu L, Jin G, Xu E. Profile analysis of differentially expressed long non‑coding RNAs in metabolic memory induced by high glucose in human umbilical vein endothelial cells. Exp Ther Med 2023; 25:288. [PMID: 37206566 PMCID: PMC10189608 DOI: 10.3892/etm.2023.11987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/21/2023] [Indexed: 05/21/2023] Open
Abstract
Numerous long non-coding RNAs (lncRNAs) are dysregulated in the hyperglycemia-induced phenomenon of metabolic memory (MM). In the present study, the significance of these lncRNAs in MM was explored by screening for MM-involved differentially expressed lncRNAs (MMDELs) in human umbilical vein endothelial cells (HUVECs) induced by high glucose. A total of nine HUVEC samples were divided into three groups to mimic conditions of low and high glucose environments, as well as induce the state of metabolic memory. The expression of lncRNAs was profiled using RNA sequencing. Bioinformatic analysis was performed using the Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes databases to explore the parental genes from which the lncRNAs are transcribed and target genes of the MMDELs and generate enrichment datasets. Reverse transcription-quantitative PCR was performed to validate the expression levels of the selected lncRNAs. The present study identified 308 upregulated and 157 downregulated MMDELs, which were enriched in numerous physiologic processes. Key functional enrichment terms included 'cell cycle', 'oocyte meiosis' and 'p53 signaling pathway'. In conclusion, certain MMDELs may regulate the expression level of highly associated mRNAs through various mechanisms and pathways, thereby interfering with several processes, such as the regulation of the cell cycle, and affecting vascular endothelial cell function. Furthermore, the disorders of these lncRNAs can be retained in MM, further investigation into the functions of these lncRNAs may result in novel insights and treatments, which could help control MM in patients with diabetes.
Collapse
Affiliation(s)
- Jingya Cheng
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Anqi Huang
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Ji Cheng
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Xiaoyan Pei
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Lei Yu
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Guoxi Jin
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
- Correspondence to: Professor Guoxi Jin, Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Longzihu, Bengbu, Anhui 233004, P.R. China
| | - Erqin Xu
- Department of Physical Diagnostics, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
13
|
Fu F, Chen C, Du K, Li LS, Li R, Lei TY, Deng Q, Wang D, Yu QX, Yang X, Han J, Pan M, Zhen L, Zhang LN, Li J, Li FT, Zhang YL, Jing XY, Li FC, Li DZ, Liao C. Ndufa4 Regulates the Proliferation and Apoptosis of Neurons via miR-145a-5p/Homer1/Ccnd2. Mol Neurobiol 2023; 60:2986-3003. [PMID: 36763283 PMCID: PMC10122635 DOI: 10.1007/s12035-023-03239-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/09/2023] [Indexed: 02/11/2023]
Abstract
The Dandy-Walker malformation (DWM) is characterized by neuron dysregulation in embryonic development; however, the regulatory mechanisms associated with it are unclear. This study aimed to investigate the role of NADH dehydrogenase 1 alpha subcomplex 4 (NDUFA4) in regulating downstream signaling cascades and neuronal proliferation and apoptosis. Ndufa4 overexpression promoted the proliferation of neurons and inhibited their apoptosis in vitro, which underwent reverse regulation by the Ndufa4 short hairpin RNAs. Ndufa4-knockout (KO) mice showed abnormal histological alterations in the brain tissue, in addition to impaired spatial learning capacity and exploratory activity. Ndufa4 depletion altered the microRNA expressional profiles of the cerebellum: Ndufa4 inhibited miR-145a-5p expression both in the cerebellum and neurons. miR-145a-5p inhibited the proliferation of neurons and promoted their apoptosis. Ndufa4 promoted and miR-145a-5p inhibited the expression of human homer protein homolog 1 and cyclin D2 in neurons. Thus, Ndufa4 promotes the proliferation of neurons and inhibits their apoptosis by inhibiting miR-145a-5p, which directly targets and inhibits the untranslated regions of Homer1 and Ccnd2 expression.
Collapse
Affiliation(s)
- Fang Fu
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Chen Chen
- Department of Respirator, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Kun Du
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Lu-Shan Li
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Ru Li
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Ting-Ying Lei
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Qiong Deng
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Dan Wang
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Qiu-Xia Yu
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Xin Yang
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Jin Han
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Min Pan
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Li Zhen
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Li-Na Zhang
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Jian Li
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Fa-Tao Li
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Yong-Ling Zhang
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Xiang-Yi Jing
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Fu-Cheng Li
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Dong-Zhi Li
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China
| | - Can Liao
- Department of Prenatal Diagnostic Centre, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 of Jinsui Road of Guangzhou, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
14
|
Li S, Ni Y, Li C, Xiang Q, Zhao Y, Xu H, Huang W, Wang Y, Wang Y, Zhan J, Liu Y. Long noncoding RNA SNHG1 alleviates high glucose-induced vascular smooth muscle cells calcification/senescence by post-transcriptionally regulating Bhlhe40 and autophagy via Atg10. J Physiol Biochem 2023; 79:83-105. [PMID: 36194366 PMCID: PMC9905201 DOI: 10.1007/s13105-022-00924-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022]
Abstract
Long noncoding RNAs (lncRNAs) are emerging regulators of vascular diseases, yet their role in diabetic vascular calcification/aging remains poorly understood. In this study, we identified a down-expressed lncRNA SNHG1 in high glucose (HG)-induced vascular smooth muscle cells (HA-VSMCs), which induced excessive autophagy and promoted HA-VSMCs calcification/senescence. Overexpression of SNHG1 alleviated HG-induced HA-VSMCs calcification/senescence. The molecular mechanisms of SNHG1 in HA-VSMCs calcification/senescence were explored by RNA pull-down, RNA immunoprecipitation, RNA stability assay, luciferase reporter assay, immunoprecipitation and Western blot assays. In one mechanism, SNHG1 directly interacted with Bhlhe40 mRNA 3'-untranslated region and increased Bhlhe40 mRNA stability and expression. In another mechanism, SNHG1 enhanced Bhlhe40 protein SUMOylation by serving as a scaffold to facilitate the binding of SUMO E3 ligase PIAS3 and Bhlhe40 protein, resulting in increased nuclear translocation of Bhlhe40 protein. Moreover, Bhlhe40 suppressed the expression of Atg10, which is involved in the process of autophagosome formation. Collectively, the protective effect of SNHG1 on HG-induced HA-VSMCs calcification/senescence is accomplished by stabilizing Bhlhe40 mRNA and promoting the nuclear translocation of Bhlhe40 protein. Our study could provide a novel approach for diabetic vascular calcification/aging.
Collapse
Affiliation(s)
- Shuang Li
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Yuqing Ni
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Chen Li
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Qunyan Xiang
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Yan Zhao
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Hui Xu
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Wu Huang
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Yanjiao Wang
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Yi Wang
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China
| | - Junkun Zhan
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China.
| | - Youshuo Liu
- Department of Geriatrics, Institute of Aging and Age-Related Disease Research, The Second Xiangya Hospital, Central SouthUniversity, Changsha, 410011, Hunan, China.
| |
Collapse
|
15
|
Gao P, Zhao K, Lu W, Wang L, Zhang P. miR-339-3p inhibits cell growth and epithelial-mesenchymal transition in nasopharyngeal carcinoma by modulating the KAT6A/TRIM24 axis. Int J Clin Oncol 2022; 27:1684-1697. [PMID: 35976474 DOI: 10.1007/s10147-022-02231-8] [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: 02/17/2022] [Accepted: 08/03/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To explore the effect and mechanism of the miR-339-3p/KAT6A/TRIM24 axis in nasopharyngeal carcinoma (NPC) cell growth and epithelial-mesenchymal transition (EMT) progression. METHODS CNE2 and 5-8F NPC cell lines were transfected with miR-339-3p-mimic or sh-KAT6A alone or co-transfected with miR-339-3p-mimic and oe-KAT6A. The expression levels of miR-339-3p, KAT6A, TRIM24, and EMT-related proteins were assessed, in addition to cell biological behaviors. Then, the relationship between miR-339-3p and KAT6A was predicted and validated. The correlations between miR-339-3p and KAT6A or between KAT6A and TRIM24 were analyzed by Pearson coefficient and the enrichment of H3K23ac in TRIM24 promoter region was measured by chromatin immunoprecipitation. RESULTS miR-339-3p was downregulated, but KAT6A and TRIM24 were highly expressed in NPC cells and tissues. Upregulated miR-339-3p or downregulated KAT6A could inhibit the growth and EMT of NPC cells. Further experiments showed that miR-339-3p regulated NPC cell growth and EMT by mediating KAT6A in a targeted fashion. KAT6A was positively correlated with TRIM24, and the enrichment of H3K23ac was much higher in NPC tissues. miR-339-3p suppressed the growth and EMT of NPC cells by the KAT6A/TRIM24 axis. In a xenograft study, miR-339-3p overexpression inhibited NPC tumor growth in vivo. CONCLUSION Conclusively, miR-339-3p inhibited the growth and EMT of NPC cells via the KAT6A/TRIM24 axis.
