1
|
Yang TF, Li XR, Kong MW. Molecular mechanisms underlying roles of long non-coding RNA small nucleolar RNA host gene 16 in digestive system cancers. World J Gastrointest Oncol 2024; 16:4300-4308. [PMID: 39554746 PMCID: PMC11551640 DOI: 10.4251/wjgo.v16.i11.4300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/13/2024] [Accepted: 07/02/2024] [Indexed: 10/25/2024] Open
Abstract
This editorial reviews the molecular mechanisms underlying the roles of the long non-coding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) in digestive system cancers based on two recent studies on lncRNAs in digestive system tumors. The first study, by Zhao et al, explored how hBD-1 affects colon cancer, via the lncRNA TCONS_00014506, by inhibiting mTOR and promoting autophagy. The second one, by Li et al, identified the lncRNA prion protein testis specific (PRNT) as a factor in oxaliplatin resistance by sponging ZNF184 to regulate HIPK2 and influence colorectal cancer progression and chemoresistance, suggesting PRNT as a potential therapeutic target for colorectal cancer. Both of these two articles discuss the mechanisms by which lncRNAs contribute to the development and progression of digestive system cancers. As a recent research hotspot, SNHG16 is a typical lncRNA that has been extensively studied for its association with digestive system cancers. The prevailing hypothesis is that SNHG16 participates in the development and progression of digestive system tumors by acting as a competing endogenous RNA, interacting with other proteins, regulating various genes, and affecting downstream target molecules. This review systematically examines the recently reported biological functions, related molecular mechanisms, and potential clinical significance of SNHG16 in various digestive system cancers, and explores the relationship between SNHG16 and digestive system cancers. The findings suggest that SNHG16 may serve as a potential biomarker and therapeutic target for human digestive system cancers.
Collapse
Affiliation(s)
- Ting-Fang Yang
- Department of Oncology, Guiqian International General Hospital, Guiyang 550018, Guizhou Province, China
| | - Xin-Rui Li
- Department of Cardiology, Guiqian International General Hospital, Guiyang 550018, Guizhou Province, China
| | - Mo-Wei Kong
- Department of Cardiology, Guiqian International General Hospital, Guiyang 550018, Guizhou Province, China
| |
Collapse
|
2
|
Khaleel AQ, Alshahrani MY, Rizaev JA, Malathi H, Devi S, Pramanik A, Mustafa YF, Hjazi A, Muazzamxon I, Husseen B. siRNA-based strategies to combat drug resistance in gastric cancer. Med Oncol 2024; 41:293. [PMID: 39428440 DOI: 10.1007/s12032-024-02528-w] [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: 08/17/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024]
Abstract
Chemotherapy is a key treatment option for gastric cancer, but over 50% of patients develop either inherent or acquired resistance to these drugs, resulting in a 5-year survival rate of only about 20%. The primary treatment for advanced gastric cancer typically involves chemotherapy based on platinum or fluorouracil. Several factors can contribute to platinum resistance, including decreased drug uptake, increased drug efflux or metabolism, enhanced DNA repair, activation of pro-survival pathways, and inhibition of pro-apoptotic pathways. In recent years, there has been significant progress in biology aimed at finding innovative and more effective methods to overcome chemotherapy resistance. Small interfering RNAs (siRNAs) have emerged as a significant advancement in gene expression regulation, showing promise in enhancing the sensitivity of gastric cancer cells to chemotherapy drugs. However, siRNA therapies still face major challenges, particularly in terms of stability and efficient delivery in vivo. This article discusses the advances in siRNA therapy and its potential role in overcoming resistance to chemotherapeutic drugs such as cisplatin, 5-FU, doxorubicin, and paclitaxel in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Abdulrahman Qais Khaleel
- Department of Medical Instruments Engineering, College of Engineering, University of Al Maarif, Ramadi, Al Anbar, 31001, Iraq.
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Jasur Alimdjanovich Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan.
| | - H Malathi
- Department of Biotechnology and Genetics, School of Sciences Jain (Deemed to be University), Bangalore, Karnataka, India
| | - Seema Devi
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjheri, Mohali, 140307, Punjab, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Ismoilova Muazzamxon
- Department of Propaedeutics of Internal Diseases, Fergana Medical Institute of Public Health, Fergana, Uzbekistan
- Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
3
|
Shi Y, Men X, Wang F, Li X, Zhang B. Role of long non-coding RNAs (lncRNAs) in gastric cancer metastasis: A comprehensive review. Pathol Res Pract 2024; 262:155484. [PMID: 39180802 DOI: 10.1016/j.prp.2024.155484] [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: 01/15/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024]
Abstract
One of the greatest frequent types of malignancy is gastric cancer (GC). Metastasis, an essential feature of stomach cancer, results in a high rate of mortality and a poor prognosis. However, metastasis biological procedures are not well recognized. Long non-coding RNAs (lncRNAs) have a role in numerous gene regulation pathways via epigenetic modification as well as transcriptional and post-transcriptional control. LncRNAs have a role in a variety of disorders, such as cardiovascular disease, Alzheimer's, and cancer. LncRNAs are substantially related to GC incidence, progression, metastasis and drug resistance. Several research released information on the molecular processes of lncRNAs in GC pathogenesis. By interacting with a gene's promoter or enhancer region to influence gene expression, lncRNAs can operate as an oncogene or a tumor suppressor. This review includes the lncRNAs associated with metastasis of GC, which may give insights into the processes as well as potential clues for GC predicting and tracking.
Collapse
Affiliation(s)
- Yue Shi
- Department of Microbiology and Immunology, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| | - Xiaoping Men
- Department of Clinical Laboratory, The First Affiliated Hospital to Changchun University of Chinese Medicine, Jilin 130021, PR China.
| | - Fang Wang
- Department of Microbiology and Immunology, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| | - Xueting Li
- Experimental Center, Changchun University of Chinese Medicine, Jilin 130021, PR China.
| | - Biao Zhang
- School of Health Management, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| |
Collapse
|
4
|
Ye H, Li MY, Shi RH. Advances in understanding of mechanism of long non-coding RNA SNHG16 in digestive system tumors. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:405-411. [DOI: 10.11569/wcjd.v32.i6.405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
|
5
|
Saeinasab M, Atlasi Y, M Matin M. Functional role of lncRNAs in gastrointestinal malignancies: the peculiar case of small nucleolar RNA host gene family. FEBS J 2024; 291:1353-1385. [PMID: 36282516 DOI: 10.1111/febs.16668] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Long noncoding RNAs (lncRNAs) play crucial roles in normal physiology and are often de-regulated in disease states such as cancer. Recently, a class of lncRNAs referred to as the small nucleolar RNA host gene (SNHG) family have emerged as important players in tumourigenesis. Here, we discuss new findings describing the role of SNHGs in gastrointestinal tumours and summarize the three main functions by which these lncRNAs promote carcinogenesis, namely: competing with endogenous RNAs, modulating protein function, and regulating epigenetic marking. Furthermore, we discuss how SNHGs participate in different hallmarks of cancer, and how this class of lncRNAs may serve as potential biomarkers in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
| | - Yaser Atlasi
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, UK
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Iran
| |
Collapse
|
6
|
Ghahramani Almanghadim H, Karimi B, Poursalehi N, Sanavandi M, Atefi Pourfardin S, Ghaedi K. The biological role of lncRNAs in the acute lymphocytic leukemia: An updated review. Gene 2024; 898:148074. [PMID: 38104953 DOI: 10.1016/j.gene.2023.148074] [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: 10/10/2023] [Revised: 11/29/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
The cause of leukemia, a common malignancy of the hematological system, is unknown. The structure of long non-coding RNAs (lncRNAs) is similar to mRNA but no ability to encode proteins. Numerous malignancies, including different forms of leukemia, are linked to Lnc-RNAs. It is verified that the carcinogenesis and growth of a variety of human malignancies are significantly influenced by aberrant lncRNA expression. The body of evidence linking various types of lncRNAs to the etiology of leukemia has dramatically increased during the past ten years. Some lncRNAs are therefore anticipated to function as novel therapeutic targets, diagnostic biomarkers, and clinical outcome predictions. Additionally, these lncRNAs may provide new therapeutic options and insight into the pathophysiology of diseases, particularly leukemia. Thus, this review outlines the present comprehension of leukemia-associated lncRNAs.
Collapse
Affiliation(s)
| | - Bahareh Karimi
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Negareh Poursalehi
- Department of Medical Biotechnology, School of Medicine Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | | | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Ave., Azadi Sq., 81746-73441 Isfahan, Iran.
| |
Collapse
|
7
|
Liu S, Rong Y, Tang M, Zhao Q, Li C, Gao W, Yang X. The Functions and Mechanisms of Long Non-coding RNA SNHGs in Gastric Cancer. Comb Chem High Throughput Screen 2024; 27:2639-2653. [PMID: 37842903 DOI: 10.2174/0113862073268591230928100922] [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: 06/22/2023] [Revised: 08/08/2023] [Accepted: 09/01/2023] [Indexed: 10/17/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. Despite significant advancements in surgical and adjuvant treatments, patient prognosis remains unsatisfactory. Long non-coding RNAs (lncRNAs) are a class of RNA molecules that lack protein-coding capacity but can engage in the malignant biological behaviors of tumors through various mechanisms. Among them, small nucleolar host genes (SNHGs) represent a subgroup of lncRNAs. Studies have revealed their involvement not only in gastric cancer cell proliferation, invasion, migration, epithelial- mesenchymal transition (EMT), and apoptosis but also in chemotherapy resistance and tumor stemness. This review comprehensively summarizes the biological functions, molecular mechanisms, and clinical significance of SNHGs in gastric cancer. It provides novel insights into potential biomarkers and therapeutic targets for the exploration of gastric cancer.
Collapse
Affiliation(s)
- Songhua Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou 730000, China
| | - Yao Rong
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou 730000, China
| | - Mingzheng Tang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou 730000, China
| | - Qiqi Zhao
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Chunyan Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, China
| | - Wenbin Gao
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou 730000, China
| | - Xiaojun Yang
- The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, China
- Gansu key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
- Gansu Research Center of Prevention and Control Project for Digestive Oncology, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Gastrointestinal Tumor Diagnosis and Treatment of National Health and Health Commission, Lanzhou, 730000, China
| |
Collapse
|
8
|
Hou W, Xu L, Su T, Wu Y, Liu Y, Wei Y. Hypoxia Induces Tumor-Derived Exosome SNHG16 to Mediate Nasopharyngeal Carcinoma Progression through the miR-23b-5p/MCM6 Pathway. Appl Biochem Biotechnol 2024; 196:275-295. [PMID: 37119503 DOI: 10.1007/s12010-023-04558-y] [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] [Accepted: 04/18/2023] [Indexed: 05/01/2023]
Abstract
This study aims to investigate the mechanism of tumor-derived exosomal (EVs) SNHG16 in promoting the progression of nasopharyngeal carcinoma (NPC). QRT-PCR was used to detect the expression of SNHG16, miR-23b-5p and MCM6 in NPC. MTT, flow cytometry and transwell were used to detect the effects of them on the proliferation, cycle, apoptosis and invasion ability of NPC. Transmission electron microscopy, Western blotting and BCA were used to verify the regulation of exosome secretion under different oxygen environments. Our results showed that hypoxia induces tumor-derived exosome SNHG16 to mediate NPC progression through the miR-23b-5p/MCM6 pathway.
