1
|
Yu Y, He N, Song Z, Wang C, Xu J. LIMD1-AS1 promotes the progression of prostate cancer and affects the function of prostate cancer cells by down-regulating miR-29c-3p. J Cancer Res Clin Oncol 2024; 151:5. [PMID: 39636414 PMCID: PMC11621152 DOI: 10.1007/s00432-024-06046-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE Prostate cancer (PCa) is a prevalent and lethal malignancy affecting males, with a considerable proportion of patients experiencing poor survival outcomes. The regulatory role of LIMD1-AS1 in the initiation and progression of PCa is emerging as a significant factor, however, the precise mechanisms governing its influence are yet to be fully elucidated. METHODS qRT-PCR was employed to assess the expression of LIMD1-AS1 and miR-29c-3p. The Cell Counting Kit-8 (CCK-8) was used to assess cell proliferation in PCa cells. Apoptosis rates were determined using flow cytometry. Cell migration and invasion were evaluated using the transwell assay. The targeted relationship of LIMD1-AS1 and miR-29c-3p was confirmed through dual-luciferase reporter gene analysis. RESULTS Increased expression of LIMD1-AS1 and decreased expression of miR-29c-3p were observed in both tumor tissues and serum from PCa patients. LIMD1-AS1 exhibited diagnostic and prognostic significance in PCa patients. Functionally, LIMD1-AS1 modulated the expression of miR-29c-3p to potentiate the proliferative, migratory, and invasive capabilities of PCa cells while concurrently inhibiting apoptosis. CONCLUSION LncRNA LIMD1-AS1 promotes the advancement of PCa by regulating miR-29c-3p, indicating that LIMD1-AS1/miR-29c-3p axis could serve as potential therapeutic targets for the therapeutic intervention of PCa.
Collapse
Affiliation(s)
- Yongsheng Yu
- Department of Urinary Surgery, The Fourth Affiliated Hospital of Guangzhou Medical University, Guangdong, 511300, China
| | - Nan He
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Hubei, 430071, China
| | - Zhaolu Song
- Department of Urology Surgery, Jiaozhou Central Hospital of Qingdao, Shandong, 266300, China
| | - Chang Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan Second Road, Guangzhou City, Guangdong Province, 510080, China.
| | - Jinhuang Xu
- Department of Trauma and Joint Surgery, The Fourth Affiliated Hospital of Guangzhou Medical University, No. 1, Guangming East Road, Zengcheng District, Guangzhou City, Guangdong Province, 511300, China.
| |
Collapse
|
2
|
Zhu Z, Tang G, Shi M, Fang M, Zhang X, Xu H. Identification of the Oncogenic Role of the Circ_0001326/miR-577/VDAC1 Cascade in Prostate Cancer. J Biochem Mol Toxicol 2024; 38:e70034. [PMID: 39555732 DOI: 10.1002/jbt.70034] [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: 06/30/2024] [Revised: 09/19/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024]
Abstract
Prostate cancer (PCa) is one of the leading causes of cancer death among men worldwide. Circular RNAs (circRNAs) have been implicated in the pathogenesis of PCa. However, the precise action of circ_0001326 in PCa malignant progression is still unknown. The levels of circ_0001326, miR-577 and voltage dependent anion channel 1 (VDAC1) were determined by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Cell proliferation, colony formation, apoptosis, migration and invasion were evaluated by the Cell Counting Kit-8 (CCK-8), EdU staining, colony formation, flow cytometry, wound-healing and transwell assays, respectively. Targeted relationships among circ_0001326, miR-577 and VDAC1 were confirmed by dual-luciferase reporter assays. Xenograft experiments were performed to detect the role of circ_0001326 in tumor growth. Our data revealed that circ_0001326 was overexpressed in PCa tissues and cells. Circ_0001326 depletion repressed PCa cell proliferation, migration, and invasion and enhanced apoptosis in vitro, as well as hampered tumor growth in vivo. Mechanistically, circ_0001326 directly targeted miR-577, and VDAC1 was directly targeted and suppressed by miR-577. Moreover, the effects of circ_0001326 knockdown on PCa cell functional behaviors were mediated by miR-577. VDAC1 silencing phenocopied miR-577 overexpression in regulating PCa cell functional behaviors in vitro. Furthermore, circ_0001326 regulated VDAC1 expression through sponging miR-577. Our findings showed that circ_0001326 regulated PCa cell functional behaviors at least partly through targeting the miR-577/VDAC1 axis.
Collapse
Affiliation(s)
- Zhirong Zhu
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| | - Guiliang Tang
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| | - Mengqi Shi
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| | - Mengjie Fang
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| | - Xiaolong Zhang
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| | - Huali Xu
- Department of Urology, Shaoxing People's Hospital, Shaoxing, China
| |
Collapse
|
3
|
Qi XL, Luo GQ, Tuersun A, Chen M, Wu GB, Zheng L, Li HJ, Lou XL, Luo M. Construction of an endoplasmic reticulum stress and cuproptosis -related lncRNAs signature in chemosensitivity in hepatocellular carcinoma by comprehensive bioinformatics analysis. Heliyon 2024; 10:e38342. [PMID: 39398070 PMCID: PMC11471205 DOI: 10.1016/j.heliyon.2024.e38342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Endoplasmic reticulum stress (ERS) and cuproptosis have remarkable effects on hepatocellular carcinoma (HCC) leading to a poor prognosis. The current study aimed to explore credible signature for predicting the prognosis of HCC based on ERS and cuproptosis-related lncRNAs. In our study, clinical and transcriptomic profiles of HCC patients were obtained from the Cancer Genome Atlas (TCGA) database. An ERS and cuproptosis-related lncRNA prognostic signature, including NRAV, SNHG3, LINC00839 and AC004687.1, was determined by correlation tests, Cox regression analysis, least absolute shrinkage, and selection operator (LASSO) methods. Survival and predictive value were evaluated using Kaplan-Meier and receiver operating characteristic (ROC) curves, while calibration and nomograms curves were developed. Besides the enrichment analyses for ERS and cuproptosis-related lncRNAs, mutational status and immune status were assessed with TMB and ESTIMATE. Additionally, consensus cluster analysis was employed to compare cancer subtype differences, while drug sensitivity and immunologic efficacy were evaluated for further exploration. qRT-PCR and CCK-8 were utilized to verify the alteration of the prognostic lncRNAs expression and proliferation in vitro. High-risk groups exhibited poorer prognosis. The signature exhibited robust predictive value as an independent prognostic indicator and showed significant correlation with clinicopathological features. In the enriched analysis, biological membrane pathways were enriched. Low-risk patients had lower TMB and higher immune status. A cluster analysis revealed that cluster 2 had the best clinical immunological efficacy and most active immune function. In brief, our constructed signature with ERS and cuproptosis-related lncRNAs was associated survival outcomes of HCC, and can be used to predict the clinical classification and curative effect.
Collapse
Affiliation(s)
- Xiao-Liang Qi
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gu-Qing Luo
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Abudukadier Tuersun
- Department of General Surgery, Kashgar Prefecture Second People's Hospital of Xinjiang Uygur Autonomous Regions, Kashgar, Xinjiang, China
| | - Min Chen
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang-Bo Wu
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zheng
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Jie Li
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Lou Lou
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Luo
- Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Peng Y, Long XD. The role of the ceRNA network mediated by lncRNA SNHG3 in the progression of cancer. Discov Oncol 2024; 15:514. [PMID: 39349640 PMCID: PMC11442963 DOI: 10.1007/s12672-024-01184-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/22/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are a distinct class of RNAs with longer than 200 base pairs that are not translated into proteins. Small Nucleolar RNA Host Gene 3 (SNHG3) is a lncRNA and frequently dysregulated in various human cancers. OBJECTIVE This review provides a comprehensive analysis of current research on lncRNA SNHG3, focusing on its role within the competitive endogenous RNA (ceRNA) network and its implications in cancer. METHODS A systematic literature review was conducted using PubMed up to October 2023. The search strategy included keywords such as "lncRNA SNHG3", "competitive endogenous RNA", "cancer", and related terms. Studies were selected based on relevance to SNHG3's involvement in cancer pathogenesis and progression. RESULTS Disruptions in the ceRNA network involving lncRNA SNHG3 can impair normal cell growth and differentiation, significantly contributing to disease pathogenesis, particularly cancer. This review highlights SNHG3's substantial impact on various cancer processes and its potential as a diagnostic and therapeutic tool for aggressive cancers. CONCLUSION The findings underscore SNHG3's pivotal role in cancer prevention, diagnosis, and treatment, laying a foundation for future research in cancer management. Insights from this review emphasize the necessity for further exploration and development of SNHG3-based diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Ying Peng
- Department of Pathology, the First Affiliated Hospital, Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China
- Department of Pathology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518000, Guangdong, People's Republic of China
- Department of Pathology, the Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, Baise, 533000, People's Republic of China
| | - Xi-Dai Long
- Department of Pathology, the Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, Baise, 533000, People's Republic of China.
- Department of Tumor Pathology, Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Guangxi Zhuang Autonomous Region, Baise, 533000, China.
| |
Collapse
|
5
|
Khalilollah S, Kalantari Soltanieh S, Obaid Saleh R, Ali Alzahrani A, Ghaleb Maabreh H, Mazin Al-Hamdani M, Dehghani-Ghorbi M, Shafiei Khonachaei M, Akhavan-Sigari R. LncRNAs involvement in pathogenesis of immune-related disease via regulation of T regulatory cells, an updated review. Cytokine 2024; 179:156585. [PMID: 38579428 DOI: 10.1016/j.cyto.2024.156585] [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: 05/24/2023] [Revised: 03/03/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024]
Abstract
The pathophysiology of several illnesses, including cancer and autoimmune diseasesdepends on human regulatory T cells (Tregs), and abnormalities in these cells may function as triggers for these conditions. Cancer and autoimmune, and gynecological diseases are associated with the differentiation of the proinflammatory T cell subset TH17 and its balance with the production of Treg. Recently, long non-coding RNAs (lncRNAs) have become important regulatory molecules in a wide range of illnesses. During epigenetic regulation, they can control the expression of important genes at several levels by affecting transcription, post-transcriptional actions, translation, and protein modification. They might connect with different molecules, such as proteins, DNA and RNA, and their structural composition is intricate. Because lncRNAs regulatebiological processes, including cell division, death, and growth, they are linked to severaldiseases. A notable instance of this is the lncRNA NEAT1, which has been the subject of several investigations to ascertain its function in immune cell development. In the context of immune cell development, several additional lncRNAs have been connected to Treg cell differentiation. In this work, we summarize current findings about the diverse functions of lncRNAs in Treg cell differentiation and control of the Th17/Treg homeostasis in autoimmune disorders, cancers, as well as several gynecological diseases where Tregs are key players.