Collapse
Affiliation(s)
- Pei Gao
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Erqi District, Zhengzhou, Henan, 450052, People's Republic of China.
| | - Kun Zhao
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Wuhao Lu
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Erqi District, Zhengzhou, Henan, 450052, People's Republic of China
| | - Liang Wang
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Erqi District, Zhengzhou, Henan, 450052, People's Republic of China
| | - Peng Zhang
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1, East Jianshe Road, Erqi District, Zhengzhou, Henan, 450052, People's Republic of China
| |
Collapse
|
16
|
Hsieh PL, Chao SC, Chu PM, Yu CC. Regulation of Ferroptosis by Non-Coding RNAs in Head and Neck Cancers. Int J Mol Sci 2022; 23:3142. [PMID: 35328568 PMCID: PMC8950679 DOI: 10.3390/ijms23063142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is a newly identified mode of programmed cell death characterized by iron-associated accumulation of lipid peroxides. Emerging research on ferroptosis has suggested its implication in tumorigenesis and stemness of cancer. On the other hand, non-coding RNAs have been shown to play a pivotal role in the modulation of various genes that affect the progression of cancer cells and ferroptosis. In this review, we summarize recent advances in the theoretical modeling of ferroptosis and its relationship between non-coding RNAs and head and neck cancers. Aside from the significance of ferroptosis-related non-coding RNAs in prognostic relevance, we also review how these non-coding RNAs participate in the regulation of iron, lipid metabolism, and reactive oxygen species accumulation. We aim to provide a thorough grounding in the function of ferroptosis-related non-coding RNAs based on current knowledge in an effort to develop effective therapeutic strategies for head and neck cancers.
Collapse
Affiliation(s)
- Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404333, Taiwan; (P.-L.H.); (P.-M.C.)
| | - Shih-Chi Chao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Medical Research and Education, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan 265, Taiwan
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404333, Taiwan; (P.-L.H.); (P.-M.C.)
| | - Cheng-Chia Yu
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan;
- School of Dentistry, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
17
|
Jia Y, Chen X, Zhao D, Ma S. SNHG1/miR-194-5p/MTFR1 Axis Promotes TGFβ1-Induced EMT, Migration and Invasion of Tongue Squamous Cell Carcinoma Cells. Mol Biotechnol 2022; 64:780-790. [PMID: 35107755 DOI: 10.1007/s12033-021-00445-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022]
Abstract
Tongue squamous cell carcinoma (TSCC) is a common malignancy with aggressive biological behaviors. Mitochondrial fission regulator 1 (MTFR1), is aberrantly expressed in head and neck squamous cell carcinoma (HNSC), but its role in TSCC remains unclear. We aimed to explore the role of MTFR1 in TSCC. The expression of long non-coding RNA small nucleolar RNA host gene 1 (SNHG1), microRNA-194-5p and MTFR1 in TSCC cells was measured by RT-qPCR. Luciferase reporter assay and RNA pull down assay were applied to confirm the binding capacity between miR-194-5p and SNHG1 (or MTFR1). TSCC cell invasion and migration were accessed by Transwell assays. The protein levels of MTFR1 and epithelial-mesenchymal transition (EMT) markers were examined by western blot. MTFR1 had high expression level in TSCC. MTFR1 knockdown inhibited transforming growth factor β1 (TGFβ1)-induced EMT, migration and invasion of TSCC cells in vitro. MiR-194-5p targeted MTFR1 and negatively regulated its expression. In addition, SNHG1 upregulated the expression of MTFR1 by binding with miR-194-5p. Importantly, SNHG1 promoted EMT, invasion and migration of TSCC cells by upregulating MTFR1. SNHG1/miR-194-5p/MTFR1 axis promotes TGFβ1-induced EMT, migration and invasion of cells in TSCC, which could be potential targets for treating TSCC patients.
Collapse
Affiliation(s)
- Yonglu Jia
- Department of Stomotology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, No. 118 Wansheng Street, Suzhou Industrial Park, Suzhou, 215028, Jiangsu, China
| | - Xiaojuan Chen
- Department of Stomotology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, No. 118 Wansheng Street, Suzhou Industrial Park, Suzhou, 215028, Jiangsu, China
| | - Dayong Zhao
- Department of Stomotology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, No. 118 Wansheng Street, Suzhou Industrial Park, Suzhou, 215028, Jiangsu, China
| | - Sancheng Ma
- Department of Stomotology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, No. 118 Wansheng Street, Suzhou Industrial Park, Suzhou, 215028, Jiangsu, China.
| |
Collapse
|
18
|
Hong X, Wang G, Xu G, Shi W, Wang T, Rong Z, Mo C. Prognostic value of EGFR and p-EGFR in nasopharyngeal carcinoma: A systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e28507. [PMID: 35060503 PMCID: PMC8772636 DOI: 10.1097/md.0000000000028507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 12/13/2021] [Indexed: 01/05/2023] Open
Abstract
PURPOSE To evaluate the prognostic effect and clinical significance of epidermal growth factor receptor and its phosphorlated form (EGFR/p-EGFR) in nasopharyngeal carcinoma. METHODS A systematic review and meta-analysis was designed. We visited PubMed, Embase, China National Knowledge Infrastructure Database, Database of Chinese sci-tech periodicals, WanFang Database, and China Biology Medicine disc to search for Chinese and English publications of prospective studies and retrospective studies investigating the association of EGFR/p-EGFR and nasopharyngeal carcinoma prognosis from inception to April 2021. The inclusion criteria were that the samples should be pathologically confirmed as nasopharyngeal carcinoma and the expression of EGFR/p-EGFR should be detected via immunohistochemistry; the study should analyze the prognostic significance of EGFR/p-EGFR in nasopharyngeal carcinoma; hazard ratio (HR) and 95% confidence interval (CI) should be reported in the study or could be derived from survival curves; and the outcomes of the study should include overall survival (OS), disease-free survival (DFS), progression-free survival (PFS), and distant metastasis-free survival (DMFS). RESULTS A total of 18 studies evaluating 1451 samples were included. For studies that reported OS as an outcome, EGFR overexpression indicated worse OS of nasopharyngeal carcinoma patients. The heterogeneity between studies was high (I2 = 91%, P < .01), and a random-effect model was used to combine the effect size (HR = 1.71, 95% CI [1.21, 2.41], P < .01). Further sensitivity analysis and prespecified subgroup analysis were performed to detect the source of heterogeneity, and the results showed that the heterogeneity could not be eliminated. Publication bias assessed by funnel plots and Begg test and Egger test was low (Begg test: P = .846 and Egger test: P = .074). p-EGFR was not correlated with the OS of nasopharyngeal carcinoma patients (HR = 1.01, 95% CI [0.88, 1.15], P = .92). For studies that reported DFS, EGFR overexpression was associated with worse DFS in patients with nasopharyngeal carcinoma (HR = 2.53, 95% CI [1.84, 3.47], P < .01). For studies that reported PFS, EGFR overexpression was not correlated with the PFS of nasopharyngeal carcinoma patients (HR = 1.86, 95% CI [0.90, 3.82], P = .09). For studies that reported DMFS, EGFR overexpression was not correlated with the DMFS of nasopharyngeal carcinoma patients, and high heterogeneity between studies was detected (I2 = 97%, P < .01). A random-effect model was used to combine the effect size (HR = 1.80, 95% CI [0.56, 5.76], P = .32). A sensitivity analysis was conducted. Publication bias was detected to be low (Begg test: P = .817 and Egger test: P = .954). There was no correlation between p-EGFR overexpression and DMFS in patients with nasopharyngeal carcinoma (HR = 1.20, 95% CI [0.95, 1.52], P = .12). CONCLUSION In nasopharyngeal carcinoma patients, EGFR overexpression could be used as a biomarker that predicts poor OS and DFS, but not a prognostic biomarker for PFS and DMFS. The overexpression of p-EGFR was not shown to be associated with the prognosis of nasopharyngeal carcinoma patients and could not be used as a prognostic biomarker. ETHICS AND DISSEMINATION This study was registered on the International Platform of Registered Systematic Review and Meta-analysis Protocols (INPLASY), and reported as stated by the Preferred Reporting Items for Systematic reviews and Meta-Analyses. Neither ethical approval nor informed consent was required since this study was conducted based on previous publications. INPLASY REGISTRATION NUMBER INPLASY 202150010.
Collapse
Affiliation(s)
- Xiaohua Hong
- Guangxi University of Chinese medicine, NanNing Guangxi, China
| | - Guangyao Wang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning Guangxi, China
| | - Guanglan Xu
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning Guangxi, China
| | - Wei Shi
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning Guangxi, China
| | - Tongbiao Wang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning Guangxi, China
| | - Zhen Rong
- Bao’an Authentic TCM Therapy Hospital, Shenzhen Guangdong, China
| | - Chunmei Mo
- Bao’an Authentic TCM Therapy Hospital, Shenzhen Guangdong, China
| |
Collapse
|
19
|
Function of miRNA-145-5p in the pathogenesis of human disorders. Pathol Res Pract 2022; 231:153780. [DOI: 10.1016/j.prp.2022.153780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/08/2022] [Accepted: 01/22/2022] [Indexed: 01/09/2023]
|
20
|
Homayoonfal M, Asemi Z, Yousefi B. Targeting long non coding RNA by natural products: Implications for cancer therapy. Crit Rev Food Sci Nutr 2021:1-29. [PMID: 34783279 DOI: 10.1080/10408398.2021.2001785] [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] [Indexed: 12/23/2022]
Abstract
In spite of achieving substantial progress in its therapeutic strategies, cancer-associated prevalence and mortality are persistently rising globally. However, most malignant cancers either cannot be adequately diagnosed at the primary phase or resist against multiple treatments such as chemotherapy, surgery, radiotherapy as well as targeting therapy. In recent decades, overwhelming evidences have provided more convincing words on the undeniable roles of long non-coding RNAs (lncRNAs) in incidence and development of various cancer types. Recently, phytochemical and nutraceutical compounds have received a great deal of attention due to their inhibitory and stimulatory effects on oncogenic and tumor suppressor lncRNAs respectively that finally may lead to attenuate various processes of cancer cells such as growth, proliferation, metastasis and invasion. Therefore, application of phytochemicals with anticancer characteristics can be considered as an innovative approach for treating cancer and increasing the sensitivity of cancer cells to standard prevailing therapies. The purpose of this review was to investigate the effect of various phytochemicals on regulation of lncRNAs in different human cancer and evaluate their capabilities for cancer treatment and prevention.