Collapse
Affiliation(s)
- Wei Hou
- Department of Otolaryngology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Lu Xu
- Department of Otolaryngology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Tao Su
- Department of Otolaryngology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Yunxiao Wu
- Department of Neurology, Yang Ling Demonstration Zone Hospital, Yangling, 712100, Shaanxi, China
| | - Yujuan Liu
- Department of Otolaryngology, Yang Ling Demonstration Zone Hospital, Yangling, 712100, Shaanxi, China
| | - Yangao Wei
- Department of Otolaryngology, Yang Ling Demonstration Zone Hospital, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
9
|
Xia Y, Guan J, Lu X, Liu Y, Luan W. Novel lncRNA SNHG16 Promotes the Growth and Metastasis of Malignant Melanoma by Regulating miR-205-5p/PAK2 Axis. Clin Cosmet Investig Dermatol 2022; 15:1615-1625. [PMID: 35983126 PMCID: PMC9379127 DOI: 10.2147/ccid.s374404] [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: 05/22/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022]
Abstract
Background Long non-coding RNAs (lncRNAs) play an key role in the biological processes of various malignant tumors. SNHG16 has been confirmed to be associated with the progression of various cancers. However, function and molecular mechanism of SNHG16 in melanoma have not been studied by scholars. Methods The expression of SNHG16 in melanoma tissues were detected by using qRT-PCR. Melanoma cases from The Cancer Genome Atlas (TCGA) and GEO#GSE15605 were included in this study. CCK-8 assay, EdU assay, transwell and scratch wound assay were used to explore the role of SNHG16 in melanoma cells. Luciferase reporter assays and RNA pull-down assay were used to explore the molecular mechanism of SNHG16 in melanoma. Results Here, we found that SNHG16 was up-regulated in melanoma. SNHG16 enhances the growth and metastasis of melanoma. SNHG16 can promote the expression of P21-activated kinases 2 (PAK2) by sponging miR-205-5p. PAK2 is the target gene of miR-205-5p. We demonstrated that SNHG16 promotes the metastasis and growth of melanoma through miR-205-5p/PAK2 axis. Conclusion This study firstly confirmed the role and mechanism of SNHG16 in the occurrence and development of melanoma. Therefore, SNHG16 may become a key point in the diagnosis, prognosis and treatment of melanoma patients in the future.
Collapse
Affiliation(s)
- Yun Xia
- Department of Plastic Surgery, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu, People's Republic of China
| | - Jing Guan
- Department of Plastic Surgery, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu, People's Republic of China
| | - Xu Lu
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Yifan Liu
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Wenkang Luan
- Department of Plastic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| |
Collapse
|
10
|
Han Z, Zhuang X, Yang B, Jin L, Hong P, Xue J, Chen S, Tian Z. SYDE1 Acts as an Oncogene in Glioma and has Diagnostic and Prognostic Values. Front Mol Biosci 2021; 8:714203. [PMID: 34722629 PMCID: PMC8552071 DOI: 10.3389/fmolb.2021.714203] [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: 05/24/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022] Open
Abstract
Objectives: Gliomas remain one of serious public health problems worldwide which demand further and deeper investigation. The aim of this study was to explore the association between synapse defective protein 1 homolog 1 (SYDE1) and gliomas via public database analysis and in vitro validation to determine the potential diagnostic and prognostic values. Methods and Results: Compared with healthy brain tissues, there was a significant increase in SYDE1 expression in glioma tissues. Additionally, SYDE1 exhibited higher expression levels in glioma patients with unfavorable clinicopathological factors. In vitro knockdown of SYDE1 in glioma cell lines A172 inhibited their migrative and invasive ability but not the proliferative ability. GO and KEGG pathway analysis of the top 100 genes coexpressed with SYDE1 showed enrichments of tumor-associated terms. Further bioinformatic analysis revealed that the SNHG16/hsa-miR-520e/SYDE1 axis might be involved in glioma development. Conclusions: SYDE1 is expressed at higher levels in gliomas than in healthy brains, and can promote metastasis and invasion but not proliferation of gliomas. Furthermore, SYDE1 has values in the diagnosis and prognosis prediction of gliomas.
Collapse
Affiliation(s)
- Zhenyuan Han
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China.,Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China
| | - Xiaodong Zhuang
- Department of Stomatology, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, China
| | - Biao Yang
- Department of Neurosurgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Lihui Jin
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengjie Hong
- Department of Neurosurgery, Huashan Hospital of Fudan University, Shanghai, China
| | - Junqing Xue
- Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunjie Chen
- Department of Nephrology, Shanghai Fourth People's Hospital Affiliated to Tongji Universtiy, Shanghai, China
| | - Zhen Tian
- Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Weng W, Liu C, Li G, Ruan Q, Li H, Lin N, Chen G. Long non-‑coding RNA SNHG16 functions as a tumor activator by sponging miR‑373‑3p to regulate the TGF‑β‑R2/SMAD pathway in prostate cancer. Mol Med Rep 2021; 24:843. [PMID: 34643247 DOI: 10.3892/mmr.2021.12483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/14/2021] [Indexed: 01/27/2023] Open
Abstract
Long non‑coding RNAs (lncRNAs) are involved in the pathogenesis of prostate cancer (PCa) as competitive endogenous RNA. The present study aimed to investigate the molecular mech--anisms of lncRNA small nucleolar RNA host gene 16 (SNHG16) in the proliferation and metastasis of PCa cells. Cancer tissues and adjacent normal tissues were collected from 80 patients with PCa who did not receive any treatment. Reverse transcription‑quantitative PCR analysis was performed to detect the expression levels of SNHG16, hsa‑microRNA (miRNA/miR)‑373‑3p and transforming growth factor‑β receptor type 2 (TGF‑β‑R2), and Spearman's correlation coefficient analysis was performed to assess the correlations between these molecules. Furthermore, the effects of SNHG16 knockdown and overexpression on the biological functions of DU‑145 PCa cells and TGF‑β‑R2/SMAD signaling were analyzed. The dual‑luciferase reporter assay was performed to assess the associations between SNHG16 and miR‑373‑3p, and TGF‑β‑R2 and miR‑373‑3p, the effects of which were verified via rescue experiments. The results demonstrated that the expression levels of SNHG16 and TGF‑β‑R2 were significantly upregulated in PCa tissues, whereas miR‑373‑3p expression was significantly downregulated (P<0.001). In addition, negative correlations were observed between SNHG16 and miR‑373‑3p (rho, ‑0.631) and miR‑373‑3p and TGF‑β‑R2 (rho, ‑0.516). Overexpression of SNHG16 significantly promoted the proliferation, migration and invasion of PCa cells (P<0.05), and significantly increased the protein expression levels of TGF‑β‑R2, phosphorylated (p)‑SMAD2, p‑SMAD3, c‑Myc and E2F4 (P<0.001). Notably, the results revealed that miR‑373‑3p is a target of SNHG16, and miR‑373‑3p knockdown rescued short hairpin (sh)‑SNHG16‑suppressed cellular functions by promoting TGF‑β‑R2/SMAD signaling. The results also revealed that miR‑373‑3p targets TGF‑β‑R2. Notably, transfection with miR‑373‑3p inhibitor rescued sh‑TGF‑β‑R2‑suppressed cell proliferation and migration. Taken together, the results of the present study suggest that SNHG16 promotes the proliferation and migration of PCa cells by targeting the miR‑373‑3p/TGF‑β‑R2/SMAD axis.
Collapse
Affiliation(s)
- Wubin Weng
- Department of Urology, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian 355000, P.R. China
| | - Changming Liu
- Department of Urology, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian 355000, P.R. China
| | - Guomin Li
- Department of Urology, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian 355000, P.R. China
| | - Qiongfang Ruan
- Department of Respiratory Medicine, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian 355000, P.R. China
| | - Huizhang Li
- Department of Urology, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian 355000, P.R. China
| | - Ningfeng Lin
- Department of Urology, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian 355000, P.R. China
| | - Guangbing Chen
- Department of Urology, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian 355000, P.R. China
| |
Collapse
|
12
|
Moghbeli M. Molecular interactions of miR-338 during tumor progression and metastasis. Cell Mol Biol Lett 2021; 26:13. [PMID: 33827418 PMCID: PMC8028791 DOI: 10.1186/s11658-021-00257-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cancer, as one of the main causes of human deaths, is currently a significant global health challenge. Since the majority of cancer-related deaths are associated with late diagnosis, it is necessary to develop minimally invasive early detection markers to manage and reduce mortality rates. MicroRNAs (miRNAs), as highly conserved non-coding RNAs, target the specific mRNAs which are involved in regulation of various fundamental cellular processes such as cell proliferation, death, and signaling pathways. MiRNAs can also be regulated by long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs). They are highly stable in body fluids and have tumor-specific expression profiles, which suggest their suitability as efficient non-invasive diagnostic and prognostic tumor markers. Aberrant expression of miR-338 has been widely reported in different cancers. It regulates cell proliferation, migration, angiogenesis, and apoptosis in tumor cells. Main body In the present review, we have summarized all miR-338 interactions with other non-coding RNAs (ncRNAs) and associated signaling pathways to clarify the role of miR-338 during tumor progression. Conclusions It was concluded that miR-338 mainly functions as a tumor suppressor in different cancers. There were also significant associations between miR-338 and other ncRNAs in tumor cells. Moreover, miR-338 has a pivotal role during tumor progression using the regulation of WNT, MAPK, and PI3K/AKT signaling pathways. This review highlights miR-338 as a pivotal ncRNA in biology of tumor cells.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Yang B, Wang X, Ying C, Peng F, Xu M, Chen F, Cai B. Long Noncoding RNA SNHG16 Facilitates Abdominal Aortic Aneurysm Progression through the miR-106b-5p/STAT3 Feedback Loop. J Atheroscler Thromb 2021; 28:66-78. [PMID: 32612026 PMCID: PMC7875146 DOI: 10.5551/jat.52274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 03/01/2020] [Indexed: 12/11/2022] Open
Abstract
AIM Deepening our understanding of the molecular mechanism of abdominal aortic aneurysm (AAA) progression will help set up novel avenues for therapeutic target identification. Our aim here was to unveil the mechanism function of STAT3 in AAA progression. METHODS We investigated the functional role of STAT3 in AAA by evaluating vascular smooth muscle cell (VSMC) apoptosis and proliferation via terminal deoxynucleotidyl transferase dUTP nick end labeling, western blotting, 5-ethynyl-2´-deoxyuridine, and Cell Counting Kit-8 assays. The interplay of lncRNA-miRNA-mRNA was verified using the luciferase reporter assay and the RNA pull-down, RNA immunoprecipitation, and chromatin immunoprecipitation assays. Quantitative real-time polymerase chain reaction and western blot were utilized to quantitate the RNA and protein levels of the indicated molecules. RESULTS Inhibition of STAT3 facilitated VSMC proliferation and repressed VSMC apoptosis. Moreover, It was demonstrated that small nucleolar RNA host gene 16 (SNHG16) sponged miR-106b-5p to release STAT3 from the inhibitory effect of miR-106b-5p. SNHG16 led to the upregulation of STAT3, and STAT3 was an upstream factor in the activation of SNHG16 transcription. Moreover, rescue experiments indicated that SNHG16 depended on STAT3 to regulate VSMC apoptosis and proliferation. In vivo assays showed that SNHG16 knockdown retarded the formation of AAA and upregulated STAT3 in vivo. CONCLUSIONS We identified that SNHG16/miR-106b-5p/STAT3 formed a complex circuitry for the deterioration of AAA via regulating VSMCs, suggesting a possible target for the pathogenesis of AAA.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/pathology
- Apoptosis
- Cell Line
- Gene Deletion
- Gene Knockdown Techniques
- Humans
- Mice, Inbred C57BL
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- RNA, Long Noncoding/genetics
- STAT3 Transcription Factor/genetics
- Transcriptional Activation
- Up-Regulation
- Mice
Collapse
Affiliation(s)
- Baihui Yang
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xi Wang
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chenyong Ying
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fei Peng
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ming Xu
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Feiyun Chen
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bing Cai
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
14
|
Huang L, Ding Y, Yang L, Jiang X, Xia Z, You Z. The effect of LncRNA SNHG16 on vascular smooth muscle cells in CHD by targeting miRNA-218-5p. Exp Mol Pathol 2020; 118:104595. [PMID: 33359036 DOI: 10.1016/j.yexmp.2020.104595] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 01/21/2023]
Abstract
PURPOSE To explore the role of SNHG16 in coronary heart disease (CHD) and its effect on vascular smooth muscle cells via miR-218-5p. METHODS A quantitative real time polymerase chain reaction (qRT-PCR) assay was carried out to determine the expression of serum SNHG16 and miR-218-5p in the observation group before and after treatment and in the control group. Then, receiver operating characteristic (ROC) curves were drawn to analyze the value of SNHG16 and miR-218-5p in the diagnosis and prognosis prediction of CHD. Furthermore, purchased coronary artery smooth muscle cells (HCASMC) were transfected with SNHG16 mimics, SNHG16 inhibitor, miR-218-5p mimics, miR-218-5p inhibitor, or negative control, and then the cell proliferation, migration, apoptosis, and apoptosis-related proteins (Bax, Bcl-2, and Caspase-3) and Wnt/β-catenin signaling pathway-related proteins (c-myc and β-catenin) in the cells were detected. RESULTS Both SNHG16 and miR-218-5 had good predictive value for the development and recurrence of CHD (P < 0.001). In addition, cell experiments showed that inhibition of SNHG16 weakened the proliferation and migration of HCASMC cells and intensified their apoptosis, SNHG16 and miR-218-5p had the same binding sites, and the dual luciferase reporter assay revealed that the fluorescence activity of HG16-WT was inhibited by transfected miR-mimics, but enhanced by transfected miR-inhibitor (both P < 0.050). Furthermore, the rescue experiment revealed that the effect of inhibiting SNHG16 on HCASMC cells was completely reversed by miR-218-5p (P > 0.050). CONCLUSIONS Highly expressed SNHG16 targetedly regulates miR-218-5p and promotes the proliferation and migration of HCASMC via the Wnt/β-catenin signaling pathway, giving rise to CHD.