Collapse
Affiliation(s)
- Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | | | - Raed Obaid Saleh
- Department of Pathological Analysis, College of Applied Science, University of Fallujah, Al-Anbar, Iraq.
| | | | - Hatem Ghaleb Maabreh
- Department of Dermatovenerology, Foreign Languages, RUDN University (Peoples' Friendship University of Russia named after Patrice Lumumba), Moscow, Russia.
| | | | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Poland.
| |
Collapse
|
6
|
Xu D, Feng H, Ren Z, Li X, Jiang C, Chen Y, Liu L, Chen W, Cui Z, Cang S. SNHG3/WISP2 Axis Promotes Hela Cell Migration and Invasion via Activating Wnt/β-Catenin Signaling. Cancer Genomics Proteomics 2023; 20:744-753. [PMID: 38035707 PMCID: PMC10687733 DOI: 10.21873/cgp.20421] [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: 08/01/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND/AIM Cervical cancer (CC) poses a significant threat to women's health and has a relatively poor prognosis due to local invasion and metastasis. It is, therefore, crucial to elucidate the molecular mechanisms of CC metastasis. SNHG3 has been implicated in various tumor metastasis processes, but its involvement in CC has not been thoroughly studied. Our study aimed to investigate the role of SNHG3 in metastasis and elucidate its underlying mechanisms in CC. MATERIALS AND METHODS LncRNA SNHG3 expression in CC tissues was analyzed using TCGA and GSE27469 databases. Normal cervical epithelial cells and CC cell lines were used to detect mRNA expression of SNHG3 via quantitative reverse transcription polymerase chain reaction (qRT-PCR). With RNA interference (RNAi) technology, antisense oligonucleotides (ASO) can act on HeLa cells to knockdown target gene expression. The influence of SNHG3 on cell migration and invasion were determined by wound healing and transwell assays. Transcriptome sequencing (RNA-seq) was used to seek abnormally expressed genes between SNHG3 knockdown cells and control cells. The expressions of epithelial-mesenchymal transition (EMT) and Wnt/β-catenin signaling related proteins were detected using western blot. RESULTS SNHG3 was obviously up-regulated in CC tissues and cell lines, and ectopic expression of SNHG3 was associated with lymph node metastasis of CC. Knockdown of SNHG3 significantly inhibited cell migration and invasion in CC. Further molecular mechanism studies showed that SNHG3 knockdown could down-regulate the expression of WNT1 Inducible Signaling Pathway Protein 2 (WISP2) so as to inhibit the activation of the Wnt/β-catenin signaling pathway, and regulated the expression of EMT-related markers, that promoted the protein expression of E-cadherin, as well as decreased the expression of N-cadherin and vimentin. CONCLUSION SNHG3 appears to exert a pro-metastatic effect in CC, as evidenced by inhibition of cell migration and invasion upon SNHG3 knockdown. EMT also appears to be attenuated. Of interest is the down-regulation of WISP2 following SNHG3 knockdown leads to the inactivation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Dengfei Xu
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Zheng Zhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Hao Feng
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Zirui Ren
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Zheng Zhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Xiang Li
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Zheng Zhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Chenyang Jiang
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Zheng Zhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Yuming Chen
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Zheng Zhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Lina Liu
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Zheng Zhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, P.R. China
| | - Wenchao Chen
- Department of Gastrointestinal Surgery, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, P.R. China
| | - Zhilei Cui
- Department of Respiratory Medicine, XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Shundong Cang
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Zheng Zhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, P.R. China;
| |
Collapse
|
7
|
Li W, Li D, Ma Q, Chen Y, Hu Z, Bai Y, Xie L. Targeted inhibition of mTOR by BML-275 induces mitochondrial-mediated apoptosis and autophagy in prostate cancer. Eur J Pharmacol 2023; 957:176035. [PMID: 37657741 DOI: 10.1016/j.ejphar.2023.176035] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer among men and the second leading cause of death in Western countries. Clinically, screening drugs and develop developing new therapeutics to treat PCa is of great significance. In this study, BML-275 was demonstrated to exert potent antitumor effects in PCa by antagonizing mTOR activity. In cultured PCa cells, BML-275 treatment reduced the expression levels of c-Myc and survivin, promoted the activation of p53, and thereby induced p21/cyclin D1/CDK4/6-dependent cell cycle G1/S arrest. As a result, BML-275 inhibited cellular proliferation and induced mitochondrial-mediated apoptosis. In addition, BML-275 treatment triggered autophagy. Interestingly, EACC-mediated suppression of autophagy did not affect BML-275-induced proliferation and apoptosis. Nude mouse tumorigenic experiments also confirmed that BML-275 inhibited PCa growth, induced PCa cell apoptosis and autophagy. Mechanistically, the activities of PI3K/AKT and AMPK pathways were downregulated by BML-275 treatment in vitro and in vivo. Importantly, mTOR, a common downstream negative protein of PI3K/AKT and AMPK signaling, was induced to inactivate, which may be associated with the induction of apoptosis and autophagy. The pharmacological activation of mTOR by MHY1485 abolished the induction of apoptosis and autophagy of BML-275. Molecular docking results showed that BML-275 can bind to the FKRP12-rapamycin binding site on mTOR protein, and thereby may have the same inhibitory activity on mTOR as rapamycin. Thus, these findings indicated that BML-275 induces mitochondrial-mediated apoptosis and autophagy in PCa by targeting mTOR inhibition. BML-275 may be a potential candidate for the treatment of PCa.
Collapse
Affiliation(s)
- Wangjian Li
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China; Department of Urology, The Central Hospital Affiliated to Shaoxing University, Shaoxing, 312030, China
| | - Dongzhang Li
- Department of Urology, The Central Hospital Affiliated to Shaoxing University, Shaoxing, 312030, China
| | - Quan Ma
- Department of Urology, The Central Hospital Affiliated to Shaoxing University, Shaoxing, 312030, China
| | - Yongliang Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zujian Hu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Liping Xie
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
8
|
Huldani H, Gandla K, Asiri M, Romero-Parra RM, Alsalamy A, Hjazi A, Najm MAA, Fawaz A, Hussien BM, Singh R. A comprehensive insight into the role of small nucleolar RNAs (snoRNAs) and SNHGs in human cancers. Pathol Res Pract 2023; 249:154679. [PMID: 37567032 DOI: 10.1016/j.prp.2023.154679] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 08/13/2023]
Abstract
Long non-coding RNAs (lncRNAs), which comprise most non-coding RNAs (ncRNAs), have recently become a focus of cancer research. How many functional ncRNAs exist is still a matter of debate. Although insufficient evidence supports that most lncRNAs function as transcriptional by-products, it is widely known that an increasing number of lncRNAs play essential roles in cells. Small nucleolar RNAs (snoRNAs), 60-300 nucleotides in length, have been better studied than long non-coding RNAs (lncRNAs) and are predominantly present in the nucleolus. Most snoRNAs are encoded in introns of protein- and non-protein-coding genes called small nucleolar RNA host genes (SNHGs). In this article, we explore the biology and characteristics of SNHGs and their role in developing human malignancies. In addition, we provide an update on the ability of these snoRNAs to serve as prognostic and diagnostic variables in various forms of cancer.
Collapse
Affiliation(s)
- Huldani Huldani
- Department of Physiology, Faculty of Medicine, Lambung Mangkurat University, Banjarmasin, South Kalimantan, Indonesia
| | - Kumaraswamy Gandla
- Department of Pharmaceutical Analysis, Chaitanya Deemed to be University, Hanamkonda, India.
| | - Mohammed Asiri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Ali Alsalamy
- College of Medical Technology, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mazin A A Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Albab Fawaz
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Beneen M Hussien
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Rajesh Singh
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun 248007, India
| |
Collapse
|
9
|
Tang J, Huang XX. Knockdown of long non-coding RNA SNHG3 inhibits proliferation, migration and invasion of human thyroid cancer via miR-339-5p/GPR62 axis. Heliyon 2023; 9:e19713. [PMID: 37809703 PMCID: PMC10559012 DOI: 10.1016/j.heliyon.2023.e19713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Previous studies have implicated SNHG3, a long non-coding RNA, in various human cancers, suggesting its oncogenic role. However, its specific involvement in thyroid cancer and the underlying molecular mechanisms remain unclear. Therefore, this study aims to elucidate the role of SNHG3 in human thyroid cancer and its interaction with the miR-339-5p/GPR62 axis. Understanding these mechanisms could provide insights into potential therapeutic targets for managing thyroid cancer. Results revealed significant upregulation of SNHG3 in human thyroid cancer tissues and cell lines. Knockdown of SNHG3 significantly suppressed proliferation, migration and invasion of CUTC5 and IHH-4 thyroid cancer cells. Knockdown of SNHG3 induces apoptosis in CUTC5 and IHH-4 cells and also inhibits the growth of xenografted tumors in vivo. Different in vitro assays revealed the interaction of SNHG3 with microRNA-339-5p (miR-339-5p) in thyroid cancer cells. Expression of miR-339-5p was significantly downregulated in thyroid cancer tissues and cell lines. However, the knockdown of SNHG3 caused significant upregulation of miR-339-5p. Interestingly, overexpression of miR-339-5p exerted tumor-suppressive effects in CUTC5 and IHH-4 cells via post-transcriptional suppression of GPR62. Knockdown of GPR62 significantly inhibited the proliferation, migration and invasion of CUTC5 and IHH-4 cells. Nonetheless, inhibition of miR-339-5p or overexpression of GPR62 avoids the growth inhibitory effects of SNHG3 knockdown in CUTC5 and IHH-4 cells. Results indicated that SNHG3 exerts oncogenic molecular function in thyroid cancer via miR-339-5p/GPR62 axis and may act as a therapeutic target for its management.