Collapse
Affiliation(s)
- Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Wu Q, Zhao Y, Shi R, Wang T. LncRNA SNHG16 Facilitates Nasopharyngeal Carcinoma Progression by Acting as ceRNA to Sponge miR-520a-3p and Upregulate MAPK1 Expression. Cancer Manag Res 2021; 13:4103-4114. [PMID: 34045897 PMCID: PMC8147710 DOI: 10.2147/cmar.s305544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Background Accumulating evidence shows that lncRNAs are widely involved cellular processes of various tumors. The aim of this study was to explore the potential role and molecular mechanism of lncRNA SNHG16 in nasopharyngeal carcinoma (NPC). Methods SNHG16, miR-520a-3p, and MAPK1 levels were measured by RT-qPCR assay. CCK-8, colony formation, transwell, and flow cytometry assays were adopted to analyze the proliferation, migration, invasion, and apoptosis of NPC cell lines (SUNE1 and 5–8F). Murine xenograft model was used to investigate tumor growth and metastasis in vivo. Immunohistochemical staining was employed to evaluate the levels of Bcl-2, cleaved caspase-3, Bax, and Ki-67. Dual-luciferase reporter assays were conducted to analyze the binding ability between miR-520a-3p and SNHG16 or MAPK1. Results SNHG16 was overexpressed in NPC tissues and cells. High SNHG16 expression indicated a poor prognosis. SNHG16 knockdown could cause significant inhibition on cell proliferation and metastasis, induce cell apoptosis in NPC cells, and repressed tumor growth and metastasis in vivo. Additionally, SNHG16 could directly bind to miR-520a-3p, thus positively regulating MAPK1 expression. Moreover, functional analysis indicated that miR-520a-3p exerted a tumor-suppressing role in NPC progression. Rescue assays demonstrated that MAPK1 upregulation could abrogate the inhibitory effects on NPC cell proliferation and metastasis, as well as the promoting effects on NPC cell apoptosis caused by SNHG16 knockdown. In conclusion, SNHG16 contributed to the proliferation and metastasis of NPC cells by modulating the miR-520a-3p/MAPK1 axis. Conclusion These results suggest that SNHG16 acts as an oncogene in the progression of NPC via modulating the miR-520a-3p/MAPK1 axis.
Collapse
Affiliation(s)
- Qingwei Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yingying Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Runjie Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Tao Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
22
|
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
|
23
|
Biagioni A, Tavakol S, Ahmadirad N, Zahmatkeshan M, Magnelli L, Mandegary A, Samareh Fekri H, Asadi MH, Mohammadinejad R, Ahn KS. Small nucleolar RNA host genes promoting epithelial-mesenchymal transition lead cancer progression and metastasis. IUBMB Life 2021; 73:825-842. [PMID: 33938625 DOI: 10.1002/iub.2501] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/22/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023]
Abstract
The small nucleolar RNA host genes (SNHGs) belong to the long non-coding RNAs and are reported to be able to influence all three levels of cellular information-bearing molecules, that is, DNA, RNA, and proteins, resulting in the generation of complex phenomena. As the host genes of the small nucleolar RNAs (snoRNAs), they are commonly localized in the nucleolus, where they exert multiple regulatory functions orchestrating cellular homeostasis and differentiation as well as metastasis and chemoresistance. Indeed, worldwide literature has reported their involvement in the epithelial-mesenchymal transition (EMT) of different histotypes of cancer, being able to exploit peculiar features, for example, the possibility to act both in the nucleus and the cytoplasm. Moreover, SNHGs regulation is a fundamental topic to better understand their role in tumor progression albeit such mechanism is still debated. Here, we reviewed the biological functions of SNHGs in particular in the EMT process and discussed the perspectives for new cancer therapies.
Collapse
Affiliation(s)
- Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Florence, Italy
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nooshin Ahmadirad
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Zahmatkeshan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Florence, Italy
| | - Ali Mandegary
- Department of Pharmacology & Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Hojjat Samareh Fekri
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.,Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Malek Hossein Asadi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Reza Mohammadinejad
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Wu Y, Zhu B, Yan Y, Bai S, Kang H, Zhang J, Ma W, Gao Y, Hui B, Li R, Zhang X, Ren J. Long non-coding RNA SNHG1 stimulates ovarian cancer progression by modulating expression of miR-454 and ZEB1. Mol Oncol 2021; 15:1584-1596. [PMID: 33641229 PMCID: PMC8096788 DOI: 10.1002/1878-0261.12932] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/21/2020] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer (OC) is highly prevalent and is associated with high mortality rates due to metastasis and relapse. In this study, we assessed the role of long non-coding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) in OC to gain further insight into mechanisms that contribute to its aggressiveness. We analyzed the correlation between SNHG1, miR-454 and zinc finger E-box-binding homeobox 1 (ZEB1) using a dual-luciferase reporter assay. Alterations in cell metastasis and invasiveness were observed using wound-healing and Transwell invasion assays, respectively. Tumor xenografts allowed us to monitor liver metastasis of mice injected with A2780 cells. We found that SNHG1 is overexpressed in OC. Downregulation of SNHG1 promoted miR-454 expression and reduced ZEB1 levels. In addition, knockdown of SNHG1, also reduced the aggressiveness of A2780 and SK-OV3 cells. Furthermore, SNHG1 downregulation by siRNA hindered cell migration and invasion; however, this effect was reversed by co-transfection of miR-454 into A2780 and SK-OV3 cells. Moreover, SNHG1 increased ZEB1 expression by downregulating miR-454 and activated Akt signaling, thereby promoting epithelial-mesenchymal transition and enhancing the invasiveness of OC cells. Tumor xenograft analyses confirmed that SNHG1 affects OC proliferation and metastasis in vivo. In summary, our data demonstrate that SNHG1 plays crucial roles in tumor progression and may be a useful maker for OC prognosis.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/pathology
- Cell Line, Tumor
- Cell Proliferation/genetics
- Disease Progression
- Epithelial-Mesenchymal Transition/genetics
- Female
- Gene Expression Regulation, Neoplastic
- HEK293 Cells
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- MicroRNAs/genetics
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/physiology
- Zinc Finger E-box-Binding Homeobox 1/genetics
Collapse
Affiliation(s)
- YinYing Wu
- Department of Chemotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Bo Zhu
- Department of Pulmonary and Critical Care MedicineFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Yanli Yan
- Department of Radiotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Shuheng Bai
- Department of Radiotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Haojing Kang
- Department of Radiotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | | | - Wen Ma
- Medical SchoolXi’an Jiaotong UniversityChina
| | - Ying Gao
- Department of Radiotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Beina Hui
- Department of Radiotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Rong Li
- Department of Radiotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Xiaozhi Zhang
- Department of Radiotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Juan Ren
- Department of Radiotherapy, Oncology DepartmentFirst Affiliated Hospital of Xi’an Jiaotong UniversityChina
| |
Collapse
|
25
|
Kong D, Long D, Liu B, Pei D, Cao N, Zhang G, Xia Z, Luo M. Downregulation of long non-coding RNA LOC101928477 correlates with tumor progression by regulating the epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Thorac Cancer 2021; 12:1303-1311. [PMID: 33713583 PMCID: PMC8088935 DOI: 10.1111/1759-7714.13858] [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: 12/02/2020] [Revised: 01/09/2021] [Accepted: 01/09/2021] [Indexed: 02/07/2023] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is one of the deadliest malignancies. There is a growing body of evidence showing that long non‐coding RNAs (lncRNAs) play critical roles in ESCC oncogenesis. The present study aimed to explore the role of LOC101928477, a newly discovered lncRNA, in the development and metastasis of ESCC. Methods In this study, real‐time PCR, western blotting, cell counting kit‐8 (CCK‐8), flow cytometry, colony formation, wound healing, transwell migration/invasion assay, immunofluorescence, and immunohistochemistry were used. We also applied an in situ xenograft mouse model and a lung metastasis mouse model to verify our findings. Results We determined that LOC101928477 expression was inhibited in ESCC tissue and ESCC cell lines when compared with controls. Moreover, forced expression of LOC101928477 effectively inhibited ESCC cell proliferation, colony formation, migration, and invasion via suppression of epithelial‐mesenchymal transition (EMT). Furthermore, LOC101928477 overexpression inhibited in situ tumor growth and lung metastasis in a mouse model. Conclusions Together, our results suggested that LOC101928477 could be a novel suppressor gene involved in ESCC progression.
Collapse
Affiliation(s)
- Demiao Kong
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Dali Long
- Department of Intensive Care Unit, Guizhou Provincial People's Hospital, Guiyang, China
| | - Bo Liu
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Dengke Pei
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Na Cao
- Department of Logistics, Guizhou Provincial People's Hospital, Guizhou, Guiyang, China
| | - Guihua Zhang
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Zhenkun Xia
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Meng Luo
- Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
26
|
Ma H, Dong A. Dysregulation of lncRNA SNHG1/miR-145 axis affects the biological function of human carotid artery smooth muscle cells as a mechanism of carotid artery restenosis. Exp Ther Med 2021; 21:423. [PMID: 33777187 PMCID: PMC7967805 DOI: 10.3892/etm.2021.9867] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Carotid angioplasty and stenting have developed into reliable options for patients with carotid stenosis. However, postoperative restenosis remains a serious and unresolved problem. Restenosis is partly caused by the proliferation of vascular smooth muscle cells. As certain long non-coding RNAs (lncRNAs) affect cell proliferation and migration, the present study aimed to investigate them as novel biomarkers for restenosis development and to further reveal the potential underlying mechanisms. The expression of lncRNA small nucleolar RNA host gene 1 (SNHG1) and microRNA145 (miR-145) in human carotid artery smooth muscle cells (hHCtASMCs) was analyzed using reverse transcription-quantitative PCR. In addition, a luciferase reporter assay was performed to investigate the interaction between SNHG1 and miR-145. The effects of the SNHG1/miR-145 axis on the proliferation and migration of hHCtASMCs were evaluated by Cell Counting Kit-8 and Transwell assays. Serum SNHG1 and miR-145 expression levels were increased and decreased, respectively, in patients with restenosis (all P<0.001). High SNHG1 and low miR-145 were identified as risk factors for restenosis onset (all P<0.01). Furthermore, decreasing SNHG1 expression levels in hHCtASMCs inhibited cell proliferation and migration. The luciferase reporter assay and expression results demonstrated that miR-145 may be a target of SNHG1 and mediated the effects of SNHG1 on hHCtASMC proliferation and migration. The results obtained suggested that abnormal expression of SNHG1 and miR-145 may be risk factors for restenosis. The present study revealed that the SNHG1/miR-145 axis regulates hHCtASMC proliferation and migration, indicating its potential for restenosis prevention and treatment.