Collapse
Affiliation(s)
- Lin Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of NanChang University, Nanchang 330006, Jiangxi Province, China
| | - Ying Ding
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of NanChang University, Nanchang 330006, Jiangxi Province, China
| | - Lu Yang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of NanChang University, Nanchang 330006, Jiangxi Province, China
| | - Xinghua Jiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of NanChang University, Nanchang 330006, Jiangxi Province, China
| | - Zhen Xia
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of NanChang University, Nanchang 330006, Jiangxi Province, China
| | - Zhigang You
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of NanChang University, Nanchang 330006, Jiangxi Province, China.
| |
Collapse
|
15
|
BC032913 as a Novel Antisense Non-coding RNA is Downregulated in Gastric Cancer. J Gastrointest Cancer 2020; 52:928-931. [DOI: 10.1007/s12029-020-00517-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Xiao Y, Xiao T, Ou W, Wu Z, Wu J, Tang J, Tian B, Zhou Y, Su M, Wang W. LncRNA SNHG16 as a potential biomarker and therapeutic target in human cancers. Biomark Res 2020; 8:41. [PMID: 32944244 PMCID: PMC7487997 DOI: 10.1186/s40364-020-00221-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/26/2020] [Indexed: 01/27/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) represent an important class of RNAs comprising more than 200 nucleotides, which are produced by RNA polymerase II. Although lacking an open reading framework and protein-encoding activity, lncRNAs can mediate endogenous gene expression by serving as chromatin remodeler, transcriptional or post-transcriptional modulator, and splicing regulator during gene modification. In recent years, increasing evidence shows the significance of lncRNAs in many malignancies, with vital roles in tumorigenesis and cancer progression. Moreover, lncRNAs were also considered potential diagnostic and prognostic markers in cancer. The lncRNA small nuclear RNA host gene 16 (SNHG16), found on chromosome 17q25.1, represents a novel tumor-associated lncRNA. SNHG16 was recently found to exhibit dysregulated expression in a variety of malignancies. There are growing evidence of SNHG16's involvement in characteristics of cancer, including proliferation, apoptosis, together with its involvement in chemoresistance. In addition, SNHG16 has been described as a promising diagnostic and prognostic biomarker in cancer patients. The current review briefly summarizes recently reported findings about SNHG16 and discuss its expression, roles, mechanisms, and diagnostic and prognostic values in human cancers.
Collapse
Affiliation(s)
- Yuhang Xiao
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
- Department of Pharmacy, Xiangya Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410001 PR China
| | - Ta Xiao
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu 210042 China
| | - Wei Ou
- Department of Pharmacy, The First People’s Hospital of Yue Yang, Yue Yang, PR China
| | - Zhining Wu
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Jie Wu
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Jinming Tang
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Bo Tian
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Yong Zhou
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
| | - Min Su
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenxiang Wang
- Thoracic Surgery Department 2, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013 PR China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
17
|
Assessment of expression of vitamin D receptor-associated lncRNAs in gastric cancer. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
18
|
Aravindan N, Herman T, Aravindan S. Emerging therapeutic targets for neuroblastoma. Expert Opin Ther Targets 2020; 24:899-914. [PMID: 33021426 PMCID: PMC7554151 DOI: 10.1080/14728222.2020.1790528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Neuroblastoma (NB) is the prime cancer of infancy, and accounts for 9% of pediatric cancer deaths. While children diagnosed with clinically stable NB experience a complete cure, those with high-risk disease (HR-NB) do not recover, despite intensive therapeutic strategies. Development of novel and effective targeted therapies is needed to counter disease progression, and to benefit long-term survival of children with HR-NB. AREAS COVERED Recent studies (2017-2020) pertinent to NB evolution are selectively reviewed to recognize novel and effective therapeutic targets. The prospective and promising therapeutic targets/strategies for HR-NB are categorized into (a) targeting oncogene-like and/or reinforcing tumor suppressor (TS)-like lncRNAs; (b) targeting oncogene-like microRNAs (miRs) and/or mimicking TS-miRs; (c) targets for immunotherapy; (d) targeting epithelial-to-mesenchymal transition and cancer stem cells; (e) novel and beneficial combination approaches; and (f) repurposing drugs and other strategies in development. EXPERT OPINION It is highly unlikely that agents targeting a single candidate or signaling will be beneficial for an HR-NB cure. We must develop efficient drug deliverables for functional targets, which could be integrated and advance clinical therapy. Fittingly, the looming evidence indicated an aggressive evolution of promising novel and integrative targets, development of efficient drugs, and improvised strategies for HR-NB treatment.
Collapse
Affiliation(s)
| | - Terence Herman
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
- Stephenson Cancer Center, Oklahoma City, USA
| | | |
Collapse
|
19
|
Jiao R, Jiang W, Wei X, Zhang M, Zhao S, Huang G. Clinicopathological significance and prognosis of long noncoding RNA SNHG16 expression in human cancers: a meta-analysis. BMC Cancer 2020; 20:662. [PMID: 32677912 PMCID: PMC7366298 DOI: 10.1186/s12885-020-07149-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 07/07/2020] [Indexed: 12/31/2022] Open
Abstract
Background Recent studies have highlighted the important role of long non-coding RNA SNHG16 in various human cancers. Here, we conducted a meta-analysis to investigate the effect of SNHG16 expression on clinicopathological features and prognosis in patients with different kinds of human cancers. Methods We performed a systematic search in electronic databases including PubMed, EMBASE, Cochrane Library and Web of Science, to investigate the potential association between SNHG16 expression and prognostic significance and clinical features in cancer patients. Odds ratios (ORs) or hazards ratios (HRs) with corresponding 95% confidence intervals (95% CIs) were pooled to estimate the prognosis value of SNHG16 by StataSE 15.0 software. Results A total of 16 eligible studies with 1299 patients were enrolled in our meta-analysis. The results revealed that increased expression level of SNHG16 was significantly associated with larger tumor size (OR: 3.357; 95% CI: 2.173–5.185; P < 0.001), advanced TNM stage (OR: 2.930; 95% CI: 1.522–5.640; P = 0.001) and poor histological grade (OR: 3.943; 95% CI: 1.955–7.952; P < 0.001), but not correlated with smoking status (P = 0.489), sex (P = 0.932), distant metastasis (P = 0.052), or lymph node metastasis (P = 0.155). Moreover, the pooled HR showed that elevated expression SNHG16 was associated with a significantly poorer overall survival (OS) (HR = 1.866, 95% CI: 1.571–2.216, P < 0.001). For the set of cancer types, high expression of SNHG16 was significantly associated with shorter OS in patients with cancers of the urinary system (HR: 2.523, 95% CI:1.540–4.133; P <0.001), digestive system (HR: 2.406, 95% CI:1.556–3.721; P <0.001), and other cancers (including glioma and non-small cell lung cancer) (HR: 1.786, 95% CI:1.406–2.267; P <0.001). Conclusions LncRNA SNHG16 overexpression might serve as an unfavorable prognostic factor, which provides a basis for medical workers to evaluate the prognosis of patients and to help the decision-making process.
Collapse
Affiliation(s)
- Ruonan Jiao
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Wei Jiang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Xin Wei
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Mengpei Zhang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Si Zhao
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Guangming Huang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| |
Collapse
|
20
|
SNHG16 promotes tumorigenesis and cisplatin resistance by regulating miR-338-3p/PLK4 pathway in neuroblastoma cells. Cancer Cell Int 2020; 20:236. [PMID: 32536824 PMCID: PMC7291484 DOI: 10.1186/s12935-020-01291-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023] Open
Abstract
Background Long noncoding RNA small nucleolar RNA host gene 16 (lncRNA SNHG16) has been revealed to be involved in the tumorigenesis of neuroblastoma. However, the role of SNHG16 in regulating cisplatin sensitivity in neuroblastoma remains largely unknown. Methods The expression of SNHG16, microRNA (miR)-338-3p and polo-like kinase 4 (PLK4) mRNA was measured using quantitative real-time polymerase chain reaction. The protein levels of PLK4, multidrug resistance protein 1 (MRP1), multidrug-resistance gene 1-type p-glycoprotein (P-gp) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway-related proteins were detected by Western blot. The half maximal inhibitory concentration (IC50) value, cell proliferation, migration and invasion were analyzed using Cell Counting Kit-8 assays or Transwell assay. Apoptotic cells were measured by Flow cytometry. The interaction between miR-338-3p and SNHG16 or PLK4 was confirmed by dual-luciferase reporter and RNA immunoprecipitation assay. In vivo experiments were conducted through the murine xenograft model. Results SNHG16 was up-regulated, while miR-338-3p was down-regulated in cisplatin-resistant neuroblastoma tissues and cells. SNHG16 silencing weakened cisplatin resistance, reflected by the reduction of IC50 value, down-regulation of MRP-1 and P-gp protein expression, suppression of proliferation, migration and invasion, as well as enhancement of apoptosis in SNHG16 deletion cisplatin-resistant neuroblastoma cells. Besides that, SNHG16 could regulate PLK4 expression by sponging miR-338-3p and SNHG16/miR-338-3p/PLK4 axis could affect the activation of PI3K/AKT pathway in cisplatin-resistant neuroblastoma cells. MiR-338-3p inhibition attenuated SNHG16 deletion-mediated impairment on cisplatin resistance and PLK4 overexpression reversed the decrease of cisplatin-resistance induced by miR-338-3p re-expression. Furthermore, SNHG16 knockdown contributed to the anti-tumor effect of cisplatin in neuroblastoma in vivo. Conclusion SNHG16 contributed to the tumorigenesis and cisplatin resistance in neuroblastoma possibly through miR-338-3p/PLK4 pathway, indicating a novel insight for overcoming chemoresistance in neuroblastoma patients.
Collapse
|
21
|
Deng D, Yang S, Wang X. Long non-coding RNA SNHG16 regulates cell behaviors through miR-542-3p/HNF4α axis via RAS/RAF/MEK/ERK signaling pathway in pediatric neuroblastoma cells. Biosci Rep 2020; 40:BSR20200723. [PMID: 32412051 PMCID: PMC7251324 DOI: 10.1042/bsr20200723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 01/27/2023] Open
Abstract
Neuroblastoma (NB) is an extracranial solid tumor in children with complex mechanism. Increasing reports indicated that long non-coding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) account for the pathogenesis of NB. Nevertheless, the precise functions of SNHG16 needed to be further exposed in NB progression. Our data revealed that SNHG16 and hepatocyte nuclear factor 4 α (HNF4α) were up-regulated, but miR-542-3p was down-regulated in NB. Knockdown of SNHG16 or HNF4α could impede cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) in vitro. Interestingly, the role of SNHG16 detetion in cell behaviors was rescued by HNF4α overexpression in NB cells. Mechanically, SNHG16 modulated the progression of tumor growth via miR-542-3p/HNF4α axis in NB. Also, SNHG16 knockdown inactivated rat sarcoma/effector of RAS/mitogen-activated extracellular signal-regulated kinase/extracellular regulated protein kinases (RAS/RAF/MEK/ERK) signaling pathway through HNF4α. Therefore, SNHG16/miR-542-3p/HNF4α axis modified NB progression via RAS/RAF/MEK/ERK signaling pathway, might highlight a novel therapeutic approach for NB.
