1
|
Chen M, Wang T, Tian D, Hai C, Qiu Z. Induction, growth, drug resistance, and metastasis: A comprehensive summary of the relationship between STAT3 and gastric cancer. Heliyon 2024; 10:e37263. [PMID: 39309860 PMCID: PMC11416542 DOI: 10.1016/j.heliyon.2024.e37263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Gastric cancer is a prevalent and highly lethal malignancy that poses substantial challenges to healthcare systems globally. Owing to its often asymptomatic nature in early stages, diagnosis frequently occurs at advanced stages when surgical intervention is no longer a viable option, forcing most patients to rely on nonsurgical treatments such as chemotherapy, targeted therapies, and emerging immunotherapies. Unfortunately, the therapeutic response rates for these treatments are suboptimal, and even among responders, the eventual development of drug resistance remains a significant clinical hurdle. Signal transducer and activator of transcription 3 (STAT3) is a widely expressed cellular protein that plays crucial roles in regulating cellular processes such as growth, metabolism, and immune function. Aberrant activation of the STAT3 pathway has been implicated in the initiation, progression, and therapeutic resistance of several cancers, with gastric cancer being particularly affected. Dysregulated STAT3 signaling not only drives tumorigenesis but also facilitates the development of resistance to chemotherapy and targeted therapies, as well as promotes metastatic dissemination. In this study, we explored the critical role of the STAT3 signaling cascade in the pathogenesis of gastric cancer, its contribution to drug resistance, and its involvement in the metastatic process. Furthermore, we assess recent advances in the development of STAT3 inhibitors and their potential application as therapeutic agents in the treatment of gastric cancer. This work provides a comprehensive overview of the current understanding of STAT3 in gastric cancer and offers a foundation for future research aimed at improving therapeutic outcomes in this challenging disease.
Collapse
Affiliation(s)
- Muyang Chen
- School of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Tongshan Wang
- Gastric Cancer Center, Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dianzhe Tian
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chaorui Hai
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Zixuan Qiu
- School of Public Health, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
2
|
Chen F, Huang Y, Qian Y, Zhao Y, Bu C, Zhang D. A Machine Learning-Driven Surface-Enhanced Raman Scattering Analysis Platform for the Label-Free Detection and Identification of Gastric Lesions. Int J Nanomedicine 2024; 19:9305-9315. [PMID: 39282579 PMCID: PMC11401524 DOI: 10.2147/ijn.s471392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024] Open
Abstract
Background Gastric lesions pose significant clinical challenges due to their varying degrees of malignancy and difficulty in early diagnosis. Early and accurate detection of these lesions is crucial for effective treatment and improved patient outcomes. Methods This paper proposed a label-free and highly sensitive classification method for serum of patients with different degrees of gastric lesions by combining surface-enhanced Raman scattering (SERS) and machine learning analysis. Specifically, we prepared Au lotus-shaped (AuLS) nanoarrays substrates using seed-mediated and liquid-liquid interface self-assembly method for measuring SERS spectra of serum, and then the collected spectra were processed by principal component analysis (PCA) - multi-local means based nearest neighbor (MLMNN) model to achieve differentiation. Results By employing this pattern analysis, AuLS nanoarray substrates can achieve fast, sensitive, and label-free serum spectral detection. The classification accuracy can reach 97.5%, the sensitivity is higher than 96.7%, and the specificity is higher than 95.0%. Moreover, by analyzing the PCs loading plots, the most critical spectral features distinguishing different degrees of gastric lesions were successfully captured. Conclusion This discovery lays the foundation for combining SERS with machine learning for real-time diagnosis and recognition of gastric lesions.