Collapse
Affiliation(s)
- Huanhuan Ma
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Aiqin Dong
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
27
|
Zhang L, Wu X, Li Y, Teng X, Zou L, Yu B. LncRNA SNHG5 promotes cervical cancer progression by regulating the miR-132/SOX4 pathway. Autoimmunity 2021; 54:88-96. [PMID: 33622094 DOI: 10.1080/08916934.2020.1864731] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND The long non-coding RNA (lncRNA) small nucleolar RNA host gene 5 (SNHG5) has been verified as a crucial regulator in many types of tumours but not clear in cervical cancer (CC). This study aims to investigate the effect and further mechanisms of lncRNA SNHG5 in CC. METHODS The expression of SNHG5 and miR-132, as well as SOX4 (sex-determining region Y-box 4) mRNA expression were determined by quantitative real-time PCR (qRT-PCR). The protein level of SOX4 and epithelial-mesenchymal transition (EMT)-related proteins were evaluated by western blot. Then, Edu and Transwell assay were performed to assess the proliferation, migration and invasion of CC cells. RNA immunoprecipitation (RIP) and RNA pull-down assay were conducted to explore the relationship between SNHG5 and miR-132. RESULTS SNHG5 and SOX4 were upregulated, and miR-132 was downregulated in CC tissues and cell lines. SNHG5 was positively correlated with FIGO stage (p = .003) and lymph node metastasis (p = .001). Pearson's correlation analysis conveyed that SNHG5 was positively correlated with SOX4, and miR-132 was negatively correlated with SOX4 and SNHG5. Knockdown of SNHG5 in vitro reduced CC cell proliferation, migration and invasion through regulating miR-132. Moreover, overexpression of miR-132 restrained CC cell proliferation, migration, and invasion through targeting SOX4, and SNHG5 enhanced SOX4 expression via negatively regulating miR-132. CONCLUSION SNHG5 promotes SOX4 expression to accelerate CC cell proliferation, migration and invasion through negatively regulating miR-132.
Collapse
Affiliation(s)
- Liqin Zhang
- Department of Laboratory, Jinhua People's Hospital, China
| | - Xiaoming Wu
- Department of Laboratory, Hangzhou Jianggan District People's Hospital, Hangzhou, China
| | - Yue Li
- Department of Laboratory, Jinhua People's Hospital, China
| | - Xianlin Teng
- Department of Laboratory, Jinhua People's Hospital, China
| | - Libo Zou
- Department of Laboratory, Jinhua People's Hospital, China
| | - Beiwei Yu
- Department of Laboratory, Hangzhou Jianggan District People's Hospital, Hangzhou, China
| |
Collapse
|
28
|
Shen P, Qu L, Wang J, Ding Q, Zhou C, Xie R, Wang H, Ji G. LncRNA LINC00342 contributes to the growth and metastasis of colorectal cancer via targeting miR-19a-3p/NPEPL1 axis. Cancer Cell Int 2021; 21:105. [PMID: 33588834 PMCID: PMC7885559 DOI: 10.1186/s12935-020-01705-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Background Long intergenic non-protein coding RNA 00342 (LINC00342) has been identified as a novel oncogene. However, the functional role of LINC00342 in colorectal cancer (CRC) remains unclear. Methods The expression of LINC00342 is detected by real-time PCR (RT-PCR) analysis. Cell proliferation, migration and invasion and xenograft model are examined to analyze the biological functions of LINC00342 in vitro and in vivo using colony formation, would healing and transwell analyses. Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays are used to identify the target interactions between LINC00342, miR-19a-3p and aminopeptidase like 1 (NPEPL1). Results LINC00342 was highly expressed in CRC. Down-regulation of LINC00342 inhibited cell proliferation and metastasis of CRC cells. Moreover, knocking down LINC00342 inhibited the tumor growth in vivo. Mechanistic investigation revealed that LINC00342 might sponge miR-19a-3p to regulate NPEPL1 expression. Further investigation indicated that the ontogenesis facilitated by LINC00342 was inhibited due to the depletion of NPEPL1. Conclusion LINC00342 promotes CRC progression by competitively binding miR-19a-3p with NPEPL1.
Collapse
Affiliation(s)
- Peng Shen
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Lili Qu
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Jingjing Wang
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Quchen Ding
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Chuanwen Zhou
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Rui Xie
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Honggang Wang
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Guozhong Ji
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
29
|
Grubelnik G, Boštjančič E, Aničin A, Dovšak T, Zidar N. MicroRNAs and Long Non-Coding RNAs as Regulators of NANOG Expression in the Development of Oral Squamous Cell Carcinoma. Front Oncol 2021; 10:579053. [PMID: 33643897 PMCID: PMC7906007 DOI: 10.3389/fonc.2020.579053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
NANOG is a stem cell transcription factor that is believed to play an important role in the development of oral squamous cell carcinoma (OSCC), but there is limited data regarding the role of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) in the regulation of NANOG expression. We therefore analyzed expression of NANOG, NANOG-regulating miRNAs and lncRNAs in OSCC cancerogenesis, using oral biopsy samples from 66 patients including normal mucosa, dysplasia, and OSCC. Expression analysis of NANOG, miR-34a, miR-145, RoR, SNHG1, AB209630, and TP53 was performed using qPCR and immunohistochemistry for NANOG protein detection. NANOG protein showed no staining in normal mucosa, very weak in low-grade dysplasia, and strong staining in high-grade dysplasia and OSCC. NANOG, miR-145, RoR, and SNHG1 showed up-regulation, TP53 and miR-34a showed down-regulation, and AB209630 showed variable expression during cancerogenesis. NANOG mRNA was up-regulated early in cancerogenesis, before strong protein expression can be detected. NANOG was in correlation with miR-145 and RoR. Our results suggest that miRNAs and lncRNAs, particularly miR-145 and RoR, might be important post-transcription regulatory mechanisms of NANOG in OSCC cancerogenesis. Furthermore, NANOG protein detection has a diagnostic potential for oral high-grade dysplasia, distinguishing it from low-grade dysplasia and non-neoplastic reactive lesions.
Collapse
Affiliation(s)
- Gašper Grubelnik
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Emanuela Boštjančič
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Aleksandar Aničin
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tadej Dovšak
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Department of Maxillofacial and Oral Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Nina Zidar
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
30
|
Zheng C, Yu S. Expression and gene regulatory network of SNHG1 in hepatocellular carcinoma. BMC Med Genomics 2021; 14:28. [PMID: 33499863 PMCID: PMC7836560 DOI: 10.1186/s12920-021-00878-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/17/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Small nucleolar RNA host gene 1 (SNHG1), a long noncoding RNA (lncRNA), is a transcript that negatively regulates tumour suppressor genes, such as p53. Abnormal SNHG1 expression is associated with cell proliferation and cancer. We used sequencing data downloaded from Genomic Data Commons to analyse the expression and interaction networks of SNHG1 in hepatocellular carcinoma (HCC). METHODS Expression was examined using the limma package of R and verified by Gene Expression Profiling Interactive Analysis. We also obtained miRNA expression data from StarBase to determine the lncRNA-miRNA-mRNA-related RNA regulatory network in HCC. Kaplan-Meier (KM) analysis was performed using the survival package of R. Gene Ontology annotation of genes was carried out using Metascape. RESULTS We found that SNHG1 was overexpressed and often amplified in HCC patients. In addition, SNHG1 upregulation was associated with the promotion of several primary biological functions, including cell proliferation, transcription and protein binding. Moreover, we found similar trends of small nucleolar RNA host gene 1 (SNHG1), E2F8 (E2F transcription factor 8), FANCE (FA complementation group E) and LMNB2 (encodes lamin B2) expression. In the SNHG1-associated network, high expression levels of SNHG1 (log-rank P value = 0.0643), E2F8 (log-rank P value = 0.000048), FANCE (log-rank P value = 0.00125) and LMNB2 (log-rank P value = 0.0392) were significantly associated with poor survival. Single-cell analysis showed that E2F8 may play an important role in tumorigenesis or cancer development. CONCLUSIONS Our results highlight the benefit of utilizing multiple datasets to understand the functional potential regulatory networks of SNHG1 and the role of SNHG1 in tumours.