Collapse
Affiliation(s)
- Defeng Deng
- Department of Hematology, Kaifeng Children’s Hospital, Kaifeng, Henan, China
| | - Shuangjie Yang
- Department of Hematology, Kaifeng Children’s Hospital, Kaifeng, Henan, China
| | - Xiang Wang
- Department of Hematology, Kaifeng Children’s Hospital, Kaifeng, Henan, China
| |
Collapse
|
22
|
Wang X, Liu L, Zhao W, Li Q, Wang G, Li H. LncRNA SNHG16 Promotes the Progression of Laryngeal Squamous Cell Carcinoma by Mediating miR-877-5p/FOXP4 Axis. Onco Targets Ther 2020; 13:4569-4579. [PMID: 32547087 PMCID: PMC7251222 DOI: 10.2147/ott.s250752] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/26/2020] [Indexed: 12/24/2022] Open
Abstract
Objective Laryngeal cancer is a common malignant tumor in the ENT, of which laryngeal squamous cell carcinoma (LSCC) accounts for more than 90% of laryngeal cancer. The purpose of this study is to investigate the regulatory mechanism of lncRNA SNHG16 in LSCC. Materials and Methods Real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to measure mRNA expression. Cell Counting Kit (CCK-8), Transwell and luciferase reporter assays, flow cytometric analysis and Western blot analysis were used to investigate the function of lncRNA SNHG16 in LSCC. Results SNHG16 expression was increased in LSCC tissues and cells. The abnormal expression of SNHG16 was associated with clinical stage and lymph node metastasis in LSCC patients. In addition, knockdown of SNHG16 restrained cell proliferation, migration and invasion in LSCC. More importantly, SNHG16 acted as a competitive endogenous RNA in LSCC and regulated FOXP4 expression by making miR-877-5p sponge. Further, SNHG16 promoted LSCC progression by interacting with miR-877-5p and FOXP4. Conclusion LncRNA SNHG16 promotes the progression of LSCC by sponging miR-877-5p and upregulating FOXP4.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Clinical Laboratory, Jinan City People's Hospital, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan 271199, People's Republic of China
| | - Liming Liu
- Department of Otorhinolaryngology, Juye County Hospital of TCM, Heze 274900, People's Republic of China
| | - Wenfei Zhao
- Department of Comprehensive Oncology Therapy, Qingdao Central Hospital, Qingdao University, Qingdao 266043, People's Republic of China
| | - Qingyan Li
- Department of Clinical Laboratory, The People's Hospital of Zhangqiu Area, Jinan 250200, People's Republic of China
| | - Guangsheng Wang
- Department of Neurology, The People's Hospital of Zhangqiu Area, Jinan 250200, People's Republic of China
| | - Huahui Li
- Department of Clinical Laboratory, Qingdao Municipal Hospital, Qingdao 266071, People's Republic of China
| |
Collapse
|
23
|
Ba MC, Ba Z, Long H, Cui SZ, Gong YF, Yan ZF, Lin KP, Wu YB, Tu YN. LncRNA AC093818.1 accelerates gastric cancer metastasis by epigenetically promoting PDK1 expression. Cell Death Dis 2020; 11:64. [PMID: 31988283 PMCID: PMC6985138 DOI: 10.1038/s41419-020-2245-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/18/2022]
Abstract
Gastric cancer (GC) is a highly prevalent type of metastatic tumor. The mechanisms underlying GC metastasis are poorly understood. Some long noncoding RNAs (lncRNAs) reportedly play key roles in regulating metastasis of GC. However, the biological roles of five natural antisense lncRNAs (AC093818.1, CTD-2541M15.1, BC047644, RP11-597M12.1, and RP11-40A13.1) in GC metastasis remain unclear. In this study, the expression of these lncRNAs was measured by quantitative reverse transcription-polymerase chain reaction. Migration and invasion were evaluated by wound-healing and the Transwell assay, respectively. Stable cells were injected into the tail veins of nude mice. Sections of collected lung and liver tissues were stained using hematoxylin and eosin. Protein expression was analyzed by western blot. RNA immunoprecipitation (RIP) assay was used to verify whether the STAT3 and SP1 transcription factors bound to AC093818.1 in GC cells. Expression levels of the five lncRNAs, especially AC093818.1, were significantly upregulated in metastatic GC tissues relative to those in nonmetastatic GC tissues. AC093818.1 expression was correlated with invasion, lymphatic metastasis, distal metastasis, and tumor-node-metastasis stage. AC093818.1 expression was highly sensitive and specific in the diagnosis of metastatic or nonmetastatic GC. AC093818.1 overexpression promoted GC migration and invasion in vitro and in vivo. AC093818.1 overexpression increased PDK1, p-AKT1, and p-mTOR expression levels. AC093818.1 silencing decreased these expressions. AC093818.1 bound to transcription factors STAT3 and SP1, and SP1 or STAT3 silencing could alleviated the effect of AC093818.1 overexpression. The data demonstrate that lncRNA AC093818.1 accelerates gastric cancer metastasis by epigenetically promoting PDK1 expression. LncRNA AC093818.1 may be a potential therapeutic target for metastatic GC.
Collapse
Affiliation(s)
- Ming-Chen Ba
- Intracelom Hyperthermic Perfusion Therapy Center, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P.R. China.
| | - Zheng Ba
- Intensive Care Unit, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Hui Long
- Department of Pharmacy, Guangzhou Dermatology Institute, Guangzhou, 510095, P.R. China
| | - Shu-Zhong Cui
- Intracelom Hyperthermic Perfusion Therapy Center, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P.R. China
| | - Yuan-Feng Gong
- Intracelom Hyperthermic Perfusion Therapy Center, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P.R. China
| | - Zhao-Fei Yan
- Intracelom Hyperthermic Perfusion Therapy Center, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P.R. China
| | - Kun-Peng Lin
- Intracelom Hyperthermic Perfusion Therapy Center, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P.R. China
| | - Yin-Bing Wu
- Intracelom Hyperthermic Perfusion Therapy Center, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P.R. China
| | - Yi-Nuo Tu
- Intracelom Hyperthermic Perfusion Therapy Center, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P.R. China
| |
Collapse
|
24
|
Xu P, Xiao H, Yang Q, Hu R, Jiang L, Bi R, Jiang X, Wang L, Mei J, Ding F, Huang J. The USP21/YY1/SNHG16 axis contributes to tumor proliferation, migration, and invasion of non-small-cell lung cancer. Exp Mol Med 2020; 52:41-55. [PMID: 31956270 PMCID: PMC7000404 DOI: 10.1038/s12276-019-0356-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/18/2019] [Accepted: 09/25/2019] [Indexed: 12/30/2022] Open
Abstract
Deubiquitinases (DUBs) and noncoding RNAs have been the subjects of recent extensive studies regarding their roles in lung cancer, but the mechanisms involved are largely unknown. In our study, we used The Cancer Genome Atlas data set and bioinformatics analyses and identified USP21, a DUB, as a potential contributor to oncogenesis in non-small-cell lung cancer (NSCLC). We further demonstrated that USP21 was highly expressed in NSCLCs. We then conducted a series of in vitro and in vivo assays to explore the effect of USP21 on NSCLC progression and the underlying mechanism involved. USP21 promoted NSCLC cell proliferation, migration, and invasion and in vivo tumor growth by stabilizing a well-known oncogene, Yin Yang-1 (YY1), via mediating its deubiquitination. Furthermore, YY1 transcriptionally regulates the expression of SNHG16. Moreover, StarBase bioinformatics analyses predicted that miR-4500 targets SNHG16 and USP21. A series of in vitro experiments indicated that SNHG16 increased the expression of USP21 through miR-4500. In summary, the USP21/YY1/SNHG16 axis plays a role in promoting the progression of NSCLC. Therefore, the USP21/YY1/SNHG16/miR-4500 axis may be a potential therapeutic target in NSCLC treatment. Therapies targeting a molecular feedback loop involved in tumor growth may prove valuable for treating non-small-cell lung cancer. Fangbao Ding, Jianbing Huang, and co-workers at Shanghai Jiao Tong University in Shanghai, China, have shown how an enzyme called USP21 promotes cancer cell proliferation and tumor growth in non-small-cell lung cancer. The team took cancerous and non-cancerous lung tissue samples from 42 patients, and analyzed the expression and behavior of USP21. The enzyme was highly expressed in cancerous tissues, where it stabilized a known gene with the potential to cause cancer called YY1. This gene also regulated the expression of a particular RNA molecule, which in turn worked to increase levels of USP21. This cyclical process encouraged the proliferation, migration and invasion of non-small-cell lung cancer cells, and may provide a future therapeutic target.
Collapse
Affiliation(s)
- Pei Xu
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Haibo Xiao
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Qi Yang
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Rui Hu
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Lianyong Jiang
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Rui Bi
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Xueyan Jiang
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Lei Wang
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Ju Mei
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Fangbao Ding
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China.
| | - Jianbing Huang
- Department of Cardiothoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China.
| |
Collapse
|
25
|
Wen Y, Gong X, Dong Y, Tang C. Long Non Coding RNA SNHG16 Facilitates Proliferation, Migration, Invasion and Autophagy of Neuroblastoma Cells via Sponging miR-542-3p and Upregulating ATG5 Expression. Onco Targets Ther 2020; 13:263-275. [PMID: 32021273 PMCID: PMC6959506 DOI: 10.2147/ott.s226915] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Neuroblastoma (NB) is a heterogeneous pediatric malignant tumor with many biological and clinical characteristics. Long non-coding RNA small nucleolar RNA host gene 16 (SNHG16) plays vital role in the development of NB. However, the potential mechanism of SNHG16 in the progression of NB is rarely reported. METHODS The expression levels of SNHG16, miR-542-3p and autophagy-related gene 5 (ATG5) were measured with quantitative real-time polymerase chain reaction (qRT-PCR). The proliferation, migration and invasion of NB cells were determined using 3-(4, 5-dimethylthiazol-2-YI)-2, 5-diphenyltetrazolium bromide (MTT) or transwell assay. Protein levels of ATG5, microtubule-associated protein A1/1B-light chain3 (LC3-I/II) and p62 were detected by Western blot analysis. The interaction between miR-542-3p and SNHG16 or ATG5 was predicted by starBase and confirmed by dual luciferase reporter assay. Xenograft mice models were constructed to confirm the role of SNHG16 in vivo. RESULTS SNHG16 was upregulated in NB tissues and cells and associated with clinical stage and poor prognosis of NB. Knockdown of SNHG16 impeded proliferation, migration, invasion and autophagy of NB cells in vitro, and suppressed tumor growth in vivo. Interestingly, SNHG16 mediated ATG5 expression through sponging miR-542-3p in NB cells. Moreover, miR-542-3p downregulation reversed the inhibitory effects of SNHG16 silencing on proliferation, migration, invasion and autophagy of NB cells. Besides, ATG5 overturned the regulatory effects on proliferation, migration, invasion and autophagy of NB cells induced by SNHG16 or miR-542-3p knockdown. CONCLUSION SNHG16 facilitated proliferation, migration, invasion and autophagy of NB cells via sponging miR-542-3p and upregulating ATG5 expression in NB.