Collapse
Affiliation(s)
- Jin Tang
- Department of Clinical Laboratory, Hanzhong Central Hospital, Hanzhong 723000, Shaanxi, China
| | - Xiao-xia Huang
- Department of Clinical Laboratory, Hanzhong Central Hospital, Hanzhong 723000, Shaanxi, China
| |
Collapse
|
10
|
Datkhayev UM, Rakhmetova V, Shepetov AM, Kodasbayev A, Datkayeva GM, Pazilov SB, Farooqi AA. Unraveling the Complex Web of Mechanistic Regulation of Versatile NEDD4 Family by Non-Coding RNAs in Carcinogenesis and Metastasis: From Cell Culture Studies to Animal Models. Cancers (Basel) 2023; 15:3971. [PMID: 37568787 PMCID: PMC10417118 DOI: 10.3390/cancers15153971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 08/13/2023] Open
Abstract
Discoveries related to an intriguing feature of ubiquitination have prompted a detailed analysis of the ubiquitination patterns in malignant cells. How the "ubiquitinome" is reshaped during multistage carcinogenesis has garnered significant attention. Seminal studies related to the structural and functional characterization of NEDD4 (Neuronal precursor cell-expressed developmentally downregulated-4) have consolidated our understanding at a new level of maturity. Additionally, regulatory roles of non-coding RNAs have further complicated the complex interplay between non-coding RNAs and the members of NEDD4 family. These mechanisms range from the miRNA-mediated targeting of NEDD4 family members to the regulation of transcriptional factors for a broader range of non-coding RNAs. Additionally, the NEDD4-mediated degradation of different proteins is modulated by lncRNAs and circRNAs. The miRNA-mediated targeting of NEDD4 family members is also regulated by circRNAs. Tremendous advancements have been made in the identification of different substrates of NEDD4 family and in the comprehensive analysis of the molecular mechanisms by which various members of NEDD4 family catalyze the ubiquitination of substrates. In this review, we have attempted to summarize the multifunctional roles of the NEDD4 family in cancer biology, and how different non-coding RNAs modulate these NEDD4 family members in the regulation of cancer. Future molecular studies should focus on the investigation of a broader drug design space and expand the scope of accessible targets for the inhibition/prevention of metastasis.
Collapse
Affiliation(s)
- Ubaidilla M. Datkhayev
- Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan
| | | | - Abay M. Shepetov
- Department of Nephrology, Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan;
| | - Almat Kodasbayev
- Department of Cardiovascular Surgery, Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan
| | | | - Sabit B. Pazilov
- Department of Healthcare of Kyzylorda Region, Kyzylorda, Abay Avenue, 27, Kyzylorda 120008, Kazakhstan;
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan
| |
Collapse
|
11
|
Shree B, Das K, Sharma V. Emerging role of transforming growth factor-β-regulated long non-coding RNAs in prostate cancer pathogenesis. CANCER PATHOGENESIS AND THERAPY 2023; 1:195-204. [PMID: 38327834 PMCID: PMC10846338 DOI: 10.1016/j.cpt.2022.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/01/2022] [Accepted: 12/20/2022] [Indexed: 02/09/2024]
Abstract
Prostate cancer (PCa) is the most common malignancy in men. Despite aggressive therapy involving surgery and hormonal treatments, the recurrence and emergence of metastatic castration-resistant prostate cancer (CRPCa) remain a major challenge. Dysregulation of the transforming growth factor-β (TGF-β) signaling pathway is crucial to PCa development and progression. This also contributes to androgen receptor activation and the emergence of CRPC. In addition, TGF-β signaling regulates long non-coding RNA (lncRNA) expression in multiple cancers, including PCa. Here, we discuss the complex regulatory network of lncRNAs and TGF-β signaling in PCa and their potential applications in diagnosing, prognosis, and treating PCa. Further investigations on the role of lncRNAs in the TGF-β pathway will help to better understand PCa pathogenesis.
Collapse
Affiliation(s)
- Bakhya Shree
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Koyel Das
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Vivek Sharma
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad 500078, India
| |
Collapse
|
12
|
Tian X, Chen Y, Peng Z, Lin Q, Sun A. NEDD4 E3 ubiquitin ligases: promising biomarkers and therapeutic targets for cancer. Biochem Pharmacol 2023:115641. [PMID: 37307883 DOI: 10.1016/j.bcp.2023.115641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
Accumulating evidence has demonstrated that NEDD4 E3 ubiquitin ligase family plays a pivotal oncogenic role in a variety of malignancies via mediating ubiquitin dependent degradation processes. Moreover, aberrant expression of NEDD4 E3 ubiquitin ligases is often indicative of cancer progression and correlated with poor prognosis. In this review, we are going to address association of expression of NEDD4 E3 ubiquitin ligases with cancers, the signaling pathways and the molecular mechanisms by which the NEDD4 E3 ubiquitin ligases regulate oncogenesis and progression, and the therapies targeting the NEDD4 E3 ubiquitin ligases. This review provides the systematic and comprehensive summary of the latest research status of E3 ubiquitin ligases in the NEDD4 subfamily, and proposes that NEDD4 family E3 ubiquitin ligases are promising anti-cancer drug targets, aiming to provide research direction for clinical targeting of NEDD4 E3 ubiquitin ligase therapy.
Collapse
Affiliation(s)
- Xianyan Tian
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, China
| | - Yifei Chen
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, China
| | - Ziluo Peng
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, China
| | - Qiong Lin
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, China
| | - Aiqin Sun
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, China.
| |
Collapse
|
13
|
Taheri M, Badrlou E, Hussen BM, Kashi AH, Ghafouri-Fard S, Baniahmad A. Importance of long non-coding RNAs in the pathogenesis, diagnosis, and treatment of prostate cancer. Front Oncol 2023; 13:1123101. [PMID: 37025585 PMCID: PMC10070735 DOI: 10.3389/fonc.2023.1123101] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are regulatory transcripts with essential roles in the pathogenesis of almost all types of cancers, including prostate cancer. They can act as either oncogenic lncRNAs or tumor suppressor ones in prostate cancer. Small nucleolar RNA host genes are among the mostly assessed oncogenic lncRNAs in this cancer. PCA3 is an example of oncogenic lncRNAs that has been approved as a diagnostic marker in prostate cancer. A number of well-known oncogenic lncRNAs in other cancers such as DANCR, MALAT1, CCAT1, PVT1, TUG1 and NEAT1 have also been shown to act as oncogenes in prostate cancer. On the other hand, LINC00893, LINC01679, MIR22HG, RP1-59D14.5, MAGI2-AS3, NXTAR, FGF14-AS2 and ADAMTS9-AS1 are among lncRNAs that act as tumor suppressors in prostate cancer. LncRNAs can contribute to the pathogenesis of prostate cancer via modulation of androgen receptor (AR) signaling, ubiquitin-proteasome degradation process of AR or other important signaling pathways. The current review summarizes the role of lncRNAs in the evolution of prostate cancer with an especial focus on their importance in design of novel biomarker panels and therapeutic targets.
Collapse
Affiliation(s)
- Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Badrlou
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan, Iraq
| | - Amir Hossein Kashi
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| |
Collapse
|
14
|
Song S, Zhu Y, Zhang X, Chen S, Liu S. Prognostic values of long noncoding RNA in bone metastasis of prostate cancer: A systematic review and meta-analysis. Front Oncol 2023; 13:1085464. [PMID: 36890836 PMCID: PMC9986415 DOI: 10.3389/fonc.2023.1085464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/24/2023] [Indexed: 02/22/2023] Open
Abstract
Introduction Recent studies have shown that long non-coding RNAs are closely related to the occurrence and development of prostate cancer bone metastasis, and can be used as biomarkers to predict the prognosis of patients. Therefore, this study aimed to systematically evaluate the relationship between the expression levels of long non-coding RNAs and the prognosis of patients. Methods The studies of lncRNA in prostate cancer bone metastasis from Pubmed, Cochrane library, Embase, Ebsco, Web of science, Scopus, Ovid databases were analyzed, and Stata 15 was used for meta-analysis. Associations between lncRNA expression and patients' overall survival (OS) and bone metastasis-free survival (BMFS) were assessed by correlation analysis with pooled hazard ratios (HR) and 95% confidence intervals (CI). Furthermore, the results were validated using GEPIA2 and UALCAN, online database based on TCGA. Subsequently, the molecular mechanisms of the included lncRNAs were predicted based on the LncACTdb 3.0 database and the lnCAR database. Finally, we used clinical samples to validate lncRNAs that were significantly different in both databases. Results A total of 5 published studies involving 474 patients were included in this meta-analysis. The results showed that lncRNA overexpression was significantly associated with lower OS (HR = 2.55, 95% CI: 1.69 - 3.99, p < 0.05) and lower BMFS (OR = 3.16, 95% CI: 1.90 - 5.27, p < 0.05) in patients with prostate cancer bone metastasis. Based on validation from the GEPIA2 and UALCAN online databases, SNHG3 and NEAT1 were significantly up-regulated in prostate cancer. Further functional prediction showed that the lncRNAs included in the study were involved in regulating the occurrence and development of prostate cancer through the ceRNA axis. The result of clinical samples showed that SNHG3 and NEAT1 were expressed in prostate cancer bone metastasis at higher levels than in primary tumors. Conclusions LncRNA can be used as a novel predictive biomarker for predicting poor prognosis in patients with prostate cancer bone metastasis, which is worthy of clinical validation.