Collapse
Affiliation(s)
- Chaoran Zheng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Shicheng Yu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Luogang District, Guangzhou, 510530, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Bioland Laboratory, Guangzhou, 510005, China.
| |
Collapse
|
31
|
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
|
32
|
Liu W, Zhang Y, Luo B. Long Non-coding RNAs in Gammaherpesvirus Infections: Their Roles in Tumorigenic Mechanisms. Front Microbiol 2021; 11:604536. [PMID: 33519750 PMCID: PMC7843584 DOI: 10.3389/fmicb.2020.604536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) regulate gene expression at the epigenetic, transcriptional, or posttranscriptional level by interacting with protein, DNA, and RNA. Emerging evidence suggests that various lncRNAs are abnormally expressed and play indispensable roles in virus-triggered cancers. Besides, a growing number of studies have shown that virus-encoded lncRNAs participate in tumorigenesis. However, the functions of most lncRNAs in tumors caused by oncogenic viruses and their underlying mechanisms remain largely unknown. In this review, we summarize current findings regarding lncRNAs involved in cancers caused by Epstein–Barr virus (EBV) and Kaposi’s sarcoma herpesvirus (KSHV). Additionally, we discuss the contribution of lncRNAs to tumor occurrence, development, invasion, and metastasis; the roles of lncRNAs in key signaling pathways and their potential as biomarkers and therapeutic targets for tumor diagnostics and treatment.
Collapse
Affiliation(s)
- Wen Liu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China.,Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Bing Luo
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
33
|
Faisal M, Kim JH, Yoo KH, Roh EJ, Hong SS, Lee SH. Development and Therapeutic Potential of NUAKs Inhibitors. J Med Chem 2020; 64:2-25. [PMID: 33356242 DOI: 10.1021/acs.jmedchem.0c00533] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
NUAK isoforms, NUAK1 (ARK5) and NUAK2 (SNARK), are important members of the AMPK family of protein kinases. They are involved in a broad spectrum of physiological and cellular events, and sometimes their biological roles overlap. NUAK isoform dysregulation is associated with numerous pathological disorders, including neurodegeneration, metastatic cancer, and diabetes. Therefore, they are promising therapeutic targets in metabolic diseases and cancers; consequently, various NUAK-targeted inhibitors have been disclosed. The first part of this review comprises a brief discussion of the homology, expression, structure, and characteristics of NUAK isoforms. The second part focuses on NUAK isoforms' involvement in crucial biological operations, including mechanistic findings, highlighting how their abnormal functioning contributes to disease progression and quality of life. The third part summarizes the key findings and applications of targeting NUAK isoforms for treating multiple cancers and neurodegenerative disorders. The final part systematically presents a critical review and analysis of the literature on NUAK isoform inhibitions through small molecules.
Collapse
Affiliation(s)
- Muhammad Faisal
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jae Ho Kim
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Kyung Ho Yoo
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Eun Joo Roh
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea.,Chemical Kinomics Research Center, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Soon Sun Hong
- Department of Biomedical Sciences, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| | - So Ha Lee
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
34
|
Liu F, Feng XX, Zhu SL, Lin L, Huang HY, Zhang BY, Huang JL. Long non-coding RNA SNHG1 regulates rheumatoid synovial invasion and proliferation by interaction with PTBP1. Int Immunopharmacol 2020; 90:107182. [PMID: 33218941 DOI: 10.1016/j.intimp.2020.107182] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/04/2020] [Accepted: 11/02/2020] [Indexed: 02/09/2023]
Abstract
Fibroblast-like synoviocytes (FLSs) in rheumatoid arthritis (RA) present proliferative and aggressive cell phenotype. RA-FLSs are the essential effector cells that lead to symptoms like synovial inflammation and joint destruction. Currently, the cause of RA-FLSs involving in the pathological process of RA remains unknown. Accumulate researches have demonstrated that lncRNAs may play a critical role in regulating the biological behaviors of RA-FLSs, but the mechanism is still unclear. Here, we found that lncRNA small nucleolar RNA host gene 1 (SNHG1) is up-regulated in RA-FLSs compared with FLSs from trauma arthritis and osteoarthritis patients. The results suggest that SNHG1 in RA-FLSs helps to sustain the cellular functions of proliferation, migration and invasion. Furthermore, the regulation mechanism depends on the interaction between SNHG1 and polypyridine tract-binding protein 1 (PTBP1). This interaction influences PTBP1 expression that participates in the regulation of RA-FLSs biological behaviors. Our results suggest that up-regulated SNHG1 of RA-FLSs may contribute to synovial aggression and disease progression in RA and be favourable for RA treatment target RA-FLSs.
Collapse
Affiliation(s)
- Fang Liu
- Division of Rheumatology, Department of Internal Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Xue Feng
- Division of Rheumatology, Department of Internal Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shang-Ling Zhu
- Division of Rheumatology, Department of Internal Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lang Lin
- Division of Rheumatology, Department of Internal Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong-Yu Huang
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Bai-Yu Zhang
- Division of Rheumatology, Department of Internal Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jian-Lin Huang
- Division of Rheumatology, Department of Internal Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
35
|
Zhang H, Liu B, Shi X, Sun X. Long noncoding RNAs: Potential therapeutic targets in cardiocerebrovascular diseases. Pharmacol Ther 2020; 221:107744. [PMID: 33181193 DOI: 10.1016/j.pharmthera.2020.107744] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
Cardiocerebrovascular disease is a collective term for cardiovascular and cerebrovascular diseases. Because of the complex mechanisms involved in cardiocerebrovascular diseases, limited effective treatments have been developed. With advancements in precision medicine, studies have focused on long noncoding RNAs (lncRNAs) in cerebrovascular diseases. LncRNAs, which are over 200 nucleotides long, regulate gene expression at epigenetic, transcriptional, and post-transcriptional levels. Moreover, lncRNAs play pivotal roles in the progression of cardiocerebrovascular diseases. For example, recent studies suggested that abnormal expression of lncRNAs are closely related to the occurrence and progression of these diseases. LncRNAs regulate gene expression by specifically binding to mRNA to modulate disease progression, serving as biomarkers for the diagnosis and prognosis of cardiocerebrovascular diseases. In this review, we discuss the roles, mechanisms, and clinical value of lncRNAs in cardiocerebrovascular diseases, providing a new perspective for the diagnosis and treatment of the diseases.
Collapse
Affiliation(s)
- Hao Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Bo Liu
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
36
|
Ji YY, Meng M, Miao Y. lncRNA SNHG1 Promotes Progression of Cervical Cancer Through miR-195/NEK2 Axis. Cancer Manag Res 2020; 12:11423-11433. [PMID: 33204155 PMCID: PMC7665511 DOI: 10.2147/cmar.s277064] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/15/2020] [Indexed: 11/28/2022] Open
Abstract
Objective Cervical cancer is a common gynecologic cancer, and no study has been reported on the way through which lncRNA SNHG1, miR-195 and NEK2 jointly affect cervical cancer cells (CCCs), so this paper will explore a new approach to the development of cervical cancer in this respect. Methods Altogether 72 cervical cancer tissues and 54 adjacent tissues were collected. qPCR was performed to quantify lncRNA SNHG1 and miR-195, whose expression vectors were constructed and then transfected into CCCs, so as to observe their effects on the cells. Western blotting (WB) was carried out to detect protein levels. MTT assay was conducted to detect cell activity. Flow cytometry was performed to detect cell apoptosis. Transwell was carried out to detect cell invasion and migration. Results The expression of lncRNA SNHG1 up-regulated while that of miR-195 down-regulated in CCCs. lncRNA SNHG1 regulated NEK2 through its targeted binding to miR-195. The down-regulation of lncRNA SNHG1 or the up-regulation of miR-195 led to the decrease of NEK2 and the reduction of cells’ activity, migration and invasion, also resulting in the increase of cell apoptosis. Rescue experiments showed that the down-regulation of miR-195 could offset the cell changes caused by lncRNA SNHG1. Conclusion lncRNA SNHG1 promotes the progression of cervical cancer through the miR-195/NEK2 axis, so lncRNA SNHG1, miR-195 and NEK2 may have potential values for diagnosing and treating cervical cancer.
Collapse
Affiliation(s)
- Yuan Yuan Ji
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Man Meng
- Department of Geriatrics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Ye Miao
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People's Republic of China
| |
Collapse
|
37
|
Pang Q, Wang Y, Bi D, Lu H. LRRC75A-AS1 targets miR-199b-5p/PDCD4 axis to repress multiple myeloma. Cancer Biol Ther 2020; 21:1051-1059. [PMID: 33131397 DOI: 10.1080/15384047.2020.1831373] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Multiple functions of miR-199b-5p in diseases have been demonstrated by existing studies. However, never has the correlation between miR-199b-5p and multiple myeloma (MM) been established. METHODS qRT-PCR analyzed RNA expression and western blot measured protein expression. Cell proliferation ability was tested via colony formation and EdU assays, and apoptosis was determined via TUNEL, flow cytometry and detection of apoptosis-related proteins. Position of LRRC75A antisense RNA 1 (LRRC75A-AS1) was recognized by FISH assay. RIP, RNA pull-down and luciferase reporter experiments explored the molecular interplay. RESULTS GEO (Gene Expression Omnibus) data revealed miR-199b-5p upregulation in MM specimens, and qRT-PCR data verified miR-199b-5p upregulation in MM cells. Inhibiting miR-199b-5p markedly impeded MM cell proliferation and stimulated apoptosis. Moreover, we demonstrated the mechanism that miR-199b-5p was decoyed by LRRC75A-AS1 and miR-199b-5p targeted programmed cell death 4 (PDCD4) to repress its expression. Further, LRRC75A-AS1 was verified to repress proliferation and prompt apoptosis in a PDCD4-dependent way in MM cells. CONCLUSION Our data displayed that miR-199b-5p was sequestered by LRRC75A-AS1 so that PDCD4 was released to repress MM, implying the targeting miR-199b-5p as a novel thought for improving MM therapy.