Collapse
Affiliation(s)
- Yi Wen
- Neonatal Pediatrics, Central Hospital of Zhoukou City, Zhoukou, Henan, People’s Republic of China
| | - Xiaohui Gong
- Neonatal Pediatrics, Shanghai Children’s Hospital, Shanghai, People’s Republic of China
| | - Yubin Dong
- Neonatal Pediatrics, Central Hospital of Zhoukou City, Zhoukou, Henan, People’s Republic of China
| | - Chenghe Tang
- Neonatal Pediatrics, First Affiliated Hospital of Xinxiang Medical College, Xinxiang, Henan, People’s Republic of China
| |
Collapse
|
26
|
Yang M, Wei W. SNHG16: A Novel Long-Non Coding RNA in Human Cancers. Onco Targets Ther 2019; 12:11679-11690. [PMID: 32021246 PMCID: PMC6942535 DOI: 10.2147/ott.s231630] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/13/2019] [Indexed: 01/27/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have recently been considered as central regulators in diverse biological processes controlling tumorigenesis. Small nucleolar RNA host gene 16 (SNHG16) is an important tumor-associated lncRNA mainly involved in tumorigenesis and progression by competing with endogenous RNA (ceRNA) which sponges tumor-suppressive microRNA (miRNA), and by its recruitment mechanism. SNHG16 is overexpressed in tumor tissues and cell lines of different kinds of cancers, and its presence is associated with a poor clinical prognosis. Reviewing all publications about SNHG16 revealed that it plays a key role in the different hallmarks that define human cancer, including promoting proliferation, activating migration and invasion, inhibiting apoptosis, affecting lipid metabolism and chemoresistance. This review highlights the role that the aberrant expression of SNHG16 plays in the development and progression of cancer, and suggests that SNHG16 may function as a potential biomarker and therapeutic target for human cancers.
Collapse
Affiliation(s)
- Ming Yang
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing100730, People’s Republic of China
| | - Wenbin Wei
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology and Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing100730, People’s Republic of China
| |
Collapse
|
27
|
Ghafouri-Fard S, Taheri M. Long non-coding RNA signature in gastric cancer. Exp Mol Pathol 2019; 113:104365. [PMID: 31899194 DOI: 10.1016/j.yexmp.2019.104365] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/18/2019] [Accepted: 12/28/2019] [Indexed: 02/07/2023]
Abstract
Gastric cancer as a common human malignancy has been associated with aberrant expressions of several coding and non-coding genes. Long non-coding RNAs (lncRNAs) as regulators of gene expressions at different genomic, transcriptomic and post-transcriptomic levels are among putative biomarkers and therapeutic targets in gastric cancer. In the present study, we have searched available literature and listed lncRNAs that are involved in the pathogenesis of gastric cancer. In addition, we discuss associations between expressions of these lncRNAs and tumoral features or risk factors for gastric cancer. Based on the established role of lncRNAs in regulation of genomic stability, cell cycle, apoptosis, angiogenesis and other aspects of cell physiology, the potential of these transcripts as therapeutic targets in gastric cancer should be evaluated in future studies.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Role of SNHG16 in human cancer. Clin Chim Acta 2019; 503:175-180. [PMID: 31901482 DOI: 10.1016/j.cca.2019.12.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 01/27/2023]
Abstract
A growing body of evidence suggests that long non-coding RNAs (lncRNAs), a novel class of non-coding endogenous single-stranded RNA, play a key role in multiple physiological and pathological processes through transcriptional interference, post-transcriptional regulation, and epigenetic modification. Furthermore, many studies have shown that lncRNAs-as oncogenes or tumour suppressors-play an important role in the occurrence and development of human cancers. Small nucleolar RNA host gene 16 (SNHG16) was initially identified as an oncogenic lncRNA in neuroblastoma, and has since been identified as a carcinogenic regulator of various malignant tumours. Overexpression of SNHG16 is associated with clinical and pathological characteristics of cancer patients, and regulates cell proliferation, apoptosis, invasion and metastasis through a variety of potential mechanisms. Therefore, SNHG16 may be a promising biomarker and therapeutic target for cancers. In this review, we summarize the biological function, related mechanisms and potential clinical significance of SNHG16 in multiple human cancers.
Collapse
|
29
|
Sun G, Su G, Liu F, Han W. NRAS Contributes to Retinoblastoma Progression Through SNHG16/miR-183-5p/NRAS Regulatory Network. Onco Targets Ther 2019; 12:10703-10715. [PMID: 31827328 PMCID: PMC6902855 DOI: 10.2147/ott.s232470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose The oncogene of wild type neuroblastoma RAS viral oncogene homolog (NRAS) has been found to involve in the tumorigenesis of cancers. However, the role of NRAS in retinoblastoma (RB) progression remains largely unknown. Methods The expression levels of NRAS, miR-183-5p and small nucleolar RNA host gene 16 (SNHG16) were measured using quantitative real-time polymerase chain reaction assay or Western blot assay, respectively. Cell proliferation and apoptosis were analyzed with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay or flow cytometry, respectively. Transwell assay was used to determine cell migration and invasion abilities. The interaction between miR-183-5p and NRAS or SNHG16 was analyzed using bioinformatics analysis and dual-luciferase reporter assay. Results NRAS was elevated in RB tissues and cell lines, knockdown of NRAS could inhibit proliferation, migration and invasion but induced apoptosis in vitro and suppressed tumor growth in vivo. NRAS was confirmed to be a target of miR-183-5p and was negatively regulated by miR-183-5p in RB cells. Moreover, overexpressed NRAS reversed miR-183-5p mediated inhibition on RB cell progression. Besides that, SNHG16 directly interacted with miR-183-5p and reduced miR-183-5p expression in RB cells. The suppression of RB cell progression induced by SNHG16 silencing could be partially attenuated by the inhibition of miR-183-5p. Besides that, SNHG16 could regulate NRAS expression through competitively binding to miR-183-5p in RB cells. Conclusion NRAS functioned as an oncogene to contribute to RB progression by SNHG16/miR-183-5p/NRAS regulatory network, indicating a novel and promising therapeutic target for RB.
Collapse
Affiliation(s)
- Guangli Sun
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China
| | - Gang Su
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China
| | - Fang Liu
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China
| | - Wenjie Han
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan, People's Republic of China
| |
Collapse
|
30
|
Yu Y, Dong JT, He B, Zou YF, Li XS, Xi CH, Yu Y. LncRNA SNHG16 induces the SREBP2 to promote lipogenesis and enhance the progression of pancreatic cancer. Future Oncol 2019; 15:3831-3844. [PMID: 31664866 DOI: 10.2217/fon-2019-0321] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Blocking lipogenesis could significantly inhibit the progression of pancreatic cancer. Exploring the regulatory mechanisms of lipogenesis by lncRNA SNHG16 might be of great significance to control the development of pancreatic cancer. Methods: The proliferation, migration, invasion and lipogenesis were determined with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, wound healing, transwell and Oil Red O staining assays, respectively. The interactions among lncRNA SNHG16, miR-195 and SREBP2 were analyzed by dual luciferase reporter assays. Results: Both the knock down of lncRNA SNHG16 and SREBP2 and overexpression of miR-195 suppressed the proliferation, migration, invasion and lipogenesis in pancreatic cancer cells. LncRNA SNHG16 directly sponged miR-195 to modulate the lipogenesis via regulating the expression of SREBP2. Conclusion: LncRNA SNHG16 accelerated the development of pancreatic cancer and promoted lipogenesis via directly regulating miR-195/SREBP2 axis.
Collapse
Affiliation(s)
- Yi Yu
- Department of Pediatrics, Ruijin Hospital North, Shanghai Jiaotong University, School of Medicine, Shanghai 201801, PR China
| | - Jia-Tian Dong
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, PR China
| | - Bing He
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, PR China
| | - Yu-Feng Zou
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, PR China
| | - Xue-Song Li
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, PR China
| | - Chen-Hui Xi
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, PR China
| | - Yuan Yu
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, PR China
| |
Collapse
|
31
|
Li W, Wang S, Xu J, Mao G, Tian G, Yang J. Inferring Latent Disease-lncRNA Associations by Faster Matrix Completion on a Heterogeneous Network. Front Genet 2019; 10:769. [PMID: 31572428 PMCID: PMC6749816 DOI: 10.3389/fgene.2019.00769] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/19/2019] [Indexed: 11/26/2022] Open
Abstract
Current studies have shown that long non-coding RNAs (lncRNAs) play a crucial role in a variety of fundamental biological processes related to complex human diseases. The prediction of latent disease-lncRNA associations can help to understand the pathogenesis of complex human diseases at the level of lncRNA, which also contributes to the detection of disease biomarkers, and the diagnosis, treatment, prognosis and prevention of disease. Nevertheless, it is still a challenging and urgent task to accurately identify latent disease-lncRNA association. Discovering latent links on the basis of biological experiments is time-consuming and wasteful, necessitating the development of computational prediction models. In this study, a computational prediction model has been remodeled as a matrix completion framework of the recommendation system by completing the unknown items in the rating matrix. A novel method named faster randomized matrix completion for latent disease-lncRNA association prediction (FRMCLDA) has been proposed by virtue of improved randomized partial SVD (rSVD-BKI) on a heterogeneous bilayer network. First, the correlated data source and experimentally validated information of diseases and lncRNAs are integrated to construct a heterogeneous bilayer network. Next, the integrated heterogeneous bilayer network can be formalized as a comprehensive adjacency matrix which includes lncRNA similarity matrix, disease similarity matrix, and disease-lncRNA association matrix where the uncertain disease-lncRNA associations are referred to as blank items. Then, a matrix approximate to the original adjacency matrix has been designed with predicted scores to retrieve the blank items. The construction of the approximate matrix could be equivalently resolved by the nuclear norm minimization. Finally, a faster singular value thresholding algorithm with a randomized partial SVD combing a new sub-space reuse technique has been utilized to complete the adjacency matrix. The results of leave-one-out cross-validation (LOOCV) experiments and 5-fold cross-validation (5-fold CV) experiments on three different benchmark databases have confirmed the availability and adaptability of FRMCLDA in inferring latent relationships of disease-lncRNA pairs, and in inferring lncRNAs correlated with novel diseases without any prior interaction information. Additionally, case studies have shown that FRMCLDA is able to effectively predict latent lncRNAs correlated with three widespread malignancies: prostate cancer, colon cancer, and gastric cancer.
Collapse
Affiliation(s)
- Wen Li
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Shulin Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Junlin Xu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Guo Mao
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Geng Tian
- Geneis Beijing Co., Ltd., Beijing, China
| | | |
Collapse
|
32
|
Pang W, Zhai M, Wang Y, Li Z. Long noncoding RNA SNHG16 silencing inhibits the aggressiveness of gastric cancer via upregulation of microRNA-628-3p and consequent decrease of NRP1. Cancer Manag Res 2019; 11:7263-7277. [PMID: 31447585 PMCID: PMC6682761 DOI: 10.2147/cmar.s211856] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/05/2019] [Indexed: 12/16/2022] Open
Abstract
Background MicroRNA-628-3p (miR-628) has been reported to play important roles in the progression of multiple human cancer types. Nonetheless, whether the expression profile of miR-628 is altered in gastric cancer remains unclear and whether its aberrant expression plays a crucial part in the aggressiveness of gastric cancer is yet to be determined. Therefore, in this study, we systematically investigated the involvement of miR-628 in gastric cancer progression. Materials and methods MiR-628 expression in gastric cancer tissues and cell lines were determined via reverse transcription-quantitative polymerase chain reaction (RT-qPCR). A CCK-8 assay, flow-cytometric analysis, Transwell assays, and a xenograft model experiment were performed to evaluate the influence of miR-628 overexpression on gastric cancer cells. Notably, the mechanisms underlying the tumor-suppressive activity of miR-628 in gastric cancer cells were explored by bioinformatics analysis, a luciferase reporter assay, RT-qPCR, and Western blotting. Results MiR-628 expression was low in gastric cancer tissue samples and cell lines. The low expression of miR-628 was closely associated with the lymph node metastasis, invasive depth and TNM stage among patients with gastric cancer. Further clinical analysis indicated that patients with gastric cancer underexpressing miR-628 had a worse prognosis than did the patients with high miR-628 expression in the tumor. Overexpressed miR-628 restrained proliferation, migration, and invasion; induced apoptosis; and impaired tumor growth of gastric cancer cells. In addition, neuropilin 1 (NRP1) mRNA was validated as the direct target of miR-628 in gastric cancer. Long noncoding RNA small nucleolar RNA host gene 16 (SNHG16) was demonstrated to sponge miR-628 in gastric cancer. Moreover, miR-628 knockdown abrogated the influence of SNHG16 silencing on gastric cancer cells. Conclusion Our findings elucidate how the SNHG16–miR-628–NRP1 pathway serves as a regulatory network playing crucial roles in gastric cancer progression, suggesting that this pathway may be a novel target of anticancer therapy.