Collapse
Affiliation(s)
- Fengsong Chen
- Department of Gastroenterology, Haimen People's Hospital, Nantong, 226000, People's Republic of China
| | - Yanhua Huang
- Department of Gastroenterology, Haimen People's Hospital, Nantong, 226000, People's Republic of China
| | - Yayun Qian
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Ya Zhao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Chiwen Bu
- Institute of Surgery, Guanyun People's Hospital, Guanyun, 222200, People's Republic of China
| | - Dong Zhang
- Institute of Surgery, Guanyun People's Hospital, Guanyun, 222200, People's Republic of China
| |
Collapse
|
3
|
Ghorbani A, Hosseinie F, Khorshid Sokhangouy S, Islampanah M, Khojasteh-Leylakoohi F, Maftooh M, Nassiri M, Hassanian SM, Ghayour-Mobarhan M, Ferns GA, Khazaei M, Nazari E, Avan A. The prognostic, diagnostic, and therapeutic impact of Long noncoding RNAs in gastric cancer. Cancer Genet 2024; 282-283:14-26. [PMID: 38157692 DOI: 10.1016/j.cancergen.2023.12.006] [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: 05/28/2023] [Revised: 11/27/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
Gastric cancer (GC), ranking as the third deadliest cancer globally, faces challenges of late diagnosis and limited treatment efficacy. Long non-coding RNAs (lncRNAs) emerge as valuable treasured targets for cancer prognosis, diagnosis, and therapy, given their high specificity, convenient non-invasive detection in body fluids, and crucial roles in diverse physiological and pathological processes. Research indicates the significant involvement of lncRNAs in various aspects of GC pathogenesis, including initiation, metastasis, and recurrence, underscoring their potential as novel diagnostic and prognostic biomarkers, as well as therapeutic targets for GC. Despite existing challenges in the clinical application of lncRNAs in GC, the evolving landscape of lncRNA molecular biology holds promise for advancing the survival and treatment outcomes of gastric cancer patients. This review provides insights into recent studies on lncRNAs in gastric cancer, elucidating their molecular mechanisms and exploring the potential clinical applications in GC.
Collapse
Affiliation(s)
- Atousa Ghorbani
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Hosseinie
- Department of Nursing, Faculty of Nursing and Midwifery, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Saeideh Khorshid Sokhangouy
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Muhammad Islampanah
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Nazari
- Department of Health Information Technology and Management, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Liu S, Li W, Liang L, Zhou Y, Li Y. The regulatory relationship between transcription factor STAT3 and noncoding RNA. Cell Mol Biol Lett 2024; 29:4. [PMID: 38172648 PMCID: PMC10763091 DOI: 10.1186/s11658-023-00521-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3), as a key node in numerous carcinogenic signaling pathways, is activated in various tumor tissues and plays important roles in tumor formation, metastasis, and drug resistance. STAT3 is considered a potential subtarget for tumor therapy. Noncoding RNA (ncRNA) is a special type of RNA transcript. Transforming from "junk" transcripts into key molecules involved in cell apoptosis, growth, and functional regulation, ncRNA has been proven to be closely related to various epithelial-mesenchymal transition and drug resistance processes in tumor cells over the past few decades. Research on the relationship between transcription factor STAT3 and ncRNAs has attracted increased attention. To date, existing reviews have mainly focused on the regulation by ncRNAs on the transcription factor STAT3; there has been no review of the regulation by STAT3 on ncRNAs. However, understanding the regulation of ncRNAs by STAT3 and its mechanism is important to comprehensively understand the mutual regulatory relationship between STAT3 and ncRNAs. Therefore, in this review, we summarize the regulation by transcription factor STAT3 on long noncoding RNA, microRNA, and circular RNA and its possible mechanisms. In addition, we provide an update on research progress on the regulation of STAT3 by ncRNAs. This will provide a new perspective to comprehensively understand the regulatory relationship between transcription factor STAT3 and ncRNAs, as well as targeting STAT3 or ncRNAs to treat diseases such as tumors.