Collapse
Affiliation(s)
- Quantang Pang
- Department of Orthopaedics, Rongcheng People's Hospital of Shandong Province , Rongcheng, Shandong, China
| | - Yanyan Wang
- Department of Neurology, Rongcheng People's Hospital of Shandong Province , Rongcheng, Shandong, China
| | - Dapeng Bi
- Department of Orthopaedics, Jining Second People's Hospital of Shandong Province , Jining, Shandong, China
| | - Hongyu Lu
- Department of Neurology, Rongcheng People's Hospital of Shandong Province , Rongcheng, Shandong, China
| |
Collapse
|
38
|
Construction and Analysis of lncRNA-Mediated ceRNA Network in Nasopharyngeal Carcinoma Based on Weighted Correlation Network Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1468980. [PMID: 33102573 PMCID: PMC7569441 DOI: 10.1155/2020/1468980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/12/2020] [Accepted: 09/25/2020] [Indexed: 12/24/2022]
Abstract
Increasing evidence indicated that aberrant expression of long noncoding RNAs (lncRNAs) are involved in tumorigenesis of nasopharyngeal carcinoma (NPC). The purpose of this study was to construct a lncRNA-mediated ceRNA network based on weighted correlation network analysis (WGCNA). First, modules with highly correlated genes were identified from GSE102349 via WGCNA, and the preservation of the modules was evaluated by GSE68799. Then, the differentially expressed lncRNAs and mRNAs identified from GSE12452 which belonged to the same WGCNA modules and the differentially expressed miRNAs identified from GSE32960 were used to construct a ceRNA network. The prognostic value of the network was evaluated by survival analysis. Furthermore, a risk score model for predicting progression-free survival (PFS) of NPC patients was established via LASSO-penalized Cox regression, and the differences in the expression of the lncRNAs between high- and low-risk groups were investigated. Finally, 14 stable modules were identified, and a ceRNA network composed of 11 lncRNAs, 15 miRNAs, and 40 mRNAs was established. The lncRNAs and mRNAs in the network belonged to the turquoise and salmon modules. Survival analysis indicated that ZNF667-AS1, LDHA, LMNB2, TPI1, UNG, and hsa-miR-142-3p were significantly correlated with the prognosis of NPC. Gene set enrichment analysis indicated that the upregulation of ZNF667-AS1 was associated with some immune-related pathways. Besides, a risk score model consisting of 12 genes was constructed and showed a good performance in predicting PFS for NPC patients. Among the 11 lncRNAs in the ceRNA network, SNHG16, SNHG17, and THAP9-AS1 were upregulated in the high-risk group of NPC, while ZNF667-AS1 was downregulated in the high-risk group of NPC. These results will promote our understanding of the crosstalk among lncRNAs, miRNAs, and mRNAs in the tumorigenesis and progression of NPC.
Collapse
|
39
|
Xiang J, Fu HQ, Xu Z, Fan WJ, Liu F, Chen B. lncRNA SNHG1 attenuates osteogenic differentiation via the miR‑101/DKK1 axis in bone marrow mesenchymal stem cells. Mol Med Rep 2020; 22:3715-3722. [PMID: 32901867 PMCID: PMC7533455 DOI: 10.3892/mmr.2020.11489] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
The imbalance induced by inhibition of bone mesenchymal stem cell (BMSC) osteogenic differentiation results in osteoporosis (OP); however, the underlying regulatory mechanism is not completely understood. Long non-coding RNAs (lncRNAs) serve crucial roles in osteogenic differentiation; therefore, investigating their regulatory role in the process of osteogenic differentiation may identify a promising therapeutic target for OP. The expression of small nucleolar RNA host gene 1 (SNHG1), Dickkopf 1 (DKK1), microRNA (miR)-101, RUNX family transcription factor 2 (RUNX2), osteopontin (OPN) and osteocalin (OCN) were detected via reverse transcription-quantitative PCR. The protein expression levels of DKK1, β-catenin, RUNX2, OPN, OCN, osterix and collagen type I α1 chain were analyzed by performing western blotting. The osteoblastic phenotype was assessed by conducting alkaline phosphatase activity detection and Alizarin Red staining. The interaction between SNHG1 and miR-101 was validated by bioinformatics and luciferase assays. The regulatory role of SNHG1 in BMSC osteogenic differentiation was assessed. SNHG1 expression was downregulated in a time-dependent manner during the process of osteogenic differentiation. SNHG1 overexpression inhibited osteogenic differentiation compared with the pcDNA group. The results indicated that SNHG1 and DKK1 directly interacted with miR-101. Moreover, SNHG1 regulated the Wnt/β-catenin signaling pathway to inhibit osteogenic differentiation via the miR-101/DKK1 axis. The present study indicated that lncRNA SNHG1 could attenuate BMSC osteogenic differentiation via the miR-101/DKK1 axis as a competitive endogenous RNA. Therefore, the present study furthered the current understanding of the potential mechanism underlying lncRNAs in in osteogenic differentiation.
Collapse
Affiliation(s)
- Jie Xiang
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hai-Qing Fu
- Department of Orthopedics and Traumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhun Xu
- Department of Orthopedics and Traumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei-Jie Fan
- Department of Orthopedics and Traumatology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Fei Liu
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Bin Chen
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
40
|
Li Q, Jiang B, Qi Y, Zhang H, Ma H. Long non-coding RNA SLCO4A1-AS1 drives the progression of non-small-cell lung cancer by modulating miR-223-3p/IKKα/NF-κB signaling. Cancer Biol Ther 2020; 21:806-814. [PMID: 32687454 DOI: 10.1080/15384047.2020.1787757] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Globally, lung cancer is known as a major cause of cancer-associated death and non-small-cell lung cancer (NSCLC) accounts for majority of all cases. Growing evidence has emerged that long non-coding RNAs (lncRNAs) act as vital regulatory molecules in various malignancies. Nevertheless, the function of SLCO4A1 antisense RNA 1(SLCO4A1-AS1) in NSCLC is vague. This study intended to investigate the biological role and probable regulatory mechanism of SLCO4A1-AS1 in NSCLC. qRT-PCR revealed that SLCO4A1-AS1 level was upregulated in NSCLC. Function assays manifested that silence of SLCO4A1-AS1 attenuated NSCLC cell proliferation, migration and invasion but promoted NSCLC cell apoptosis. Furthermore, we disclosed that SLCO4A1-AS1 activated NF-κB pathway in NSCLC, and that IKKα, an NF-κB pathway-related gene, possessed an enhanced level in NSCLC tissues and cells. Importantly, miR-223-3p bound with SLCO4A1-AS1 and IKKα. Further, SLCO4A1-AS1 competitively bound with miR-223-3p to increase IKKα expression, thereby activating NF-κB signaling pathway. In conclusion, SLCO4A1-AS1 drove NSCLC progression by activating NF-κB signaling pathway via sponging miR-223-3p to enhance IKKα expression. Thus, SLCO4A1-AS1 might be a promising biomarker for NSCLC treatment.
Collapse
Affiliation(s)
- Qingpeng Li
- Thoracic Surgery Department, The First Affiliated Hospital of Suzhou University , Suzhou, Jiangsu, China.,Thoracic Surgery Department, The Affiliated Hospital of Xuzhou Medical University , Xuzhou, Jiangsu, China
| | - Bo Jiang
- Thoracic Surgery Department, The Affiliated Hospital of Xuzhou Medical University , Xuzhou, Jiangsu, China
| | - Yang Qi
- Thoracic Surgery Department, The Affiliated Hospital of Xuzhou Medical University , Xuzhou, Jiangsu, China
| | - Hu Zhang
- Thoracic Surgery Department, The Affiliated Hospital of Xuzhou Medical University , Xuzhou, Jiangsu, China
| | - Haitao Ma
- Thoracic Surgery Department, The First Affiliated Hospital of Suzhou University , Suzhou, Jiangsu, China
| |
Collapse
|
41
|
Qiongna D, Jiafeng Z, Yalin H, Ping H, Chuan Z, Xiaojie J, Miaomiao Z, Yiting S, Hui Z. Implication of hsa_circ_0028007 in reinforcing migration, invasion, and chemo-tolerance of nasopharyngeal carcinoma cells. J Clin Lab Anal 2020; 34:e23409. [PMID: 32524687 PMCID: PMC7521330 DOI: 10.1002/jcla.23409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Background Given the reliability of circRNAs in symbolizing cancer progression, this investigation was designed to expound the involvement of hsa_circ_0028007 in regulating chemosensitivity of nasopharyngeal carcinoma (NPC) cells. Methods Altogether, 241 pairs of NPC tissues and para‐cancerous normal tissues were collected to identify NPC‐symbolic circRNAs, which have been screened by circRNA microarray in advance. Expressions of the circRNAs were determined by means of real‐time polymerase chain reaction (PCR). Besides, human NPC cell lines (ie, CNE2 and HONE1) were transfected by si‐hsa_circ_0028007 and si‐NC. Scratch assay, transwell assay, and MTT assay were performed to assess migration, invasion, and paclitaxel/cisplatin‐resistance of NPC cell lines. Results Hsa_circ_0028007 expression was abnormally heightened within NPC tissues in comparison with matched non‐tumor tissues (P < .05). Over‐expressed hsa_circ_0028007 was strongly associated with advanced (III‐IV) tumor stage, aggressive infiltration, and metastatic lymph nodes of NPC patients (P < .05). Regarding in vitro experiments, hsa_circ_0028007 expression was elevated in CNE2 and HONE1 cell lines as compared with HENE cell line (P < .05). Silencing of hsa_circ_0028007 not merely sensitized CNE2 and HONE1 cells against paclitaxel and cisplatin (P < .05), but also significantly repressed migration and invasion of the cell lines (P < .05). Conclusion Hsa_circ_0028007 was involved in facilitating progression and chemo‐resistance of NPC, which might offer an alternative for NPC treatment.