Collapse
Affiliation(s)
- Weifeng Pang
- Department of Internal Oncology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Mingcui Zhai
- Department of Burn, Heilongjiang Province Hospital, Harbin, People's Republic of China
| | - Yue Wang
- Department of Pharmacology and Toxicology, Wright State University, Fairborn, OH, USA
| | - Zhiqiang Li
- Department of General Surgery, Suihua First Hospital in Heilongjiang Province, Suihua, People's Republic of China
| |
Collapse
|
33
|
Yu J, Xuan Z, Feng X, Zou Q, Wang L. A novel collaborative filtering model for LncRNA-disease association prediction based on the Naïve Bayesian classifier. BMC Bioinformatics 2019; 20:396. [PMID: 31315558 PMCID: PMC6637631 DOI: 10.1186/s12859-019-2985-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/03/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Since the number of known lncRNA-disease associations verified by biological experiments is quite limited, it has been a challenging task to uncover human disease-related lncRNAs in recent years. Moreover, considering the fact that biological experiments are very expensive and time-consuming, it is important to develop efficient computational models to discover potential lncRNA-disease associations. RESULTS In this manuscript, a novel Collaborative Filtering model called CFNBC for inferring potential lncRNA-disease associations is proposed based on Naïve Bayesian Classifier. In CFNBC, an original lncRNA-miRNA-disease tripartite network is constructed first by integrating known miRNA-lncRNA associations, miRNA-disease associations and lncRNA-disease associations, and then, an updated lncRNA-miRNA-disease tripartite network is further constructed through applying the item-based collaborative filtering algorithm on the original tripartite network. Finally, based on the updated tripartite network, a novel approach based on the Naïve Bayesian Classifier is proposed to predict potential associations between lncRNAs and diseases. The novelty of CFNBC lies in the construction of the updated lncRNA-miRNA-disease tripartite network and the introduction of the item-based collaborative filtering algorithm and Naïve Bayesian Classifier, which guarantee that CFNBC can be applied to predict potential lncRNA-disease associations efficiently without entirely relying on known miRNA-disease associations. Simulation results show that CFNBC can achieve a reliable AUC of 0.8576 in the Leave-One-Out Cross Validation (LOOCV), which is considerably better than previous state-of-the-art results. Moreover, case studies of glioma, colorectal cancer and gastric cancer demonstrate the excellent prediction performance of CFNBC as well. CONCLUSIONS According to simulation results, due to the satisfactory prediction performance, CFNBC may be an excellent addition to biomedical researches in the future.
Collapse
Affiliation(s)
- Jingwen Yu
- grid.448798.eCollege of Computer Engineering & Applied Mathematics, Changsha University, Changsha, Hunan People’s Republic of China
- 0000 0000 8633 7608grid.412982.4Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, XiangTan, People’s Republic of China
| | - Zhanwei Xuan
- grid.448798.eCollege of Computer Engineering & Applied Mathematics, Changsha University, Changsha, Hunan People’s Republic of China
- 0000 0000 8633 7608grid.412982.4Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, XiangTan, People’s Republic of China
| | - Xiang Feng
- grid.448798.eCollege of Computer Engineering & Applied Mathematics, Changsha University, Changsha, Hunan People’s Republic of China
- 0000 0000 8633 7608grid.412982.4Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, XiangTan, People’s Republic of China
| | - Quan Zou
- 0000 0004 0369 4060grid.54549.39Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
- 0000 0004 1761 2484grid.33763.32School of Computer Science and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Lei Wang
- grid.448798.eCollege of Computer Engineering & Applied Mathematics, Changsha University, Changsha, Hunan People’s Republic of China
- 0000 0000 8633 7608grid.412982.4Key Laboratory of Intelligent Computing & Information Processing, Xiangtan University, XiangTan, People’s Republic of China
| |
Collapse
|
34
|
Xie X, Xu X, Sun C, Yu Z. Long intergenic noncoding RNA SNHG16 interacts with miR-195 to promote proliferation, invasion and tumorigenesis in hepatocellular carcinoma. Exp Cell Res 2019; 383:111501. [PMID: 31306653 DOI: 10.1016/j.yexcr.2019.111501] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/26/2019] [Accepted: 07/11/2019] [Indexed: 02/07/2023]
Abstract
Long non-coding RNAs (lncRNAs) have been confirmed crucial regulators in tumorgenesis. Small nucleolar RNA host gene 16 (SNHG16) has been recently shown to be dysregulated, which uncovered to be a potential oncogene in some cancers. However, the biological function and potential mechanism of SNHG16 in hepatocellular carcinoma (HCC) remain unclear. In our study, our observations showed that the expression level of SNHG16 in HCC tissues and cell lines was upregulated compared with adjacent noncancerous tissues and normal cells. In vitro, loss-of-function experiments revealed that SNHG16 knockdown suppressed the proliferation and weakened invasion of SMMC7721 and HepG2 cells. miR-195 expression was significantly decreased in HCC tissues and negatively correlated with SNHG16 expression. Furthermore, RIP and dual luciferase reporter assays showed that SNHG16 acted as an endogenous sponge by directly binding to miR-195 and downregulated its expression. SNHG16 overexpression inverted the inhibitory effect of miR-195 on proliferation and invasion of SMMC7721 and HepG2 cells. Additionally, SNHG16 depletion resulted in lower tumor growth and weight loss, in vivo. In conclusion, our findings reported that the oncogenic role of SNHG16 in HCC tumorigenesis through a novel SNHG16-miR-195 axis, which provided a novel insight for HCC and helped to probe a potential therapeutic target for the deadly disease.
Collapse
Affiliation(s)
- Xuhua Xie
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Xiaopei Xu
- Department of Physical Examination, The Third People's Hospital of Henan Province, Zhengzhou, 450006, PR China
| | - Changyu Sun
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zujiang Yu
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
35
|
Lin Q, Zheng H, Xu J, Zhang F, Pan H. LncRNA SNHG16 aggravates tumorigenesis and development of hepatocellular carcinoma by sponging miR-4500 and targeting STAT3. J Cell Biochem 2019; 120:11604-11615. [PMID: 30779219 DOI: 10.1002/jcb.28440] [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] [Received: 10/22/2018] [Revised: 11/29/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver tumor and becomes a lethal malignancy on account of high mortality and increasing incidence. A growing body of studies has proved that long noncoding RNAs (lncRNAs) participate in the development of diverse cancers. Although it has been commonly accepted that SNHG16 is a procancer gene in numerous cancers, the regulatory mechanism of SNHG16 in HCC still needs more explorations. In this study, our results delineated that SNHG16 presented much higher expression levels in HCC tissues and cells, particularly in advanced stages of HCC. Enhanced SNHG16 expression was strongly related to poor prognosis. SNHG16 facilitated HCC progression by promoting cell proliferation, migration, invasion, and epithelial-mesenchymal transition process as well as inhibiting cell apoptosis. SNHG16 served as a sponge for miR-4500 in HCC and miR-4500 neutralized the influences of SNHG16 knockdown on HCC. SNHG16 was confirmed to compete with signal transducer and activator of transcription 3 (STAT3) to bind with miR-4500. SNHG16 aggravated the development of HCC via sponging miR-4500 so as to upregulate STAT3. In other words, this study was the first to investigate the potential mechanism of SNHG16 in HCC and verified SNHG16 exerted its carcinogenesis by miR-4500/STAT3 axis, suggesting SNHG16 may be a new underlying therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Qing Lin
- Clinical Lab, Infectious Disease Hospital of Yantai, Yantai, Shandong, China
| | - Hongying Zheng
- Clinical Lab, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jianjun Xu
- Clinical Lab, Qingdao Chengyang People's Hospital, Qingdao, Shandong, China
| | - Feng Zhang
- Clinical Lab, Rizhao City Hospital of Traditional Chinese Medicine, Rizhao, Shandong, China
| | - Huazheng Pan
- Clinical Lab, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
36
|
Zhong JH, Xiang X, Wang YY, Liu X, Qi LN, Luo CP, Wei WE, You XM, Ma L, Xiang BD, Li LQ. The lncRNA SNHG16 affects prognosis in hepatocellular carcinoma by regulating p62 expression. J Cell Physiol 2019; 235:1090-1102. [PMID: 31256427 DOI: 10.1002/jcp.29023] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023]
Abstract
Long noncoding RNAs (lncRNAs) regulate tumor development and progression by promoting proliferation, invasion, and metastasis. The oncogenic role of lncRNA SNHG16 in hepatocellular carcinoma (HCC) has not been revealed. LncRNA SNHG16 is upregulated in HCC and correlates with poorer prognosis. Patients with high SNHG16 expression showed lower rates of overall and disease-free survival than patients with low SNHG16 expression. Multivariate Cox regression revealed that SNHG16 expression was an independent predictor of poor overall and disease-free survival. In vitro, SNHG16 promoted HCC cell proliferation, migration, and invasion while inhibiting apoptosis; in vivo, it accelerated tumor development. Altering SNHG16 expression altered levels of miR-17-5p, which in turn modified expression of p62, which has been shown to regulate the mTOR and NF-κB pathways. Indeed, altering SNHG16 expression in HCC cells activated mTOR and NF-κB signaling. These results reveal a potential mechanism for the oncogenic role of SNHG16 in HCC. SNHG16 may therefore be a promising diagnostic marker as well as therapeutic target in HCC.
Collapse
Affiliation(s)
- Jian-Hong Zhong
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China.,Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
| | - Xiao Xiang
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China.,Department of Hepatobilliary Surgery, Beijing Key Surgical Basic Research, Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, China
| | - Yan-Yan Wang
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Xu Liu
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Lu-Nan Qi
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China.,Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
| | - Cheng-Piao Luo
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Wen-E Wei
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Xue-Mei You
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China.,Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
| | - Liang Ma
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China.,Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
| | - Bang-De Xiang
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China.,Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
| | - Le-Qun Li
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China.,Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, China
| |
Collapse
|
37
|
Xu C, Hu C, Wang Y, Liu S. Long noncoding RNA SNHG16 promotes human retinoblastoma progression via sponging miR-140-5p. Biomed Pharmacother 2019; 117:109153. [PMID: 31234025 DOI: 10.1016/j.biopha.2019.109153] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Small nucleolar RNA host gene 16 (SNHG16), a long non-coding RNA, was reported to function as an oncogene in multiple cancers. However, its biological function and regulatory mechanism in retinoblastoma (RB) has not yet been revealed. In this study, we attempted to ascertain the biological role and underlying regulatory mechanism of SNHG16 in RB progression. The expression levels of SNHG16 were measured in RB tissues and cell lines. The effects of SNHG16 knockdown on the proliferation, colony formation, cell cycle progression and apoptosis were investigated using corresponding experiments. Bioinformatic analysis, luciferase reporter assay, and RNA immunoprecipitation assay were applied to identify potential microRNAs (miRs) that could bind with SNHG16. A nude model was established to investigate the effect of SNHG16 knockdown on tumor growth in vivo. We found that SNHG16 expression was upregulated in RB tissues and cell lines compared with normal controls. Knockdown of SNHG16 in RB cells significantly inhibited the proliferation and colony formation, and promoted apoptosis in vitro, as well as retarded tumor growth in vivo. Mechanistic investigation illustrated that SNHG16 acted as a sponge for miR-140-5p and regulated its expression in RB cells. Clinical evidence revealed a negative correlation between SNHG16 and miR-140-5p in RB specimens. Rescue experiments showed that inhibition of miR-140-5p partially attenuated the growth-suppressing effects of SNHG16-depletion on RB cells.. Collectively, SNHG16 exerts oncogenic role in RB by sponging miR-140-5p, suggesting that SNHG16 might be a potential therapy target for RB.