Collapse
Affiliation(s)
- Silu Song
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Department of Basic Medical, Jiamusi University, Jiamusi, China
| | - Yanli Zhu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Department of Basic Medical, Jiamusi University, Jiamusi, China
| | - Xue Zhang
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Department of Basic Medical, Jiamusi University, Jiamusi, China
| | - Siyu Chen
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Department of Basic Medical, Jiamusi University, Jiamusi, China
| | - Shuang Liu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Department of Basic Medical, Jiamusi University, Jiamusi, China
| |
Collapse
|
15
|
Guan Y, Yue S, Chen Y, Pan Y, An L, Du H, Liang C. Molecular Cluster Mining of Adrenocortical Carcinoma via Multi-Omics Data Analysis Aids Precise Clinical Therapy. Cells 2022; 11:cells11233784. [PMID: 36497046 PMCID: PMC9737968 DOI: 10.3390/cells11233784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a malignancy of the endocrine system. We collected clinical and pathological features, genomic mutations, DNA methylation profiles, and mRNA, lncRNA, microRNA, and somatic mutations in ACC patients from the TCGA, GSE19750, GSE33371, and GSE49278 cohorts. Based on the MOVICS algorithm, the patients were divided into ACC1-3 subtypes by comprehensive multi-omics data analysis. We found that immune-related pathways were more activated, and drug metabolism pathways were enriched in ACC1 subtype patients. Furthermore, ACC1 patients were sensitive to PD-1 immunotherapy and had the lowest sensitivity to chemotherapeutic drugs. Patients with the ACC2 subtype had the worst survival prognosis and the highest tumor-mutation rate. Meanwhile, cell-cycle-related pathways, amino-acid-synthesis pathways, and immunosuppressive cells were enriched in ACC2 patients. Steroid and cholesterol biosynthetic pathways were enriched in patients with the ACC3 subtype. DNA-repair-related pathways were enriched in subtypes ACC2 and ACC3. The sensitivity of the ACC2 subtype to cisplatin, doxorubicin, gemcitabine, and etoposide was better than that of the other two subtypes. For 5-fluorouracil, there was no significant difference in sensitivity to paclitaxel between the three groups. A comprehensive analysis of multi-omics data will provide new clues for the prognosis and treatment of patients with ACC.
Collapse
Affiliation(s)
- Yu Guan
- Department of Urology, The First Affifiliated Hospital of Anhui Medical University, 218th Jixi Road, Hefei 230022, China
- Institute of Urology, Anhui Medical University, 81th Meishan Road, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University (AHMU), 81th Meishan Road, Hefei 230022, China
| | - Shaoyu Yue
- Department of Urology, The First Affifiliated Hospital of Anhui Medical University, 218th Jixi Road, Hefei 230022, China
- Institute of Urology, Anhui Medical University, 81th Meishan Road, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University (AHMU), 81th Meishan Road, Hefei 230022, China
| | - Yiding Chen
- Department of Urology, The First Affifiliated Hospital of Anhui Medical University, 218th Jixi Road, Hefei 230022, China
- Institute of Urology, Anhui Medical University, 81th Meishan Road, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University (AHMU), 81th Meishan Road, Hefei 230022, China
| | - Yuetian Pan
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, D-81377 Munich, Germany
| | - Lingxuan An
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, D-81377 Munich, Germany
| | - Hexi Du
- Department of Urology, The First Affifiliated Hospital of Anhui Medical University, 218th Jixi Road, Hefei 230022, China
- Institute of Urology, Anhui Medical University, 81th Meishan Road, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University (AHMU), 81th Meishan Road, Hefei 230022, China
- Correspondence: (H.D.); (C.L.); Tel.: +86-18856040979 (H.D.); +86-13505604595 (C.L.)
| | - Chaozhao Liang
- Department of Urology, The First Affifiliated Hospital of Anhui Medical University, 218th Jixi Road, Hefei 230022, China
- Institute of Urology, Anhui Medical University, 81th Meishan Road, Hefei 230022, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University (AHMU), 81th Meishan Road, Hefei 230022, China
- Correspondence: (H.D.); (C.L.); Tel.: +86-18856040979 (H.D.); +86-13505604595 (C.L.)
| |
Collapse
|
16
|
Hashemi M, Hajimazdarany S, Mohan CD, Mohammadi M, Rezaei S, Olyaee Y, Goldoost Y, Ghorbani A, Mirmazloomi SR, Gholinia N, Kakavand A, Salimimoghadam S, Ertas YN, Rangappa KS, Taheriazam A, Entezari M. Long non-coding RNA/epithelial-mesenchymal transition axis in human cancers: Tumorigenesis, chemoresistance, and radioresistance. Pharmacol Res 2022; 186:106535. [DOI: 10.1016/j.phrs.2022.106535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/07/2022]
|
17
|
miR-302a-3p Promotes Radiotherapy Sensitivity of Hepatocellular Carcinoma by Regulating Cell Cycle via MCL1. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1450098. [PMID: 36262872 PMCID: PMC9576429 DOI: 10.1155/2022/1450098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022]
Abstract
Background. The relationship between tumor suppressor gene miR-302a-3p and radiotherapy for hepatocellular carcinoma (HCC) remains unclear. This study intended to illustrate the molecular mechanism how miR-302a-3p regulated radiotherapy sensitivity of HCC. Methods. miR-302a-3p expression in HCC tissues and cells was examined by qRT-PCR. The effect of miR-302a-3p on HCC radiotherapy sensitivity were detected by CCK-8, colony formation, and flow cytometry assays. The expression levels of cell cycle-related proteins were detected by Western blot. The influence of miR-302a-3p on radiotherapy sensitivity of HCC was further investigated via cell cycle inhibitor (Caudatin) treatment. The target gene (MCL1) of miR-302a-3p was obtained by bioinformatics analysis, and their binding relationship was confirmed by RNA-binding protein immunoprecipitation assay. The mechanisms of miR-302a-3p regulating cell cycle and affecting radiotherapy sensitivity of HCC cells through MCL1 were further explored through the rescue experiments. Results. miR-302a-3p expression was remarkably reduced in radiotherapy-resistant tissues and cells of HCC. miR-302a-3p overexpression restored sensitivity of radiotherapy-resistant HCC cells to radiotherapy. Treatment with cell cycle inhibitor Caudatin could reverse suppressive effect of miR-302a-3p downregulation on sensitivity of HCC to radiotherapy. Additionally, miR-302a-3p could restrain MCL1 expression. In vitro cell assays further revealed that miR-302a-3p/MCL1 axis could enhance radiotherapy sensitivity of HCC cells by inducing G0/G1 arrest. Conclusions. miR-302a-3p facilitated radiotherapy sensitivity of HCC cells by regulating cell cycle via MCL1, which provided a new underlying target for radiotherapy resistance of HCC patients.
Collapse
|
18
|
Hu M, Ren M, Zhao Z, Cui X, Shi M, Yang Y, Guo H. Long non‑coding RNA SNHG3 promotes prostate cancer progression by sponging microRNA‑1827. Oncol Lett 2022; 24:281. [PMID: 35814830 PMCID: PMC9260729 DOI: 10.3892/ol.2022.13401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/13/2021] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are important biological factors that contribute to the initiation and progression of different types of cancer, including gastric, bladder and colorectal cancer. Small nucleolar RNA host gene 3 (SNHG3) has been implicated in prostate cancer (PCa) progression. However, the expression pattern and function of SNHG3 in PCa remain unclear, impeding the development of novel treatment strategies for this cancer. The present study aimed to investigate a combination of molecular and biochemical approaches to determine the role of SNHG3 in patients at different stages of disease, and elucidate the pathway by which SNHG3 affects PCa progression. A Cell Counting Kit-8 assay was used to assess cell proliferation. Transwell assays were used to analyze cell migration and invasion. Reverse transcription-quantitative PCR and western blotting were used to evaluate the expression levels of RNAs and proteins, respectively. The results demonstrated that SNHG3 expression was upregulated in PCa tissues downloaded from The Cancer Genome Atlas database, which was associated with poor prognosis. Furthermore, cell proliferation, migration and invasion were significantly inhibited following SNHG3 knockdown in vitro, the effects of which were reversed following overexpression of SNHG3 in PCa cells. Bioinformatic analysis revealed that microRNA (miRNA/miR)-1827 was a downstream target of SNHG3. The direct interaction between SNHG3 and miR-1827 was validated via the dual-luciferase reporter and RNA immunoprecipitation assays. Pearson's correlation analysis demonstrated that SNHG3 expression was negatively correlated with miR-1827 expression at different stages of PCa. Furthermore, rescue assays indicated that cotransfection with small interfering-SNHG3 and miR-1827 inhibitor reversed the effects of SNHG3 knockdown on cell proliferation, migration and invasion. In addition, SNHG3 knockdown in vivo suppressed tumor growth. Notably, lncRNA SNHG3 promoted PCa progression through miR-1827 via the Wnt/AKT/mTOR pathway. Taken together, the results of the present study suggest that SNHG3 promotes PCa progression by sponging miR-1827, indicating that SNHG3 may be a promising diagnostic and therapeutic target of PCa.
Collapse
Affiliation(s)
- Ming Hu
- Department of Urology, The Sixth Affiliated Hospital, South China University of Technology (People's Hospital of Nanhai District), Foshan, Guangdong 528200, P.R. China
| | - Mingliang Ren
- The Affiliated Nanhai Hospital, Southern Medical University (People's Hospital of Nanhai District), Foshan, Guangdong 528200, P.R. China
| | - Zhenhua Zhao
- Department of Urology, The Sixth Affiliated Hospital, South China University of Technology (People's Hospital of Nanhai District), Foshan, Guangdong 528200, P.R. China
| | - Xuejiang Cui
- Department of Urology, The Sixth Affiliated Hospital, South China University of Technology (People's Hospital of Nanhai District), Foshan, Guangdong 528200, P.R. China
| | - Ming Shi
- Department of Urology, The Sixth Affiliated Hospital, South China University of Technology (People's Hospital of Nanhai District), Foshan, Guangdong 528200, P.R. China
| | - Yunjie Yang
- Department of Urology, The Sixth Affiliated Hospital, South China University of Technology (People's Hospital of Nanhai District), Foshan, Guangdong 528200, P.R. China
| | - Haiyan Guo
- The Affiliated Nanhai Hospital, Southern Medical University (People's Hospital of Nanhai District), Foshan, Guangdong 528200, P.R. China
| |
Collapse
|
19
|
Salifu SP, Doughan A. New Clues to Prognostic Biomarkers of Four Hematological Malignancies. J Cancer 2022; 13:2490-2503. [PMID: 35711821 PMCID: PMC9174851 DOI: 10.7150/jca.69274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 03/06/2022] [Indexed: 11/24/2022] Open
Abstract
Globally, one out of every two reported cases of hematologic malignancies (HMs) results in death. Each year approximately 1.24 million cases of HMs are recorded, of which 58% become fatal. Early detection remains critical in the management and treatment of HMs. However, this is thwarted by the inadequate number of reliable biomarkers. In this study, we mined public databases for RNA-seq data on four common HMs intending to identify novel biomarkers that could serve as HM management and treatment targets. A standard RNA-seq analysis pipeline was strictly adhered to in identifying differentially expressed genes (DEGs) with DESeq2, limma+voom and edgeR. We further performed gene enrichment analysis, protein-protein interaction (PPI) network analysis, survival analysis and tumor immune infiltration level detection on the genes using G:Profiler, Cytoscape and STRING, GEPIA tool and TIMER, respectively. A total of 2,136 highly-ranked DEGs were identified in HM vs. non-HM samples. Gene ontology and pathway enrichment analyses revealed the DEGs to be mainly enriched in steroid biosynthesis (5.075×10-4), cholesterol biosynthesis (2.525×10-8), protein binding (3.308×10-18), catalytic activity (2.158×10-10) and biogenesis (5.929×10-8). The PPI network resulted in 60 hub genes which were verified with data from TCGA, MET500, CPTAC and GTEx projects. Survival analyses with clinical data from TCGA showed that high expression of SRSF1, SRSF6, UBE2Z and PCF11, and low expression of HECW2 were correlated with poor prognosis in HMs. In summary, our study unraveled essential genes that could serve as potential biomarkers for prognosis and may serve as drug targets for HM management.