Collapse
Affiliation(s)
- Dong Qiongna
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhang Jiafeng
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hao Yalin
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - He Ping
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhou Chuan
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jin Xiaojie
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhao Miaomiao
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shao Yiting
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhao Hui
- Department of Otorhinolaryngology (South Campus), Ren Ji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
Li J, Zeng T, Li W, Wu H, Sun C, Yang F, Yang M, Fu Z, Yin Y. Long non-coding RNA SNHG1 activates HOXA1 expression via sponging miR-193a-5p in breast cancer progression. Aging (Albany NY) 2020; 12:10223-10234. [PMID: 32497022 PMCID: PMC7346023 DOI: 10.18632/aging.103123] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/24/2020] [Indexed: 01/04/2023]
Abstract
Breast cancer is the leading cause of cancer death in women worldwide. Long non-coding RNA small nucleolar RNA host gene 1 (SNHG1) has been reported to be involved in human diseases, including cancer. Here, we found that SNHG1 expression was significantly upregulated in human breast cancer tissues and cell lines. We explored the function of SNHG1 in breast cancer cells using in vitro and in vivo experiments and found that SNHG1 promotes breast cancer metastasis and proliferation. The potential molecular mechanism of SNHG1 in breast cancer cells may involve SNHG1 acting as a sponge of miR-193a-5p to activate the expression of the oncogene HOXA1. In summary, our study reveals a novel SNHG1/miR-193a-5p/HOXA1 competing endogenous RNA regulatory pathway in breast cancer progression and may provide new strategies for breast cancer therapy.
Collapse
Affiliation(s)
- Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Tianyu Zeng
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chunxiao Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Fan Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengzhu Yang
- Department of Geriatric Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ziyi Fu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Nanjing Maternal and Child Health Medical Institute, Nanjing Maternal and Child Health Care Hospital, Gynecology and Obstetrics Hospital Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
43
|
Orlandella FM, Mariniello RM, Mirabelli P, De Stefano AE, Iervolino PLC, Lasorsa VA, Capasso M, Giannatiempo R, Rongo M, Incoronato M, Messina F, Salvatore M, Soricelli A, Salvatore G. miR-622 is a novel potential biomarker of breast carcinoma and impairs motility of breast cancer cells through targeting NUAK1 kinase. Br J Cancer 2020; 123:426-437. [PMID: 32418991 PMCID: PMC7403386 DOI: 10.1038/s41416-020-0884-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 04/03/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Aberrant expression of microRNAs (miR) has been proposed as non-invasive biomarkers for breast cancers. The aim of this study was to analyse the miR-622 level in the plasma and in tissues of breast cancer patients and to explore the role of miR-622 and its target, the NUAK1 kinase, in this context. METHODS miR-622 expression was analysed in plasma and in tissues samples of breast cancer patients by q-RT-PCR. Bioinformatics programs, luciferase assay, public dataset analysis and functional experiments were used to uncover the role of miR-622 and its target in breast cancer cells. RESULTS miR-622 is downregulated in plasma and in tissues of breast cancer patients respect to healthy controls and its downregulation is significantly associated with advanced grade and high Ki67 level. Modulation of miR-622 affects the motility phenotype of breast cancer cells. NUAK1 kinase is a functional target of miR-622, it is associated with poor clinical outcomes of breast cancer patients and is inversely correlated with miR-622 level. CONCLUSIONS miR-622/NUAK1 axis is deregulated in breast cancer patients and affects the motility phenotype of breast cancer cells. Importantly, miR-622 and NUAK1 hold promises as biomarkers and as targets for breast cancers.
Collapse
Affiliation(s)
| | - Raffaela Mariarosaria Mariniello
- Dipartimento di Scienze Motorie e del Benessere, Universita' degli Studi di Napoli "Parthenope", Via Medina 40, 80133, Naples, Italy.,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | | | - Anna Elisa De Stefano
- Dipartimento di Scienze Motorie e del Benessere, Universita' degli Studi di Napoli "Parthenope", Via Medina 40, 80133, Naples, Italy.,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Paola Lucia Chiara Iervolino
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy.,Dipartimento di Scienze Biomediche Avanzate, Universita' "Federico II", Via Pansini 5, 80131, Napoli, Italy
| | - Vito Alessandro Lasorsa
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Mario Capasso
- IRCCS SDN, Via Emanuele Gianturco 113, 80143, Naples, Italy.,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | | | - Maria Rongo
- IRCCS SDN, Via Emanuele Gianturco 113, 80143, Naples, Italy
| | | | | | | | - Andrea Soricelli
- IRCCS SDN, Via Emanuele Gianturco 113, 80143, Naples, Italy.,Dipartimento di Scienze Motorie e del Benessere, Universita' degli Studi di Napoli "Parthenope", Via Medina 40, 80133, Naples, Italy
| | - Giuliana Salvatore
- IRCCS SDN, Via Emanuele Gianturco 113, 80143, Naples, Italy. .,Dipartimento di Scienze Motorie e del Benessere, Universita' degli Studi di Napoli "Parthenope", Via Medina 40, 80133, Naples, Italy. .,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Naples, Italy.
| |
Collapse
|
44
|
Xiong X, Feng Y, Li L, Yao J, Zhou M, Zhao P, Huang F, Zeng L, Yuan L. Long non‑coding RNA SNHG1 promotes breast cancer progression by regulation of LMO4. Oncol Rep 2020; 43:1503-1515. [PMID: 32323846 PMCID: PMC7107776 DOI: 10.3892/or.2020.7530] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) was reported to be a critical regulator of tumorigenesis and is frequently deregulated in several cancer types. However, the exact mechanism by which SNHG1 contributes to breast cancer progression has not been fully elucidated. The identification of the molecular mechanism of SNHG1 is important for understanding the development of breast cancer and for improving the prognosis of the patients with this disease. In the present study, increased expression levels of SNHG1 were noted in breast cancer tumors following analysis of differentially expressed lncRNAs between 1,063 tumor and 102 normal tissues derived from The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) dataset. This finding was further validated using 50 pairs of normal and tumor tissues that were collected from patients with breast cancer. Notably, SNHG1 expression was significantly correlated with estrogen receptor (ER)/progesterone receptor (PR) negative status (ER−/PR−) and advanced clinical stage in breast cancer tissues. Knockdown of SNHG1 led to cell growth arrest, cell cycle redistribution and cell migration inhibition of breast cancer cells. The miRDB database predicted that miR-573 interacts with SNHG1. RT-PCR confirmed the negative regulation of miR-573 levels by SNHG1 in breast cancer cells and the Dual-luciferase reporter assay confirmed their complementary binding. The repression of miR-573 by SNGH1 decreased LIM domain only 4 (LMO4) mRNA and protein expression levels in the breast cancer cell lines tested and induced the expression of cyclin D1 and cyclin E. In vitro experiments indicated that LMO4 overexpression could reverse siSNHG1-induced cell growth arrest, cell cycle redistribution and inhibition of cell migration in breast cancer cells. Moreover, the tumor xenograft model indicated that SNHG1 knockdown inhibited MDA-MB-231 growth in vivo and LMO4 overexpression reversed the tumor growth inhibition induced by SNHG1 knockdown. The present study demonstrated that SNHG1 acts as a novel oncogene in breast cancer via the SNHG/miR-573/LMO4 axis and that it could be a promising therapeutic target for patients with breast cancer.
Collapse
Affiliation(s)
- Xiang Xiong
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yeqian Feng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Lun Li
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Jia Yao
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Meirong Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Piao Zhao
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Feilong Huang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Liyun Zeng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Liqin Yuan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
45
|
Long non-coding RNA ATB promotes human non-small cell lung cancer proliferation and metastasis by suppressing miR-141-3p. PLoS One 2020; 15:e0229118. [PMID: 32092085 PMCID: PMC7039450 DOI: 10.1371/journal.pone.0229118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/24/2020] [Indexed: 01/22/2023] Open
Abstract
Long noncoding RNA activated by transforming growth factor-β (lncRNA-ATB) plays a critical role in progression of several cancers. In this study, lncRNA-ATB was significantly up-regulated in NSCLC tissues and cell lines, and high lncRNA-ATB expression indicated poor prognosis. Knockdown of lncRNA-ATB suppressed NSCLC cell growth, colony formation, migration, invasion and reversed epithelial-mesenchymal transition. In vivo study showed that silencing lncRNA-ATB inhibited tumor growth. Further mechanism studies demonstrated that lncRNA-ATB was a target of miR-141-3p. MiR-141-3p expression was negatively related to lncRNA-ATB expression in NSCLC tissues. These results suggested that inhibiting lncRNA-ATB might be an approach for NSCLC treatment.
Collapse
|
46
|
Liao B, Wang Z, Zhu Y, Wang M, Liu Y. Long noncoding RNA DRAIC acts as a microRNA-122 sponge to facilitate nasopharyngeal carcinoma cell proliferation, migration and invasion via regulating SATB1. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3585-3597. [PMID: 31497998 DOI: 10.1080/21691401.2019.1656638] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Increasing evidences have revealed that long noncoding RNAs (lncRNAs) are frequently involved in various cancers. However, the expression and function of lncRNA DRAIC in nasopharyngeal carcinoma (NPC) remain unknown. In this study, we found that DRAIC was significantly increased in NPC tissues. Increased expression of DRAIC was positively correlated with advanced clinical stages of NPC patients. Functional assays revealed that ectopic expression of DRAIC enhances NPC cell growth, migration and invasion. DRAIC knockdown represses NPC cell growth, migration and invasion. Mechanistically, we identified two miR-122 binding sites on DRAIC. RNA pull-down, RNA immunoprecipitation, and dual-luciferase reporter assays confirmed the binding of DRAIC to miR-122. Via binding of miR-122, DRAIC upregulated the expression of miR-122 target SATB1, which was abolished by the mutation of miR-122 binding sites on SATB1. Moreover, the oncogenic roles of DRAIC on NPC were reversed by the mutation of miR-122 binding sites on SATB1, simultaneous overexpression of miR-122, or depletion of SATB1. In addition, the expression of SATB1 was also increased and positively associated with that of DRAIC in NPC tissues. In conclusion, these findings revealed the important roles of DRAIC-miR-122-SATB1 axis in NPC and suggested that DRAIC may be a potential therapeutic target for NPC.