Collapse
Affiliation(s)
- Chunling Xu
- Department of Ophthalmology, the Second Hospital of Jilin University, 218#Ziqiang Street, Nanguan District, Changchun 130041, PR China
| | - Chunmei Hu
- Tumor and Hematology, the Second Hospital of Jilin University, 218#Ziqiang Street, Nanguan District, Changchun 130041, PR China
| | - Yingxue Wang
- Electrical Diagnosis, the Second Hospital of Jilin University, 218#Ziqiang Street, Nanguan District, Changchun 130041, PR China
| | - Shu Liu
- Department of Ophthalmology, the Second Hospital of Jilin University, 218#Ziqiang Street, Nanguan District, Changchun 130041, PR China.
| |
Collapse
|
38
|
Chen H, Li M, Huang P. LncRNA SNHG16 Promotes Hepatocellular Carcinoma Proliferation, Migration and Invasion by Regulating miR-186 Expression. J Cancer 2019; 10:3571-3581. [PMID: 31293662 PMCID: PMC6603422 DOI: 10.7150/jca.28428] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 04/30/2019] [Indexed: 12/11/2022] Open
Abstract
Long non-coding RNA (lncRNA) and microRNA (miRNA) play an important role in genesis and progression of tumors. The aim of this study was to explore the expression, biological function and molecular mechanism of small nucleolar RNA host gene 16 (SNHG16) in HCC. RT-qPCR was conducted to evaluate the expression level of SNHG16 in HCC tissues and cell lines. Our findings showed for the first time that SNHG16 was up-regulated in HCC tissues and cell lines. The expression of SNHG16 in cancer tissues was highly correlated with tumor size, TNM stage, ALT expression level and HBV DNA level. Moreover, cell proliferation, migration and invasion were detected by CCK-8 assay, transwell migration assay and transwell invasion assay, respectively. Xenograft tumor experiment was used to determine the biological function of SNHG16 in vivo. As revealed by our data, SNHG16 accelerated the proliferation, migration and invasion of HCC cell. SNHG16 facilitated tumor formation in vivo. Next, the relationship between SNHG16, miR-186 and ROCK1 were analyzed using bioinformatics analysis, qRT-PCR, luciferase reporter assay and western blot. Further molecular mechanism studies reported that the expression of SNHG16 was negatively correlated with the level of miR-186 and SNHG16 directly bound to miR-186. SNHG16 and miR-186 repressed each other. Notably, rescue experiments were conducted and showed that miR-186 reversed the effect of SNHG16 on cell. Taken together, SNHG16 promoted HCC cell proliferation, migration and invasion by functioning as a competitive endogenous RNA (ceRNA) to negatively regulate miR-186 expression. Our data suggested that SNHG16 might be a potential biomarker and a new therapeutic target for HCC.
Collapse
Affiliation(s)
- Hang Chen
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| | - Molin Li
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| | - Ping Huang
- National Key Clinical Department, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, P.R. China
| |
Collapse
|
39
|
Liu S, Zhang W, Liu K, Liu Y. LncRNA SNHG16 promotes tumor growth of pancreatic cancer by targeting miR-218-5p. Biomed Pharmacother 2019; 114:108862. [PMID: 30981105 DOI: 10.1016/j.biopha.2019.108862] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/31/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Small Nucleolar RNA Host Gene (SNHG16) is a novel cancer-related long noncoding RNA (lncRNA) and functions as an oncogene in a variety of cancers. Nonetheless, the expression patterns, biological function, and potential mechanisms in SNHG16 in pancreatic cancer (PC) remain rarely known. An increase in expression of SNHG16 in PC samples against adjacent normal tissues was shown here. Increased SNHG16 was linked intimately to the tumor-node-metastasis (TNM) stage, distant metastasis, tumor differentiation, and poor overall survival. Loss-of-function experiments revealed that SNHG16 knockdown suppressed the proliferation, formation of colonies, ability to migrate and invade in vitro, along with a lowered growth of the tumor in a mouse model. Mechanistically, SNHG16 might serve as a sponge competitive endogenous RNA (ceRNA) for miR-218-5p, thereby playing a role in regulating the expression of high mobility group box 1 (HMGB1) expression, a known direct miR-218-5p target in PC cells. These results provide novel insight into PC tumorigenesis and suggest that SNHG16 could serve as a likely therapeutic intervention in PC.
Collapse
Affiliation(s)
- Songyang Liu
- Department of Hepatopancreatobiliary Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, PR China
| | - Wei Zhang
- Department of Hepatopancreatobiliary Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, PR China
| | - Kai Liu
- Department of Hepatopancreatobiliary Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, PR China
| | - Yahui Liu
- Department of Hepatopancreatobiliary Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, PR China.
| |
Collapse
|
40
|
Yu Y, Chen F, Yang Y, Jin Y, Shi J, Han S, Chu P, Lu J, Tai J, Wang S, Yang W, Wang H, Guo Y, Ni X. lncRNA SNHG16 is associated with proliferation and poor prognosis of pediatric neuroblastoma. Int J Oncol 2019; 55:93-102. [PMID: 31180520 PMCID: PMC6561620 DOI: 10.3892/ijo.2019.4813] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma (NB) is one of the most common extracranial solid tumors in children, which has complex molecular mechanisms. Increasing evidence has suggested that long noncoding RNAs (lncRNAs) account for NB pathogenesis. However, the function of small nucleolar RNA host gene 16 (SNHG16) in NB is currently unclear. In the present study, publically available data and clinical specimens were employed to verify the expression of SNHG16 in NB. Colony formation, real‑time cell proliferation and migration assays were performed to demonstrate the status of cellular proliferation and migration. Flow cytometry was used to examine cell cycle progression in SH‑SY5Y cells, and acridine orange/ethidium bromide staining and caspase‑3/7 activity measurements were applied to study cell apoptosis. To explore the underlying mechanism of SNHG16 function, an online database was used to identify potential RNA‑binding proteins that bind SNHG16. The expression of SNHG16 was revealed to be in line with the clinical staging of NB, and high SNHG16 expression was positively associated with poor clinical outcome. Furthermore, SNHG16 silencing inhibited cell proliferation, repressed migration, and induced cell cycle arrest at the G0/G1 phase in SH‑SY5Y cells. Additionally, apoptosis was undetectable in SH‑SY5Y cells following SNHG16 silencing. Bioinformatics analysis revealed that SNHG16 regulated cell proliferation in NB through transcriptional and translational pathways. These results suggested that SNHG16 may serve important roles in the development and progression of NB, and could represent a potential target for NB therapy.
Collapse
Affiliation(s)
- Yongbo Yu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Feng Chen
- Department of Functional Neurosurgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Yeran Yang
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Jin Shi
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Shujing Han
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Ping Chu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Jie Lu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Jun Tai
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Shengcai Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Wei Yang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Huanmin Wang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health (NCCH), Beijing 100045, P.R. China
| |
Collapse
|
41
|
Peng H, Li H. The encouraging role of long noncoding RNA small nuclear RNA host gene 16 in epithelial‐mesenchymal transition of bladder cancer via directly acting on miR‐17‐5p/metalloproteinases 3 axis. Mol Carcinog 2019; 58:1465-1480. [DOI: 10.1002/mc.23028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/08/2019] [Accepted: 04/10/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Hao Peng
- Department of Urologic SurgeryZhoukou Central HospitalZhoukou Henan China
| | - Hao Li
- Department of Urologic SurgeryZhoukou Central HospitalZhoukou Henan China
| |
Collapse
|
42
|
Liu H, Chen B, Zhu Q. RETRACTED ARTICLE: Long non-coding RNA SNHG16 reduces hydrogen peroxide-induced cell injury in PC-12 cells by up-regulating microRNA-423-5p. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1444-1451. [PMID: 30977409 DOI: 10.1080/21691401.2019.1600530] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Haochuan Liu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bing Chen
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qingsan Zhu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
Su P, Mu S, Wang Z. Long Noncoding RNA SNHG16 Promotes Osteosarcoma Cells Migration and Invasion via Sponging miRNA-340. DNA Cell Biol 2019; 38:170-175. [PMID: 30726150 DOI: 10.1089/dna.2018.4424] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Long noncoding RNA host gene 16 (SNHG16) has a key role in a variety of cancer progression. However, the role and mechanism of SNHG16 in osteosarcoma (OS) remain unknown. In this study, we examined the functional role of SNHG16 in OS cells through knocked-down SNHG16 by using siRNA. We found that SNHG16 is overexpressed in OS tissues and cell lines. Inhibition of SNHG16 reduced OS cells proliferation, stimulated apoptosis, and decreased migration and invasion. In addition, SNHG16 reduced miR-340 expression in OS cells. The results showed that SNHG16 involves in the migration and invasion of OS cells through sponging miRNA-340. Together, our data support an important role of SNHG16 in regulating OS cell invasion and migration that highlights SNHG16 may be regarded as a potential target for OS treatment.
Collapse
Affiliation(s)
- Pan Su
- 1 Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Henan, China
| | - Shimin Mu
- 1 Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Henan, China
| | - Zhiyuan Wang
- 2 Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Wang X, Kan J, Han J, Zhang W, Bai L, Wu H. LncRNA SNHG16 Functions as an Oncogene by Sponging MiR-135a and Promotes JAK2/STAT3 Signal Pathway in Gastric Cancer. J Cancer 2019; 10:1013-1022. [PMID: 30854107 PMCID: PMC6400807 DOI: 10.7150/jca.29527] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
lncRNA can serve as a miRNA sponge and block the function of miRNA. High expression of lncRNA SNHG16 (small nucleolar RNAhostgene16) was discovered in gastric cancer (GC) and many other tumors. However, the mechanism of SNHG16 in GC is still unclear. In this research, we detected the expression level of SNHG16 in GC tissues and cell lines by qRT-PCR and FISH assay. RIP and Dual Luciferase Reporter Assay revealed that miR-135a is a target of SNHG16. SNHG16 gene knockdown experiment indicated that the expression level of SNHG16 can influence GC cells proliferation, colony formation, invasion ability and apoptosis in a miR-135a dependent manner. Western Blot assay showed that knockdown of SNHG16 decreased the expression of JAK2 and p-STAT3 in GC cells while miR-135a can offset the facilitated impact. Then the expression level of SNHG16 and miR-135a in the si-STAT3 GC cells was detected by qRT-PCR and the results showed that SNHG16 may be a target gene of p-STAT3. Collectively, it was suggested that SNHG16 can serve as a miR-135a sponge and block the function of miR-135a in JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Xinguo Wang
- Department of Oncology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining 810007, Qinghai Province, PR China
| | - Jie Kan
- Department of Oncology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining 810007, Qinghai Province, PR China
| | - Jun Han
- Department of Oncology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining 810007, Qinghai Province, PR China
| | - Wenyan Zhang
- Department of Oncology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining 810007, Qinghai Province, PR China
| | - Liyan Bai
- Department of Oncology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining 810007, Qinghai Province, PR China
| | - Hao Wu
- Department of Oncology, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining 810007, Qinghai Province, PR China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China
| |
Collapse
|
45
|
Wen Q, Zhao L, Wang T, Lv N, Cheng X, Zhang G, Bai L. LncRNA SNHG16 drives proliferation and invasion of papillary thyroid cancer through modulation of miR-497. Onco Targets Ther 2019; 12:699-708. [PMID: 30705598 PMCID: PMC6343509 DOI: 10.2147/ott.s186923] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Long noncoding small nucleolar RNA host gene 16 (SNHG16) has been shown to play an oncogenic role in multiple cancers. However, the biological roles and mechanism of SNHG16 action in the regulation of papillary thyroid cancer (PTC) remains unknown. The aims of this study were to investigate the roles and the possible mechanism of SNHG16 in PTC progression. Materials and methods The expression of SNHG16 PTC tissues and cell lines was detected by reverse-transcription quantitative PCR (qRT-PCR). The effect of SNHG16 on cell proliferation, apoptosis, migration, and invasion was detected by Cell Counting Kit-8, flow cytometry, wound-healing assay, and Matrigel invasion assay, respectively. In addition, the regulatory relationships between SNHG16 and miR-497 were explored by luciferase reporter assay and qRT-PCR. Results The SNHG16 expression was upregulated in PTC tissues and cell lines, whose expression was positively associated with advanced TNM stage and lymph node metastasis. Function analysis demonstrated that depletion of SNHG16 in PTC cells significantly inhibited cell proliferation, induced cell apoptosis, and suppressed cell migration and invasion abilities. Mechanistic studies indicated that SNHG16 functioned as an endogenous sponge for miR-497 to regulate its target genes brain-derived neurotrophic factor and yes-associated protein 1 expression. Furthermore, the inhibition of miR-497 antagonized the suppressive effect of SNHG16-depleted cells on cell proliferation, migration, and invasion. Conclusion These findings revealed that SNHG16 drived the PTC progression possibly via regulating miR-497, suggesting that SNHG16 might be a novel therapeutic agent for PTC.