Collapse
Affiliation(s)
- Samson Pandam Salifu
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Albert Doughan
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| |
Collapse
|
20
|
Hu Y, Zhao Z, Jin G, Guo J, Nan F, Hu X, Hu Y, Han Q. Long noncoding RNA regulatory factor X3- antisense RNA 1 promotes non-small cell lung cancer via the microRNA-577/signal transducer and activator of transcription 3 axis. Bioengineered 2022; 13:10749-10764. [PMID: 35475457 PMCID: PMC9208461 DOI: 10.1080/21655979.2022.2054910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the most frequent malignancy, and non-small cell lung cancer (NSCLC) is its most common pathological type. Molecular targeted therapy has been testified to be effective in intervening in the occurrence and development of malignancies. This study investigates the effect of lncRNA Regulatory Factor X3- antisense RNA 1 (RFX3-AS1) in NSCLC progression. The RFX3-AS1 profile in NSCLC tissues and cells was measured by quantitative reverse transcription PCR (qRT-PCR). The RFX3-AS1 overexpression model was constructed. The cell counting kit-8 (CCK-8) experiment and cell colony formation assay were adopted to test cell viability. The cell apoptosis was determined by flow cytometry (FCM). Cell migration and invasion were monitored by the Transwell assay, and Western blot was implemented to verify the protein profiles of signal transducer and activator of transcription 3 (STAT3), E-cadherin, Vimentin and N-cadherin. In vivo, we validated the impact of RFX3-AS1 overexpression on the NSCLC xenograft mouse model. The targeting relationships between RFX3-AS1 and miR-577, miR-577 and STAT3 were confirmed by the dual-luciferase reporter assay. The results manifested that overexpressing RFX3-AS1 markedly facilitated NSCLC cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT), and suppressed cell apoptosis. In contrast, miR-577, which was a downstream target of RFX3-AS1, dramatically impeded the malignant biological behaviors of NSCLC cells. STAT3 was a direct target of miR-577, and it was negatively regulated by the latter. STAT3 activation reversed miR-577-mediated anti-tumor roles. In brief, RFX3-AS1 aggravated NSCLC progression by regulating the miR-577/STAT3 axis.
Collapse
Affiliation(s)
- Yanjing Hu
- Department of Thoracic Surgery, The First People's Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Zhi Zhao
- Department of Thoracic Surgery, The First People's Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Gang Jin
- Department of Thoracic Surgery, The First People's Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Junhao Guo
- Department of Thoracic Surgery, The First People's Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Fangyuan Nan
- Department of Thoracic Surgery, The First People's Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Xin Hu
- Department of Thoracic Surgery, The First People's Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Yunsheng Hu
- Department of Thoracic Surgery, The First People's Hospital of Jiangxia District, Wuhan, Hubei, China
| | - Qun Han
- Department of Thoracic Surgery, The First People's Hospital of Jiangxia District, Wuhan, Hubei, China
| |
Collapse
|
21
|
Shan DD, Zheng QX, Wang J, Chen Z. Small nucleolar RNA host gene 3 functions as a novel biomarker in liver cancer and other tumour progression. World J Gastroenterol 2022; 28:1641-1655. [PMID: 35581965 PMCID: PMC9048787 DOI: 10.3748/wjg.v28.i16.1641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/09/2022] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer has become the most life-threatening disease in the world. Mutations in and aberrant expression of genes encoding proteins and mutations in noncoding RNAs, especially long noncoding RNAs (lncRNAs), have significant effects in human cancers. LncRNAs have no protein-coding ability but function extensively in numerous physiological and pathological processes. Small nucleolar RNA host gene 3 (SNHG3) is a novel lncRNA and has been reported to be differentially expressed in various tumors, such as liver cancer, gastric cancer, and glioma. However, the interaction mechanisms for the regulation between SNHG3 and tumor progression are poorly understood. In this review, we summarize the results of SNHG3 studies in humans, animal models, and cells to underline the expression and role of SNHG3 in cancer. SNHG3 expression is upregulated in most tumors and is detrimental to patient prognosis. SNHG3 expression in lung adenocarcinoma remains controversial. Concurrently, SNHG3 affects oncogenes and tumor suppressor genes through various mechanisms, including competing endogenous RNA effects. A deeper understanding of the contribution of SNHG3 in clinical applications and tumor development may provide a new target for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Dan-Dan Shan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Qiu-Xian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Jing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
22
|
Wang X, Song Y, Shi Y, Yang D, Li J, Yin B. SNHG3 could promote prostate cancer progression through reducing methionine dependence of PCa cells. Cell Mol Biol Lett 2022; 27:13. [PMID: 35123415 PMCID: PMC8903624 DOI: 10.1186/s11658-022-00313-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 01/17/2022] [Indexed: 01/21/2023] Open
Abstract
In recent years, morbidity and mortality of prostate cancer (PCa) have increased dramatically, while mechanistic understanding of its onset and progression remains unmet. LncRNA SNHG3 has been proved to stimulate malignant progression of multiple cancers, whereas its functional mechanism in PCa needs to be deciphered. In this study, our analysis in the TCGA database revealed high SNHG3 expression in PCa tissue. Further analysis in starBase, TargetScan, and mirDIP databases identified the SNHG3/miR-152-3p/SLC7A11 regulatory axis. FISH was conducted to assess the distribution of SNHG3 in PCa tissue. Dual-luciferase reporter gene and RIP assays confirmed the relationship among the three objects. Next, qRT-PCR and western blot were conducted to measure expression levels of SNHG3, miR-152-3p, and SLC7A11. CCK-8, colony formation, Transwell, and flow cytometry were carried out to assess proliferation, migration, invasion, methionine dependence, apoptosis, and the cell cycle. It was noted that SNHG3 as a molecular sponge of miR-152-3p stimulated proliferation, migration, and invasion, restrained methionine dependence and apoptosis, and affected the cell cycle of PCa cells via targeting SLC7A11. Additionally, we constructed xenograft tumor models in nude mice and confirmed that knockdown of SNHG3 could restrain PCa tumor growth and elevate methionine dependence in vivo. In conclusion, our investigation improved understanding of the molecular mechanism of SNHG3 modulating PCa progression, thereby generating novel insights into clinical therapy for PCa.
Collapse
|
23
|
Crosstalk between Long Non Coding RNAs, microRNAs and DNA Damage Repair in Prostate Cancer: New Therapeutic Opportunities? Cancers (Basel) 2022; 14:cancers14030755. [PMID: 35159022 PMCID: PMC8834032 DOI: 10.3390/cancers14030755] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Non-coding RNAs are a type of genetic material that doesn’t make protein, but performs diverse regulatory functions. In prostate cancer, most treatments target proteins, and resistance to such therapies is common, leading to disease progression. Targeting non-coding RNAs may provide alterative treatment options and potentially overcome drug resistance. Major types of non-coding RNAs include tiny ‘microRNAs’ and much longer ‘long non-coding RNAs’. Scientific studies have shown that these form a major part of the human genome, and play key roles in altering gene activity and determining the fate of cells. Importantly, in cancer, their activity is altered. Recent evidence suggests that microRNAs and long non-coding RNAs play important roles in controlling response to DNA damage. In this review, we explore how different types of non-coding RNA interact to control cell DNA damage responses, and how this knowledge may be used to design better prostate cancer treatments and tests. Abstract It is increasingly appreciated that transcripts derived from non-coding parts of the human genome, such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), are key regulators of biological processes both in normal physiology and disease. Their dysregulation during tumourigenesis has attracted significant interest in their exploitation as novel cancer therapeutics. Prostate cancer (PCa), as one of the most diagnosed malignancies and a leading cause of cancer-related death in men, continues to pose a major public health problem. In particular, survival of men with metastatic disease is very poor. Defects in DNA damage response (DDR) pathways culminate in genomic instability in PCa, which is associated with aggressive disease and poor patient outcome. Treatment options for metastatic PCa remain limited. Thus, researchers are increasingly targeting ncRNAs and DDR pathways to develop new biomarkers and therapeutics for PCa. Increasing evidence points to a widespread and biologically-relevant regulatory network of interactions between lncRNAs and miRNAs, with implications for major biological and pathological processes. This review summarises the current state of knowledge surrounding the roles of the lncRNA:miRNA interactions in PCa DDR, and their emerging potential as predictive and diagnostic biomarkers. We also discuss their therapeutic promise for the clinical management of PCa.
Collapse
|
24
|
Zhu H, Zhu C, Feng X, Luo Y. Long noncoding RNA SNHG3 promotes malignant phenotypes in cervical cancer cells via association with YAP1. Hum Cell 2021; 35:320-332. [PMID: 34816392 DOI: 10.1007/s13577-021-00644-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/03/2021] [Indexed: 12/19/2022]
Abstract
Long non-coding RNA (LncRNA) Small Nucleolar RNA Host Gene 3 (SNHG3) is involved in the occurrence and development of various cancers. However, the exact function and mechanism of SNHG3 in cervical cancer (CC) are still unclear. In this context, we identified a significant increase of SNHG3 expression in CC tissues. Upregulation of SNHG3 expression was associated with advanced FIGO stage and metastasis, and indicated poor overall survival of the CC patients. Functionally, SNHG3 enhanced the proliferation, migration and invasion of CC cells in vitro, and facilitated CC growth in vivo. Further investigation uncovered that SNHG3 interacted with oncoprotein YAP1, thus suppressing its degradation. Additionally, SNHG3 modulated the transcription of several target genes of YAP1. The oncogenic role of SNHG3 was partially attributable to YAP1. Taken together, our research revealed the prognostic and functional roles for SNHG3 in CC, suggesting that SNHG3 could serve as a biomarker for prognosis and a therapeutic target for CC.