Collapse
Affiliation(s)
- Siyi Liu
- Department of Nuclear Medicine, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Wentao Li
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Lin Liang
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China.
| | - Yanling Li
- Department of Nuclear Medicine, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China.
| |
Collapse
|
5
|
Hussain MS, Gupta G, Afzal M, Alqahtani SM, Samuel VP, Hassan Almalki W, Kazmi I, Alzarea SI, Saleem S, Dureja H, Singh SK, Dua K, Thangavelu L. Exploring the role of lncrna neat1 knockdown in regulating apoptosis across multiple cancer types: A review. Pathol Res Pract 2023; 252:154908. [PMID: 37950931 DOI: 10.1016/j.prp.2023.154908] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as pivotal regulators of gene expression, contributing significantly to a diverse range of cellular processes, including apoptosis. One such lncRNA is NEAT1, which is elevated in several types of cancer and aid in cancer growth. However, recent studies have also demonstrated that the knockdown of NEAT1 can inhibit cancer cells proliferation, movement, and infiltration while enhancing apoptosis. This article explores the function of lncRNA NEAT1 knockdown in regulating apoptosis across multiple cancer types. We explore the existing understanding of NEAT1's involvement in the progression of malignant conditions, including its structure and functions. Additionally, we investigate the molecular mechanisms by which NEAT1 modulates the cell cycle, cellular proliferation, apoptosis, movement, and infiltration in diverse cancer types, including acute myeloid leukemia, breast cancer, cervical cancer, colorectal cancer, esophageal squamous cell carcinoma, glioma, non-small cell lung cancer, ovarian cancer, prostate cancer, and retinoblastoma. Furthermore, we review the recent studies investigating the therapeutic potential of NEAT1 knockdown in cancer treatment. Targeting the lncRNA NEAT1 presents a promising therapeutic approach for treating cancer. It has shown the ability to suppress cancer cell proliferation, migration, and invasion while promoting apoptosis in various cancer types.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017 Jaipur, Rajasthan, India
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India; School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Safar M Alqahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK Medical & Health Sciences University, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah, United Arab Emirates
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Shakir Saleem
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Kamal Dua
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
6
|
Li Y, Zhang M, Shang J, Li F, Ren Q, Liu JX. iLncDA-RSN: identification of lncRNA-disease associations based on reliable similarity networks. Front Genet 2023; 14:1249171. [PMID: 37614816 PMCID: PMC10442839 DOI: 10.3389/fgene.2023.1249171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/27/2023] [Indexed: 08/25/2023] Open
Abstract
Identification of disease-associated long non-coding RNAs (lncRNAs) is crucial for unveiling the underlying genetic mechanisms of complex diseases. Multiple types of similarity networks of lncRNAs (or diseases) can complementary and comprehensively characterize their similarities. Hence, in this study, we presented a computational model iLncDA-RSN based on reliable similarity networks for identifying potential lncRNA-disease associations (LDAs). Specifically, for constructing reliable similarity networks of lncRNAs and diseases, miRNA heuristic information with lncRNAs and diseases is firstly introduced to construct their respective Jaccard similarity networks; then Gaussian interaction profile (GIP) kernel similarity networks and Jaccard similarity networks of lncRNAs and diseases are provided based on the lncRNA-disease association network; a random walk with restart strategy is finally applied on Jaccard similarity networks, GIP kernel similarity networks, as well as lncRNA functional similarity network and disease semantic similarity network to construct reliable similarity networks. Depending on the lncRNA-disease association network and the reliable similarity networks, feature vectors of lncRNA-disease pairs are integrated from lncRNA and disease perspectives respectively, and then dimensionality reduced by the elastic net. Two random forests are at last used together on different lncRNA-disease association feature sets to identify potential LDAs. The iLncDA-RSN is evaluated by five-fold cross-validation to analyse its prediction performance, results of which show that the iLncDA-RSN outperforms the compared models. Furthermore, case studies of different complex diseases demonstrate the effectiveness of the iLncDA-RSN in identifying potential LDAs.