Collapse
Affiliation(s)
- Bing Liao
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| | - Zhi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| | - Yaqiong Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| | - Meiqun Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| | - Yuehui Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| |
Collapse
|
47
|
Li HM, Yu YK, Liu Q, Wei XF, Zhang J, Zhang RX, Sun HB, Wang ZF, Xing WQ, Li Y. LncRNA SNHG1 Regulates the Progression of Esophageal Squamous Cell Cancer by the miR-204/HOXC8 Axis. Onco Targets Ther 2020; 13:757-767. [PMID: 32158227 PMCID: PMC6986417 DOI: 10.2147/ott.s224550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022] Open
Abstract
Objective Long noncoding RNA small nucleolar RNA host gene 1 (SNHG1) has been reported to be aberrantly expressed and plays an important role in human cancers, including esophageal squamous cell cancer. However, the regulatory mechanism underlying SNHG1 in the progression of esophageal squamous cell cancer is poorly defined. Materials and Methods Fifty-three esophageal squamous cell cancer patients were recruited and overall survival was analyzed. EC9706 and KYSE150 cells were cultured for study in vitro. The expression levels of SNHG1, microRNA (miR)-204 and homeobox c8 (HOXC8) were detected by quantitative real-time polymerase chain reaction and Western blot. Cell cycle distribution, apoptosis, migration and invasion were determined by flow cytometry and transwell assays, respectively. The target interaction among SNHG1, miR-204 and HOXC8 was validated by luciferase reporter assay and RNA immunoprecipitation. Xenograft model was established to investigate the role of SNHG1 in vivo. Results High expression of SNHG1 was exhibited in esophageal squamous cell cancer and indicated poor outcomes of patients. SNHG1 silence led to cell cycle arrest at G0-G1 phase, inhibition of migration and invasion and increase of apoptosis. miR-204 was validated to sponge by SNHG1 and target HOXC8 in esophageal squamous cell cancer cells. miR-204 knockdown or HOXC8 restoration reversed the inhibitive role of SNHG1 silence in the progression of esophageal squamous cell cancer cells. Furthermore, inhibiting SNHG1 decreased xenograft tumor growth by regulating miR-204 and HOXC8. Conclusion SNHG1 knockdown suppresses migration and invasion but induces apoptosis of esophageal squamous cell cancer cells by increasing miR-204 and decreasing HOXC8.
Collapse
Affiliation(s)
- Hao Miao Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yong Kui Yu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Qi Liu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xiu Feng Wei
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jun Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Rui Xiang Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hai Bo Sun
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zong Fei Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Wen Qun Xing
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yin Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Department of Thoracic Surgery, The Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
48
|
Yang H, Jiang Z, Wang S, Zhao Y, Song X, Xiao Y, Yang S. Long non-coding small nucleolar RNA host genes in digestive cancers. Cancer Med 2019; 8:7693-7704. [PMID: 31691514 PMCID: PMC6912041 DOI: 10.1002/cam4.2622] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 08/21/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022] Open
Abstract
Although long noncoding RNAs (lncRNAs) do not have protein coding capacities, they are involved in the pathogenesis of many types of cancers, including hepatocellular carcinoma, cervical cancer, and gastric cancer. Notably, the roles of lncRNAs are vital in nearly every aspect of tumor biology. Long non-coding small nucleolar RNA host genes (lnc-SNHGs) are abnormally expressed in multiple cancers, including urologic neoplasms, respiratory tumors, and digestive cancers, and play vital roles in these cancers. These host genes could participate in tumorigenesis by regulating proliferation, migration, invasion and apoptosis of tumor cells. This review focuses on the overview of the roles that lnc-SNHGs play in the formation and progression of digestive cancers.
Collapse
Affiliation(s)
- Huan Yang
- Department of GastroenterologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Zheng Jiang
- Department of GastroenterologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Shuang Wang
- Department of GastroenterologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of GastroenterologyPeople's Hospital of Changshou ChongqingChongqingChina
| | - Yongbing Zhao
- Department of GastroenterologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of GastroenterologyPeople's Hospital of Changshou ChongqingChongqingChina
| | - Xiaomei Song
- Department of GastroenterologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of GastroenterologyPeople's Hospital of Changshou ChongqingChongqingChina
| | - Yufeng Xiao
- Department of GastroenterologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Shiming Yang
- Department of GastroenterologyXinqiao HospitalArmy Medical UniversityChongqingChina
| |
Collapse
|
49
|
Chen M, Xu Z, Zhang Y, Zhang X. LINC00958 Promotes The Malignancy Of Nasopharyngeal Carcinoma By Sponging microRNA-625 And Thus Upregulating NUAK1. Onco Targets Ther 2019; 12:9277-9290. [PMID: 31819474 PMCID: PMC6842770 DOI: 10.2147/ott.s216342] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose The aberrant expression of long noncoding RNAs (lncRNAs) indicates progression of various diseases. LINC00958 has been well studied in several types of human cancer; however, the expression profile, functions, and potential mechanism of action of this lncRNA in nasopharyngeal carcinoma (NPC) remain largely unclear and still need to be elucidated. In the present study, we aimed to measure LINC00958 expression in NPC, determine its clinical value, and explore its roles in NPC progression as well as the mechanisms behind these processes. Methods The expression profile of LINC00958 in NPC was evaluated by reverse-transcription quantitative polymerase chain reaction (RT-qPCR). A series of functional assays, including the Cell Counting Kit-8 assay, flow cytometry, a Transwell assay, and an in vivo nude mouse model, were utilized to determine the participation of LINC00958 in the malignancy of NPC. Results LINC00958 was found to be upregulated in NPC tissue specimens and cell lines. The LINC00958 overexpression significantly correlated with tumor size, lymph node status, TNM stage, and worse overall survival among NPC patients. Downregulation of LINC00958 suppressed NPC cell proliferation, migration, and invasion and induced apoptosis in vitro. Additionally, the LINC00958 knockdown impaired tumor growth in vivo. Mechanistically, LINC00958 was found to serve as a molecular sponge of microRNA-625 (miR-625), thereby upregulating NUAK family SNF1-like kinase 1 (NUAK1) in NPC cells. Lastly, rescue experiments validated the involvement of the miR-625–NUAK1 axis in LINC00958-mediated biological functions in NPC. Conclusion Our results demonstrated that LINC00958 works as an oncogene in NPC and plays a key role in the malignant phenotype of NPC cells by sponging miR-625 and increasing NUAK1 expression. The LINC00958–miR-625–NUAK1 pathway might be a target for anticancer therapy in patients with NPC.
Collapse
Affiliation(s)
- Meijuan Chen
- Department of Ophthalmology and Otorhinolaryngology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, People's Republic of China
| | - Zhina Xu
- Department of Ophthalmology and Otorhinolaryngology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, People's Republic of China
| | - Yingyao Zhang
- Department of Ophthalmology and Otorhinolaryngology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, People's Republic of China
| | - Xiujuan Zhang
- Weifang People's Hospital, Weifang, Shandong 262737, People's Republic of China
| |
Collapse
|
50
|
Zheng YJ, Zhao JY, Liang TS, Wang P, Wang J, Yang DK, Liu ZS. Long noncoding RNA SMAD5-AS1 acts as a microRNA-106a-5p sponge to promote epithelial mesenchymal transition in nasopharyngeal carcinoma. FASEB J 2019; 33:12915-12928. [PMID: 31557058 DOI: 10.1096/fj.201900803r] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant epithelial cancer of the head and neck with high prevalence in southern China, which is accompanied by notable invasiveness and metastasis. Long noncoding RNAs (lncRNAs) participate in the progression of various cancers including NPC. Microarray-based analysis identified highly expressed lncRNA mothers against decapentaplegic homolog 5 (SMAD5)-antisense RNA 1 (AS1) related to NPC. Interestingly, it is found that SMAD5-AS1 competitively bound to microRNA (miR)-106a-5p to regulate SMAD5. Herein, the study aimed to clarify the role of SMAD5-AS1/miR-106a-5p/SMAD5 axis in the process of epithelial mesenchymal transition (EMT) in NPC. SMAD5-AS1 was highly expressed and miR-106a-5p was poorly expressed in NPC tissues and cell lines. The NPC cells were treated with a series of small interfering RNAs, mimics, or inhibitors to explore the effects of SMAD5-AS1, SMAD5, and miR-106a-5p on EMT, cell proliferation, migration, and invasion in NPC. Of note, SMAD5-AS1 silencing or miR-106a-5p overexpression reduced expression of N-cadherin, matrix metallopeptidase 9, Snail, and Vimentin while elevating E-cadherin expression, thus inhibiting EMT, cell proliferation, migration, and invasion in NPC by down-regulation of SMAD5. Moreover, SMAD5 silencing could reduce the ability of EMT induced by SMAD5-AS1 up-regulation. SMAD5-AS1 silencing or miR-106a-5p elevation inhibited tumorigenesis in nude mice. Taken together, SMAD5-AS1 silencing suppressed EMT, cell proliferation, migration, and invasion in NPC by elevating miR-106a-5p to down-regulate SMAD5, which provided a novel therapeutic target for NPC treatment.-Zheng, Y.-J., Zhao, J.-Y., Liang, T.-S., Wang, P., Wang, J., Yang, D.-K., Liu, Z.-S. Long noncoding RNA SMAD5-AS1 acts as a microRNA-106a-5p sponge to promote epithelial mesenchymal transition in nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Ying-Juan Zheng
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing-Yi Zhao
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tian-Song Liang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ping Wang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juan Wang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dao-Ke Yang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhang-Suo Liu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|