Collapse
Affiliation(s)
- Hongyu Zhu
- Gynecology Second Ward, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, 443003, Hubei, China.
| | - Chenyu Zhu
- Gastrointestinal Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, 443003, Hubei, China
| | - Xiang Feng
- Obstetrics Department, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, 443003, Hubei, China
| | - Youzhen Luo
- Gynecology Second Ward, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, 443003, Hubei, China.
| |
Collapse
|
25
|
Peng N, Zhang Z, Wang Y, Yang M, Fan J, Wang Q, Deng L, Chen D, Cai Y, Li Q, Wang X, Li W. Down-regulated LINC00115 inhibits prostate cancer cell proliferation and invasion via targeting miR-212-5p/FZD5/Wnt/β-catenin axis. J Cell Mol Med 2021; 25:10627-10637. [PMID: 34697900 PMCID: PMC8581327 DOI: 10.1111/jcmm.17000] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 01/21/2023] Open
Abstract
Prostate cancer is the second most frequent malignancy in men worldwide, and its incidence is increasing. Therefore, it is urgently required to clarify the underlying mechanisms of prostate cancer. Although the long non‐coding RNA LINC00115 was identified as an oncogene in several cancers, the expression and function of LINC00115 in prostate cancer have not been explored. Our results showed that LINC00115 was significantly up‐regulated in prostate cancer tissues, which was significantly associated with a poor prognosis for prostate cancer patients. Functional studies showed that knockdown LINC00115 inhibited cell proliferation and invasion. In addition, LINC00115 served as a competing endogenous RNA (ceRNA) through sponging miR‐212‐5p to release Frizzled Family Receptor 5 (FZD5) expression. The expression of miR‐212‐5p was noticeably low in tumour tissues, and FZD5 expression level was down‐regulated with the knockdown of LINC00115. Knockdown LINC00115 inhibited the Wnt/β‑catenin signalling pathway by inhibiting the expression of FZD5. Rescue experiments further showed that LINC00115 inhibits prostate cancer cell proliferation and invasion via targeting miR‐212‐5p/ FZD5/ Wnt/β‐catenin axis. The present study provided clues that LINC00115 may be a promising novel therapeutic target for prostate cancer patients.
Collapse
Affiliation(s)
- Naixiong Peng
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Zejian Zhang
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Yaomin Wang
- Wake Forest University, Winston-Salem, NC, USA
| | - Minlong Yang
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Jiqing Fan
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Qinjun Wang
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Ling Deng
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Dong Chen
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Yuefeng Cai
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Qihui Li
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Xisheng Wang
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| | - Wei Li
- Department of Urology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, China
| |
Collapse
|
26
|
Sun Z, Hu J, Hu K, Tang M, Sun S, Fang Y, Yu H, Zhang Y. [Role of long noncoding RNA SNHG3 in regulating proliferation, migration and invasion of cervical cancer SiHa cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:931-936. [PMID: 34238747 DOI: 10.12122/j.issn.1673-4254.2021.06.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the regulatory role of the long non-coding RNA (lncRNA) small nucleolar host gene 3 (SNHG3) in proliferation, migration and invasion of human cervical cancer cell line SiHa. OBJECTIVE Array data were retrieved from GEO database to analyze the expression levels of SNHG3 in cervical cancer and adjacent normal tissues. SiHa cells were transfected with a small interfering RNA (siRNA) targeting SNHG3, and the changes in the transcriptional levels of lncRNA SNHG3 and the epithelial-mesenchymal transition (EMT) markers N-cadherin, Snail, vimentin and E-cadherin were detected using real-time quantitative PCR; the protein expressions of N-cadherin, Snail, vimentin and E-cadherin were determined using Western blotting. Cell counting kit-8 (CCK8) assay was utilized to assess the proliferation capacity of the transfected cells. Wound healing assay and Transwell assay were performed to evaluate the transversal and longitudinal migration and invasion abilities of the cells. OBJECTIVE SNHG3 was over-expressed in cervical cancer tissues and SiHa cells. In SiHa cells, knocking down SNHG3 significantly inhibited the proliferation (P < 0.001), migration (P < 0.01) and invasion abilities (P < 0.001) of the cells, down-regulated the expression levels of N-cadherin, Snail and vimentin (P < 0.001) and up-regulated the expression of E-cadherin (P < 0.001). OBJECTIVE SNHG3 may promote the proliferation, migration and invasion of SiHa cells by activating the EMT signaling pathway.
Collapse
Affiliation(s)
- Z Sun
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - J Hu
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - K Hu
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - M Tang
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - S Sun
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Y Fang
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - H Yu
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Y Zhang
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
27
|
Guo X, Zheng J, Yu MJ, Piao HZ, Zhao HY. Long noncoding RNA SNHG3 promotes glioma tumorigenesis by sponging miR-485-5p to upregulate LMX1B expression. Kaohsiung J Med Sci 2021; 37:851-862. [PMID: 34153159 DOI: 10.1002/kjm2.12411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/07/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
LIM homeobox transcription factor 1-beta (LMX1B) has recently been found to be highly expressed in advanced gliomas and is associated with poor survival. However, the regulatory molecular mechanism of LMX1B expression in gliomas remains unclear. In this study, bioinformatics analysis showed that miR-485-5p may be the potential upstream regulator of LMX1B, and long noncoding RNA (lncRNA) small nucleolar RNA host gene 3 (SNHG3) may function as a competitive endogenous RNA to sponge miR-485-5p. In addition, the expression of SNHG3 and LMX1B in advanced glioma tissues was significantly upregulated, while the expression of miR-485-5p was significantly downregulated. SNHG3 overexpression reduced the expression of miR-485-5p; increased the expression of LMX1B; and promoted the proliferation, migration, and invasion of glioma cells. In contrast, miR-485-5p overexpression reduced the expression of LMX1B and inhibited cell proliferation, migration, and invasion. The luciferase reporter assay and RNA immunoprecipitation assay further confirmed the interaction between SNHG3 and miR-485-5p and between miR-485-5p and LMX1B. In addition, subcutaneous and orthotropic xenograft models confirmed that lncRNA SNHG3 silencing or miR-485-5p overexpression significantly reduced the growth of glioma xenografts and prolonged survival time. These results indicate that lncRNA SNHG3 can regulate the expression of LMX1B by sponging miR-485-5p, thereby promoting the proliferation, migration, and invasion of glioma cells. This study provides the first evidence that the SNHG3/miR-485-5p/LMX1B axis is involved in glioma tumorigenesis and highlights the potential of SNHG3 and miR-485-5p as therapeutic targets for glioma.
Collapse
Affiliation(s)
- Xu Guo
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming-Jun Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hao-Zhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Hong-Yu Zhao
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
28
|
Wang Z, Chen Y, Wang W, Wang H, Liu R. circMYC promotes cell proliferation, metastasis, and glycolysis in cervical cancer by up-regulating MET and sponging miR-577. Am J Transl Res 2021; 13:6043-6054. [PMID: 34306343 PMCID: PMC8290748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/23/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To analyze the role of circMYC in cervical cancer. METHODS Protein and RNA expression was detected by RT-qPCR and western blotting. Transwell, CCK8, and colony formation assays were used for measuring metastasis, cell viability, and proliferation, respectively. Lactate production, glucose uptake, and ATP generation were examined to evaluate cell glycolysis. Interactions between circMYC, miR-577, and MET were determined by RNA pull-down and immunoprecipitation, and dual-luciferase reporter assays. Xenografts were established in mice to evaluate the functions of circMYC in vivo. RESULTS circMYC was overexpressed in tumor tissue, which was related to poor prognosis. CircMYC knockdown reduced proliferation, colony formation, metastasis, and glycolysis in cervical cancer cells as well as inhibiting tumor growth in vivo. Mechanistically, circMYC targeted miR-577, and the effects of circMYC knockdown could be reversed by miR-577 inhibition. Moreover, miR-577 downregulated the expression of MET. Therefore, the oncogenic role of circMYC in cervical cancer was achieved by sponging miR-577 and maintaining MET expression. CONCLUSION circMYC promotes cervical cancer progression through regulation of the miR-577/MET axis. circMYC may thus be a potential target for diagnosing and treating cervical cancer.
Collapse
Affiliation(s)
- Zhizhen Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin, China
- Key Laboratory of Cancer Prevention and TherapyTianjin, China
- Tianjin’s Clinical Research Center for CancerTianjin, China
| | - Yang Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin, China
- Key Laboratory of Cancer Prevention and TherapyTianjin, China
- Tianjin’s Clinical Research Center for CancerTianjin, China
| | - Wei Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin, China
- Key Laboratory of Cancer Prevention and TherapyTianjin, China
- Tianjin’s Clinical Research Center for CancerTianjin, China
| | - Hui Wang
- Department of Radiology, Tianjin Third Central HospitalTianjin, China
| | - Ransheng Liu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin, China
- Key Laboratory of Cancer Prevention and TherapyTianjin, China
- Tianjin’s Clinical Research Center for CancerTianjin, China
| |
Collapse
|
29
|
Biagioni A, Tavakol S, Ahmadirad N, Zahmatkeshan M, Magnelli L, Mandegary A, Samareh Fekri H, Asadi MH, Mohammadinejad R, Ahn KS. Small nucleolar RNA host genes promoting epithelial-mesenchymal transition lead cancer progression and metastasis. IUBMB Life 2021; 73:825-842. [PMID: 33938625 DOI: 10.1002/iub.2501] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/22/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023]
Abstract
The small nucleolar RNA host genes (SNHGs) belong to the long non-coding RNAs and are reported to be able to influence all three levels of cellular information-bearing molecules, that is, DNA, RNA, and proteins, resulting in the generation of complex phenomena. As the host genes of the small nucleolar RNAs (snoRNAs), they are commonly localized in the nucleolus, where they exert multiple regulatory functions orchestrating cellular homeostasis and differentiation as well as metastasis and chemoresistance. Indeed, worldwide literature has reported their involvement in the epithelial-mesenchymal transition (EMT) of different histotypes of cancer, being able to exploit peculiar features, for example, the possibility to act both in the nucleus and the cytoplasm. Moreover, SNHGs regulation is a fundamental topic to better understand their role in tumor progression albeit such mechanism is still debated. Here, we reviewed the biological functions of SNHGs in particular in the EMT process and discussed the perspectives for new cancer therapies.