Collapse
Affiliation(s)
| | | | - Junliang Shang
- School of Computer Science, Qufu Normal University, Rizhao, China
| | | | | | | |
Collapse
|
7
|
Hou XR, Zhang ZD, Cao XL, Wang XP. Long noncoding RNAs, glucose metabolism and cancer (Review). Oncol Lett 2023; 26:340. [PMID: 37427347 PMCID: PMC10326653 DOI: 10.3892/ol.2023.13925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
Cancer is a serious and potentially life-threatening disease, which, despite numerous advances over several decades, remains a challenge to treat that challenging to detect at an early stage or treat during the later stages. Long noncoding RNAs are >200 nucleotides long and do not possess protein-coding capacity, instead regulating cellular processes, such as proliferation, differentiation, maturation, apoptosis, metastasis, and sugar metabolism. Several studies have shown the role of lncRNAs and glucose metabolism in regulating several key glycolytic enzymes and the activity of multiple functional signaling pathways during tumor progression. Thus, it is possible to further learn about the effects of lncRNA and glycolytic metabolism on tumor diagnosis, treatment, and prognosis through a thorough investigation of the lncRNA expression profiles and glycolytic metabolism in tumors. This may provide a novel strategy for improving the management of several types of cancer.
Collapse
Affiliation(s)
- Xin-Rui Hou
- Graduate School, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
| | - Zhen-Dong Zhang
- Graduate School, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
| | - Xiao-Lan Cao
- Graduate School, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
| | - Xiao-Ping Wang
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
- School of Medicine, Xizang Minzu University, Xianyang, Shaanxi 712082, P.R. China
| |
Collapse
|
8
|
Molecular Interactions of the Long Noncoding RNA NEAT1 in Cancer. Cancers (Basel) 2022; 14:cancers14164009. [PMID: 36011001 PMCID: PMC9406559 DOI: 10.3390/cancers14164009] [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: 07/14/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
As one of the best-studied long noncoding RNAs, nuclear paraspeckle assembly transcript 1 (NEAT1) plays a pivotal role in the progression of cancers. NEAT1, especially its isoform NEAT1-1, facilitates the growth and metastasis of various cancers, excluding acute promyelocytic leukemia. NEAT1 can be elevated via transcriptional activation or stability alteration in cancers changing the aggressive phenotype of cancer cells. NEAT1 can also be secreted from other cells and be delivered to cancer cells through exosomes. Hence, elucidating the molecular interaction of NEAT1 may shed light on the future treatment of cancer. Herein, we review the molecular function of NEAT1 in cancer progression, and explain how NEAT1 interacts with RNAs, proteins, and DNA promoter regions to upregulate tumorigenic factors.
Collapse
|
9
|
Non-coding RNAs in ferroptotic cancer cell death pathway: meet the new masters. Hum Cell 2022; 35:972-994. [PMID: 35415781 DOI: 10.1007/s13577-022-00699-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/01/2022] [Indexed: 02/08/2023]
Abstract
Despite the recent advances in cancer therapy, cancer chemoresistance looms large along with radioresistance, a major challenge in dire need of thorough and minute investigation. Not long ago, cancer cells were reported to have proven refractory to the ferroptotic cell death, a newly discovered form of regulated cell death (RCD), conspicuous enough to draw attention from scholars in terms of targeting ferroptosis as a prospective therapeutic strategy. However, our knowledge concerning the underlying molecular mechanisms through which cancer cells gain immunity against ferroptosis is still in its infancy. Of late, the implication of non-coding RNAs (ncRNAs), including circular RNAs (circRNAs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) in ferroptosis has been disclosed. Nevertheless, precisely explaining the molecular mechanisms behind the contribution of ncRNAs to cancer radio/chemotherapy resistance remains a challenge, requiring further clarification. In this review, we have presented the latest available information on the ways and means of regulating ferroptosis by ncRNAs. Moreover, we have provided important insights about targeting ncRNAs implicated in ferroptosis with the hope of opening up new horizons for overcoming cancer treatment modalities. Though a long path awaits until we make this ambitious dream come true, recent progress in gene therapy, including gene-editing technology will aid us to be optimistic that ncRNAs-based ferroptosis targeting would soon be on stream as a novel therapeutic strategy for treating cancer.