Collapse
Affiliation(s)
- Qiang Wen
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, ErDao District, Changchun 13033, China,
| | - Lina Zhao
- Department of Thyroid Surgery, China-Japan Union Hospital of Jilin University, ErDao District, Changchun 13033, China,
| | - Tongtong Wang
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, ErDao District, Changchun 13033, China,
| | - Ningning Lv
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, ErDao District, Changchun 13033, China,
| | - Xuejiao Cheng
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, ErDao District, Changchun 13033, China,
| | - Guang Zhang
- Department of Thyroid Surgery, China-Japan Union Hospital of Jilin University, ErDao District, Changchun 13033, China,
| | - Lin Bai
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, ErDao District, Changchun 13033, China,
| |
Collapse
|
46
|
Wang D, Zheng J, Liu X, Xue Y, Liu L, Ma J, He Q, Li Z, Cai H, Liu Y. Knockdown of USF1 Inhibits the Vasculogenic Mimicry of Glioma Cells via Stimulating SNHG16/miR-212-3p and linc00667/miR-429 Axis. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 14:465-482. [PMID: 30743215 PMCID: PMC6369224 DOI: 10.1016/j.omtn.2018.12.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/19/2018] [Accepted: 12/03/2018] [Indexed: 12/16/2022]
Abstract
The anti-angiogenic treatment of malignant glioma cells is an effective method to treat high-grade gliomas. However, due to the presence of vasculogenic mimicry (VM), the anti-angiogenic treatment of gliomas is not significantly effective in improving overall patient median survival. Therefore, this study investigated the mechanism of mimic formation of angiogenesis in gliomas. The results of this experiment indicate that the expression of upstream transcription factor 1 (USF1) is upregulated in glioma tissues and cells. USF1 knockdown inhibits the proliferation, migration, invasion, VM, and expression of VM-associated proteins in glioma cells by stimulating SNHG16 and linc00667. These two long non-coding RNAs (lncRNAs) regulate ALHD1A1 through the competing endogenous RNA (ceRNA) mechanism influencing the VM of glioma. This study is the first to demonstrate that the USF1/SNHG16/miR-212-3p/ALDH1A1 (aldehyde dehydrogenase-1) and USF1/linc00667/miR-429/ALDH1A1 axis regulates the VM of glioma cells, and these findings might provide a novel strategy for glioma treatment.
Collapse
Affiliation(s)
- Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Qianru He
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110122, China; Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, China; Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, China; Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, China; Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, China.
| |
Collapse
|
47
|
Xu F, Zha G, Wu Y, Cai W, Ao J. Overexpressing lncRNA SNHG16 inhibited HCC proliferation and chemoresistance by functionally sponging hsa-miR-93. Onco Targets Ther 2018; 11:8855-8863. [PMID: 30573973 PMCID: PMC6290873 DOI: 10.2147/ott.s182005] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Long noncoding RNAs (lncRNAs) have been identified as prognostic biomarkers and functional regulators in human cancers. The present study aimed to determine the expressions and functions of an lncRNA, Small Nucleolar RNA Host Gene 16 (SNHG16), in hepatocellular carcinoma (HCC). Patients and methods SNHG16 expressions were tested by quantitative real-time PCR (qRT-PCR) in HCC cell lines, as well as 43 pairs of HCC tissues and pair-matched healthy hepatic tissues. It was overexpressed in Hep3B and HuH7 cells. The effects of SNHG16 overexpression in HCC in vitro proliferation, 5-fluorouracil (5-FU) chemoresistance, and in vivo tumor growth were tested. A potential microRNA (miRNA) sponge target of SNHG16, hsa-miR-93, was tested by luciferase reporter assay and qRT-PCR. In addition, hsa-miR-93 was upregulated in SNHG16-overexpressed HCC cells to examine its effect on SNHG16-mediated cancer cell functional regulation in HCC. Results SNHG16 levels were markedly downregulated in both HCC cell lines and HCC tissues. Lentivirus-mediated SNHG16 overexpression inhibited HCC cell proliferation, 5-FU chemoresistance, and in vivo tumor growth. Hsa-miR-93 was confirmed to be directly sponging on SNHG16. Its upregulation in HCC cells reversed SNHG16 overexpression and induced tumor-suppressing effects in HCC cells. Conclusion Our data demonstrate that SNHG16 plays a critical role in HCC development via functionally sponging hsa-miR-93.
Collapse
Affiliation(s)
- Fengfeng Xu
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China,
| | - Guoqing Zha
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanpeng Wu
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China,
| | - Weilong Cai
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China,
| | - Jian Ao
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China,
| |
Collapse
|
48
|
Abstract
BACKGROUND Prostate-cancer-associated ncRNA transcript 1 (PCAT-1), a newly discovered lncRNA, was implicated in the progression of multiple tumors. We conducted a systematic review and meta-analysis to determine its prognostic potential for gastrointestinal cancers. METHODS A literature survey was conducted by searching the PubMed, Web of Science, Cochrane Library, Embase together with Wanfang, and China National Knowledge Infrastructure (CNKI) database for articles published as of October 15, 2017. Hazard ratio (HR) or odds ratio (OR) with 95% confidence intervals (95% CIs) were calculated to demonstrate prognostic value of PCAT-1 using Stata 12.0 software. RESULTS A total of 6 studies with 961 cases were pooled in the analysis to evaluate the prognostic value of PCAT-1 in gastrointestinal cancers. Increased PCAT-1 expression was significantly correlated with poor overall survival (OS) (HR = 1.04, 95% CI: 1.02-1.06). Statistical significance was also observed in subgroup meta-analysis stratified by cancer type, histology type, sample size, and analysis type. Additionally, high expression of PCAT-1 was significantly associated with deeper tumor invasion (OR = 4.46, 95% CI: 3.00-6.63), positive lymph node metastasis (OR = 3.76, 95% CI: 1.39-10.16), and advanced clinical stage (OR = 4.09, 95% CI: 1.55-10.82). CONCLUSION High expression of PCAT-1 was related to poor prognosis and could be a promising biomarker of clinicopathologic features in gastrointestinal cancers. More studies will be necessary to verify and strengthen the clinical value of PCAT-1 in gastrointestinal cancers.
Collapse
Affiliation(s)
- Wanwei Liu
- Department of Hepatobiliary Surgery, Meizhou People's Hospital (Huangtang Hospital), Meizhou, Guangdong Province, China
| | | |
Collapse
|
49
|
Yang T, Jin X, Lan J, Wang W. Long non-coding RNA SNHG16 has Tumor suppressing effect in acute lymphoblastic leukemia by inverse interaction on hsa-miR-124-3p. IUBMB Life 2018; 71:134-142. [PMID: 30380185 DOI: 10.1002/iub.1947] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/26/2018] [Accepted: 09/01/2018] [Indexed: 01/05/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is one of the deadly forms of childhood cancers in the world. In the present study, we used both in vitro and in vivo models to evaluate the functional mechanisms of a long noncoding RNA (lncRNA), small nucleolar RNA host gene 16 (SNHG16) in ALL. SNHG16 gene expression was evaluated by quantitative real-time PCR (qPCR) in both in vitro ALL cell lines and in vivo human samples of T lymphocytes. Lentivirus-mediated SNHG16 downregulation was performed in MOLT3 and SUP-B15 cells, to evaluate its functional effects on ALL cell proliferation, migration in vitro, and ALL transplant in vivo. Epigenetic regulation of SNHG16 on human miR-124-3p (hsa-miR-124-3p) was evaluated by dual-luciferase activity assay and qPCR. Hsa-miR-124-3p was inhibited in SNHG16-downregulated MOLT3 and SUP-B15 cells to further evaluate the functional correlation between SNHG16 and hsa-miR-124-3p in ALL. SNHG16 is upregulated in both in vitro ALL cell lines and in vivo human leukemic T-cells. SNHG16 downregulation suppressed ALL proliferation and migration in vitro, and ALL explant in vivo. Hsa-miR-124-3p was demonstrated to interact with SNHG16, and upregulated in SNHG16-downregulated ALL cells. In addition, inhibiting hsa-miR-124-3p reversed SNHG16-downregulation-mediated tumor suppressive functions in ALL. SNHG16 is upregulated in ALL, and its inhibition has tumor suppressive effect in ALL, likely through epigenetic interaction on hsa-miR-124-3p. © 2018 IUBMB Life, 71(1):134-142, 2019.
Collapse
Affiliation(s)
- Tianxin Yang
- Department of Hematology, Zhejiang Province People's Hospital, Hangzhou, 310014, China
| | - Xing Jin
- Department of Hematology, Zhejiang Province People's Hospital, Hangzhou, 310014, China
| | - Jianping Lan
- Department of Hematology, Zhejiang Province People's Hospital, Hangzhou, 310014, China
| | - Wensong Wang
- Department of Hematology, Zhejiang Province People's Hospital, Hangzhou, 310014, China
| |
Collapse
|
50
|
Prognostic Significance of Pretreatment Apolipoprotein A-I as a Noninvasive Biomarker in Cancer Survivors: A Meta-Analysis. DISEASE MARKERS 2018; 2018:1034037. [PMID: 30510601 PMCID: PMC6232830 DOI: 10.1155/2018/1034037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 09/13/2018] [Accepted: 09/20/2018] [Indexed: 01/17/2023]
Abstract
Background Numerous studies have reported the prognostic significance of serum apolipoprotein A-I (ApoA-I) in various cancers, but the results have been inconsistent. The current meta-analysis was performed to investigate the association between ApoA-I level and prognosis in human malignancies. Methods A literature search was performed using the electronic platforms of the PubMed, Cochrane Library, Web of Science, Embase, Wanfang, and China National Knowledge Infrastructure (CNKI) databases to obtain eligible articles published up to May 20, 2018. Pooled hazard ratios (HRs) with 95% confidence intervals (95% CIs) were calculated to assess the prognostic values of the ApoA-I level in cancers using STATA 12.0 software. Results A total of 14 studies involving 9295 patients were included. The results indicated that low ApoA-I level was significantly associated with poor overall survival (OS) (HR = 0.52, 95% CI: 0.44-0.61). Significant relationships between the ApoA-I level and OS were specifically detected in nasopharyngeal carcinoma (NPC, HR = 0.63, 95% CI: 0.54-0.73), colorectal cancer (CRC, HR = 0.48, 95% CI: 0.19-0.76), and hepatocellular carcinoma (HCC, HR = 0.46, 95% CI: 0.27-0.65). The subgroup analyses for OS also further confirmed the prognostic significance of the ApoA-I level in cancers. Moreover, lower Apo A-I was associated with unfavorable cancer-specific survival (CSS, HR: 0.47, 95% CI: 0.19-0.76) in cancers, and low ApoA-I level was clearly associated with inferior total time to recurrence (TTR, HR: 0.43, 95% CI: 0.29-0.58) in HCC, poorer locoregional recurrence-free survival (LRFS) and distant metastasis-free survival (DMFS) (HR: 0.58, 95% CI: 0.42-0.74 for LRFS; HR: 0.65, 95% CI: 0.41-0.89 for DMFS) in NPC, and shorter disease-free survival (DFS, HR: 0.64, 95% CI: 0.43-0.84) in cancers. Conclusions. Low ApoA-I level might be an unfavorable prognostic factor in multiple malignancies, and serum ApoA-I could serve as a noninvasive marker to predict cancer prognosis.
Collapse
|