Collapse
Affiliation(s)
- Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Florence, Italy
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nooshin Ahmadirad
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Zahmatkeshan
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, Florence, Italy
| | - Ali Mandegary
- Department of Pharmacology & Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Hojjat Samareh Fekri
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.,Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Malek Hossein Asadi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Reza Mohammadinejad
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
30
|
Li J, Du H, Chen W, Qiu M, He P, Ma Z. Identification of potential autophagy-associated lncRNA in prostate cancer. Aging (Albany NY) 2021; 13:13153-13165. [PMID: 33971627 PMCID: PMC8148478 DOI: 10.18632/aging.202997] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
Background: Long non-coding RNAs (lncRNAs) have been linked to autophagy. It is urgent to identify and assess the hub autophagy-associated lncRNA in prostate cancer. Methods: Differentially expressed lncRNAs associated with autophagy were identified in prostate cancer based on The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA-PRAD) data. An autophagy-mediated competing endogenous RNA network was constructed to screen for autophagy-associated lncRNA, and the preselected lncRNAs were further validated using Gene Expression Omnibus (GEO) datasets. Furthermore, a prognostic lncRNA signature was established and assessed. Additionally, Gene Set Enrichment Analysis (GSEA) revealed the underlying molecular mechanisms. Results: Using a competing endogenous RNA network, 66 differentially expressed lncRNAs associated with autophagy were identified, and the differential expression of 7 lncRNAs were verified using the TCGA-PRAD, GSE21034, and GSE94767 datasets. Additionally, a lncRNA signature associated with autophagy, including MKNK1-AS1 and INE1, was identified as an independent indicator of survival with a C-index of 0.882. The GSEA analysis indicated that several autophagy-related signaling pathways were enriched in different risk groups. Conclusions: The lncRNAs associated with autophagy were identified, and a prediction model was developed that could be used as a prognostic predictor for prostate cancer, indicating the critical role of lncRNA in the regulation of prostate cancer autophagy regulation.
Collapse
Affiliation(s)
- Jun Li
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Du
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenqiang Chen
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingxing Qiu
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng He
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhiwei Ma
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
31
|
Altschuler J, Stockert JA, Kyprianou N. Non-Coding RNAs Set a New Phenotypic Frontier in Prostate Cancer Metastasis and Resistance. Int J Mol Sci 2021; 22:ijms22042100. [PMID: 33672595 PMCID: PMC7924036 DOI: 10.3390/ijms22042100] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) mortality remains a significant public health problem, as advanced disease has poor survivability due to the development of resistance in response to both standard and novel therapeutic interventions. Therapeutic resistance is a multifaceted problem involving the interplay of a number of biological mechanisms including genetic, signaling, and phenotypic alterations, compounded by the contributions of a tumor microenvironment that supports tumor growth, invasiveness, and metastasis. The androgen receptor (AR) is a primary regulator of prostate cell growth, response and maintenance, and the target of most standard PCa therapies designed to inhibit AR from interacting with androgens, its native ligands. As such, AR remains the main driver of therapeutic response in patients with metastatic castration-resistant prostate cancer (mCRPC). While androgen deprivation therapy (ADT), in combination with microtubule-targeting taxane chemotherapy, offers survival benefits in patients with mCRPC, therapeutic resistance invariably develops, leading to lethal disease. Understanding the mechanisms underlying resistance is critical to improving therapeutic outcomes and also to the development of biomarker signatures of predictive value. The interconversions between epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET) navigate the prostate tumor therapeutic response, and provide a novel targeting platform in overcoming therapeutic resistance. Both microRNA (miRNA)- and long non-coding RNA (lncRNA)-mediated mechanisms have been associated with epigenetic changes in prostate cancer. This review discusses the current evidence-based knowledge of the role of the phenotypic transitions and novel molecular determinants (non-coding RNAs) as contributors to the emergence of therapeutic resistance and metastasis and their integrated predictive value in prostate cancer progression to advanced disease.
Collapse
Affiliation(s)
- Joshua Altschuler
- Department of Urology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.A.); (J.A.S.)
| | - Jennifer A. Stockert
- Department of Urology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.A.); (J.A.S.)
| | - Natasha Kyprianou
- Department of Urology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.A.); (J.A.S.)
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence:
| |
Collapse
|
32
|
Adylova A, Mukhanbetzhanovna AA, Attar R, Yulaevna IM, Farooqi AA. Regulation of TGFβ/SMAD signaling by long non-coding RNAs in different cancers: Dark Knight in the Castle of molecular oncology. Noncoding RNA Res 2021; 6:23-28. [PMID: 33511320 PMCID: PMC7814108 DOI: 10.1016/j.ncrna.2020.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 11/15/2022] Open
Abstract
One of the complex themes in recent years has been the multi-layered regulation of TGFβ signaling in cancer cells. TGFβ/SMAD signaling pathway is a highly complicated web of proteins which work spatio-temporally to regulate multiple steps of carcinogenesis. TGFβ/SMAD has been shown to dualistically regulate cancer progression. Therefore, TGFβ/SMAD signaling behaves as a “double-edged sword” in molecular oncology. Accordingly, regulation of TGFβ/SMAD is multi-layered because of oncogenic and tumor suppressor long non-coding RNAs (LncRNAs). In this review, we have summarized most recent breakthroughs in our understanding related to regulation of TGFβ/SMAD signaling by lncRNAs. We have comprehensively analyzed how different lncRNAs positively and negatively regulate TGFβ/SMAD signaling in different cancers. We have gathered missing pieces of an incomplete jig-saw puzzle of lncRNA-interactome ranging from “sponge effects” of lncRNAs to mechanistic modulation of TGFβ/SMAD signaling by lncRNAs.
Collapse
Affiliation(s)
- Aima Adylova
- Biomedical Engineering & Molecular Medicine PhD candidate, Guangdong Key Laboratory for Genome Stability & Disease Prevention and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen, Guangdong, 518060, China
| | | | - Rukset Attar
- Department of Obstetrics and Gynecology, Yeditepe University, Turkey
| | | | | |
Collapse
|
33
|
Bai L, Yao N, Qiao G, Wu L, Ma X. CXCL5 contributes to the tumorigenicity of cervical cancer and is post-transcriptionally regulated by miR-577. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2984-2993. [PMID: 33425099 PMCID: PMC7791384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/27/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND C-X-C motif chemokine ligand 5 (CXCL5), an important chemokine, has been validated to promote human tumorigenesis. However, the clinical significance and the underlying molecular mechanisms of CXCL5 have not been completely explored in cervical cancer. Herein, the aim was to investigate miR-577-mediated CXCL5 signaling in cervical tumorigenicity. MATERIAL AND METHODS Sixty-one pairs of cervical cancer specimens and para-carcinoma tissues were collected to measure miR-577 and CXCL5 expression levels. miR-577 mimics and/or si-CXCL5 were transfected into cervical cancer cell lines, Hela, and SiHa cells, to determine their effect on cell proliferation, migration and apoptosis. RESULTS Our results demonstrated that CXCL5 is overexpressed in cervical cancer tissues and cell lines. Knockdown of CXCL5 with specific siRNA transfection in Hela and SiHa cells significantly inhibited cell proliferation and migration and induced apoptosis in vitro. We also report that CXCL5 is a direct target of miR-577. Additionally, transfection of miR-577 mimics can inhibit CXCL5 protein expression, but not mRNA in Hela cells. miR-577 mimic transfection significantly inhibits migration and induces apoptosis in Hela and SiHa cells. However, the antineoplastic activities of miR-577 are reversed by overexpression of CXCL5 in vitro. CONCLUSIONS Overexpression of CXCL5 is involved in tumor development of cervical cancer. Inhibition of CXCL5 by its post-transcriptional regulator, miR-577, may provide a promising therapeutic strategy for patients with cervical cancer.
Collapse
Affiliation(s)
- Lu Bai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of The Air Force Medical University of PLA Xi'an 710032, Shaanxi Province, China
| | - Nianling Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of The Air Force Medical University of PLA Xi'an 710032, Shaanxi Province, China
| | - Guyuan Qiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of The Air Force Medical University of PLA Xi'an 710032, Shaanxi Province, China
| | - Liying Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of The Air Force Medical University of PLA Xi'an 710032, Shaanxi Province, China
| | - Xiangdong Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of The Air Force Medical University of PLA Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
34
|
Xia Q, Li Y, Han D, Dong L. SMURF1, a promoter of tumor cell progression? Cancer Gene Ther 2020; 28:551-565. [PMID: 33204002 DOI: 10.1038/s41417-020-00255-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/14/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022]
Abstract
Overexpression of HECT-type E3 ubiquitin ligase SMURF1 is correlated with poor prognosis in patients with various cancers, such as glioblastoma, colon cancer, and clear cell renal cell carcinoma. SMURF1 acts as a tumor promoter by ubiquitination modification and/or degradation of tumor-suppressing proteins. Combined treatment of Smurf1 knockdown with rapamycin showed collaborative antitumor effects in mice. This review described the role of HECT, WW, and C2 domains in regulating SMURF1 substrate selection. We summarized up to date SMURF1 substrates regulating different type cell signaling, thus, accelerating tumor progression, invasion, and metastasis. Furthermore, the downregulation of SMURF1 expression, inhibition of its E3 activity and regulation of its specificity to substrates prevent tumor progression. The potential application of SMURF1 regulators, specifically, wisely choose certain drugs by blocking SMURF1 selectivity in tumor suppressors, to develop novel anticancer treatments.
Collapse
Affiliation(s)
- Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yang Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Da Han
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
35
|
Zhao S, Gao X, Zhong C, Li Y, Wang M, Zang S. SNHG3 Knockdown Suppresses Proliferation, Migration and Invasion, and Promotes Apoptosis in Non-Small Cell Lung Cancer Through Regulating miR-216a/ZEB1 Axis. Onco Targets Ther 2020; 13:11327-11336. [PMID: 33177840 PMCID: PMC7649239 DOI: 10.2147/ott.s263637] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/24/2020] [Indexed: 01/14/2023] Open
Abstract
Background Tumour growth and development are dependent on many factors including long noncoding RNAs (lncRNAs). However, limited information is available on the involvement of lncRNAs in non-small cell lung cancer (NSCLC) and the molecular mechanisms have not been defined. Here, we examined the expression of small nucleolar RNA host gene 3 (SNHG3) and its contribution to the development of NSCLC. Methods We detected SNHG3, miR-216a, and ZEB1 expression in tissues from NSCLC patients and lung adenocarcinoma cell lines using quantitative real-time polymerase chain reaction. Proliferation, migrations, invasion, and apoptosis of tumour cells were assessed using cell counting kit-8, transwell experiments, and flow cytometry after SNHG3 knockdown by small interfering RNAs. Bioinformatics and luciferase reporter assays were employed for analysing the interactions between SNHG3, miR-216a, and ZEB1. Results We found highly upregulated SNHG3 in tissues and cells from NSCLC patients, which was linked to poor prognosis. SNHG3 silencing diminished the ability of NSCLC cells to proliferate, migrate, and invade and promoted apoptosis. Furthermore, SNHG3 competed with endogenous RNA and enhanced the expression of ZEB1 by interfering with miR-216a. ZEB1 overexpression or miR-216a blockade reversed SNHG3-induced tumour inhibition. Similar effects were observed in vivo where SNHG3 knockdown inhibited NSCLC tumour growth by reducing expression of miR-216a while increasing that of ZEB1. Conclusion Knockdown of SNHG3 inhibits NSCLC tumour development and progression by upregulation of ZEB1 and interference with miR-216a, revealing an attractive alternative target for patients with NSCLC.
Collapse
Affiliation(s)
- Shasha Zhao
- Department of Respiratory Medicine, The First Hospital Affiliated to the Xinxiang Medical College, Weihui, Henan 453100, People's Republic of China
| | - Xinyuan Gao
- Department of Respiratory Medicine, The First Hospital Affiliated to the Xinxiang Medical College, Weihui, Henan 453100, People's Republic of China
| | - Chunlei Zhong
- Department of Respiratory Medicine, The First Hospital Affiliated to the Xinxiang Medical College, Weihui, Henan 453100, People's Republic of China
| | - Yunxia Li
- Department of Respiratory Medicine, The First Hospital Affiliated to the Xinxiang Medical College, Weihui, Henan 453100, People's Republic of China
| | - Ming Wang
- Department of Respiratory Medicine, The First Hospital Affiliated to the Xinxiang Medical College, Weihui, Henan 453100, People's Republic of China
| | - Shuzhi Zang
- Department of Respiratory Medicine, The First Hospital Affiliated to the Xinxiang Medical College, Weihui, Henan 453100, People's Republic of China
| |
Collapse
|
36
|
Xu B, Mei J, Ji W, Bian Z, Jiao J, Sun J, Shao J. LncRNA SNHG3, a potential oncogene in human cancers. Cancer Cell Int 2020; 20:536. [PMID: 33292213 PMCID: PMC7640707 DOI: 10.1186/s12935-020-01608-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are composed of > 200 nucleotides; they lack the ability to encode proteins but play important roles in a variety of human tumors. A large number of studies have shown that dysregulated expression of lncRNAs is related to tumor oncogenesis and progression. Emerging evidence shows that SNHG3 is a novel oncogenic lncRNA that is abnormally expressed in various tumors, including osteosarcoma, liver cancer, lung cancer, etc. SNHG3 primarily competes as a competitive endogenous RNA (ceRNA) that targets tumor suppressor microRNAs (miRNAs) and ceRNA mechanisms that regulate biological processes of tumors. In addition, abnormal expression of SNHG3 is significantly correlated with patient clinical features. Upregulation of SNHG3 contributes to biological functions, including tumor cell proliferation, migration, invasion and EMT. Therefore, SNHG3 may represent a potential diagnostic and prognostic biomarker, as well as a novel therapeutic target.
Collapse
Affiliation(s)
- Bin Xu
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China
| | - Jie Mei
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, P. R. China
| | - Wei Ji
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China
| | - Zheng Bian
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China
| | - Jiantong Jiao
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China
| | - Jun Sun
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China.
| | - Junfei Shao
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
37
|
LINC01094/miR-577 axis regulates the progression of ovarian cancer. J Ovarian Res 2020; 13:122. [PMID: 33069244 PMCID: PMC7568364 DOI: 10.1186/s13048-020-00721-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background Long intergenic non-coding RNA 01094 (LINC01094) is probably a novel regulator in cancer biology. This study aimed to probe into the function and mechanism of LINC01094 in ovarian cancer (OC). Methods Quantitative real-time polymerase chain reaction (qRT-PCR) assay was utilized to measure LINC01094 and miR-577 expressions in OC tissues and cell lines. Western blot was used to examine the expressions of epithelial-mesenchymal transition (EMT)-related proteins, β-catenin, c-Myc and cyclin D1. Cell counting kit-8 (CCK-8) and Transwell assays were used to detect the proliferation, migration and invasion of SKOV3 and 3AO cells, respectively. Eventually, dual-luciferase reporter gene assay was employed to detect the regulatory relationship between miR-577 and LINC01094. Results LINC01094 expression was elevated in OC tissues and cell lines. High LINC01094 expression was associated with higher FIGO stage, lymph node metastasis and the shorter overall survival rate in patients with OC. Meanwhile, LINC01094 knockdown inhibited OC cell proliferation, migration, invasion and EMT. In addition, miR-577 was demonstrated to be a direct downstream target of LINC01094 in OC and inhibition of miR-577 reversed the biological effects of LINC01094 knockdown on OC cells. Additionally, LINC01094 / miR-577 axis regulated the expressions of β-catenin, c-Myc and cyclin D1 in OC cells. Conclusion LINC01094 promotes the proliferation, migration, invasion and EMT of OC cells by adsorbing miR-577.
Collapse
|
38
|
He S, Wang X, Zhang J, Zhou F, Li L, Han X. TRG-AS1 is a potent driver of oncogenicity of tongue squamous cell carcinoma through microRNA-543/Yes-associated protein 1 axis regulation. Cell Cycle 2020; 19:1969-1982. [PMID: 32615889 PMCID: PMC7469544 DOI: 10.1080/15384101.2020.1786622] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The long noncoding RNA T cell receptor gamma locus antisense RNA 1 (TRG-AS1) plays an important role in glioblastoma progression. The objective of this study was to determine the expression status of TRG-AS1 in tongue squamous cell carcinoma (TSCC). The regulatory effects of TRG-AS1 depletion on the malignant processes of TSCC cells were illustrated both in vitro and in vivo. Additionally, the precise molecular mechanisms through which TRG-AS promotes TSCC oncogenicity were investigated. TRG-AS1 expression in TSCC tissues and cell lines was detected using reverse transcription-quantitative PCR. Functional experiments including Cell Counting Kit-8 assay, flow cytometric apoptotic assay, migration and invasion assays, and xenograft tumor model analysis were conducted to severally determine the effects of TRG-AS1 on TSCC cell proliferation, apoptosis, migration, and invasion in vitro and tumor growth in vivo. Herein, TRG-AS1 was highly expressed in TSCC and closely associated with advanced TNM stage, high lymph node metastasis, and poor overall survival. Functionally, TRG-AS1 depletion suppressed TSCC cell proliferation, migration, and invasion in vitro; promoted cell apoptosis; and attenuated tumor growth in vivo. Mechanistically, TRG-AS1 served as a molecular sponge for microRNA-543 (miR-543), thereby contributing to the increased expression of Yes-associated protein 1 (YAP1) - a miR-543 target. Rescue experiments confirmed that miR-543 inhibition or YAP1 overexpression abrogated the anticancer effects of TRG-AS1 silencing in TSCC cells. In conclusion, TRG-AS1 aggravates TSCC malignancy by regulating the miR-543/YAP1 axis. Identification of the TRG-AS1/miR-543/YAP1 regulatory pathway may provide novel insights into TSCC diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Shuwei He
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Jingjing Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Fan Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Lei Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xingmin Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China,CONTACT Xingmin Han
| |
Collapse
|
39
|
Li T, Xing Y, Yang F, Sun Y, Zhang S, Wang Q, Zhang W. LncRNA SNHG3 sponges miR-577 to up-regulate SMURF1 expression in prostate cancer. Cancer Med 2020; 9:3852-3862. [PMID: 32248648 PMCID: PMC7286463 DOI: 10.1002/cam4.2992] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/25/2020] [Accepted: 03/02/2020] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer remains one of the most prevalent cancers and the main causes of cancer-related deaths in males. Various articles introduced that long noncoding RNAs (lncRNAs) are found in vital functions in the development and progression of cancers. Although SNHG3 (small nucleolar RNA host gene 3) has been investigated in many cancers, now researches on the role and mechanism of SNHG3 in prostate cancer are lacked. In this work, SNHG3 exerted high expression in prostate cancer cell lines. Suppression of SNHG3 inhibited cell proliferation, migration, EMT (epithelial-mesenchymal transition) process and promoted cell apoptosis. Additionally, it was found that SNHG3 could bind with miR-577. Subsequently, SMURF1 (Smad ubiquitination regulatory factor 1) was identified as a downstream target of miR-577 and had a negative correlation with miR-577. SNHG3 was found to positively regulate SMURF1 expression. Furthermore, rescue assays demonstrated that co-transfection of pcDNA3.1/SMURF1 reversed the effects of SNHG3 knockdown in cell proliferation, migration, EMT process and cell apoptosis. SNHG3 also promoted tumorigenesis in vivo. All the results above explained that SNHG3 accelerated prostate cancer progression by sponging miR-577 to up-regulate SMURF1 expression, suggesting that SNHG3 may act as a biomarker for prostate cancer patients.
Collapse
Affiliation(s)
- Teng Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Xing
- Department of Opthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fan Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yangyang Sun
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shaojin Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingwei Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weixing Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|