Collapse
|
10
|
Jiang L, Zhang Y, Su P, Ma Z, Ye X, Kang W, Liu Y, Yu J. Long non-coding RNA HNF1A-AS1 induces 5-FU resistance of gastric cancer through miR-30b-5p/EIF5A2 pathway. Transl Oncol 2022; 18:101351. [PMID: 35092904 PMCID: PMC8802127 DOI: 10.1016/j.tranon.2022.101351] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/22/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the leading causes of cancer-related deaths worldwide and chemoresistance is a major cause for its poor prognosis. Long non-coding RNAs (lncRNAs) are associated with cancer chemoresistance. The current study sought to explore the mechanism of lncRNA HNF1A antisense RNA 1 (HNF1A-AS1) in mediating 5-fluorouracil (5-FU) resistance of GC. METHODS qRT-PCR was performed to detect the expression level of HNF1A-AS1 in GC tissues and cells. Abnormal expression of HNF1A-AS1 in GC cells was induced by lentivirus infection. Protein levels of EIF5A2, E-Cadherin, Vimentin and N-Cadherin were detected using western blot. Competitive endogenous RNA (ceRNA) mechanisms were explored through luciferase assays and RNA immunoprecipitation (RIP) assays. Functional experiments of chemoresistance were performed by CCK-8 assays, colony formation assays and flow cytometry with the treatment of 5-FU. Mouse tumor xenograft assays were performed to verify the findings in vivo. RESULTS The findings showed HNF1A-AS1 was significantly upregulated in GC tissues especially in chemoresistance group. Findings from in vitro and in vivo experiments showed HNF1A-AS1 increased cell viability and proliferation, repressed apoptosis and promoted xenograft tumors growth in the presence of 5-FU. Mechanistic studies revealed HNF1A-AS1 promoted chemoresistance by facilitating epithelial mesenchymal transition (EMT) process through upregulating EIF5A2 expression and HNF1A-AS1 acted as a sponge of miR-30b-5p. CONCLUSIONS The findings from the current study showed HNF1A-AS1 promoted 5-FU resistance by acting as a ceRNA of miR-30b-5p and promoting EIF5A2-induced EMT process in GC. This indicates that HNF1A-AS1 is a potential therapeutic target for alleviating GC chemoresistance.
Collapse
Affiliation(s)
- Lin Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujin, Dongcheng District, Beijing 100730, China; Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yingjing Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujin, Dongcheng District, Beijing 100730, China; Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Pengfei Su
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujin, Dongcheng District, Beijing 100730, China; Graduate School, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Zhiqiang Ma
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujin, Dongcheng District, Beijing 100730, China
| | - Xin Ye
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujin, Dongcheng District, Beijing 100730, China
| | - Weiming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujin, Dongcheng District, Beijing 100730, China
| | - Yuqin Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Jianchun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujin, Dongcheng District, Beijing 100730, China.
| |
Collapse
|
11
|
Zhou H, Wang Y, Liu Z, Zhang Z, Xiong L, Wen Y. Recent advances of NEAT1-miRNA interactions in cancer. Acta Biochim Biophys Sin (Shanghai) 2022; 54:153-162. [PMID: 35538025 PMCID: PMC9827865 DOI: 10.3724/abbs.2021022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/20/2021] [Indexed: 11/25/2022] Open
Abstract
With high incidence rate, cancer is the main cause of death in humans. Non-coding RNAs, as novel master regulators, especially long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), play important roles in the regulation of tumorigenesis. lncRNA NEAT1 has recently gained much attention, as it is dysregulated in a broad spectrum of cancers, where it acts as either an oncogene or a tumor suppressor gene. Accumulating evidence shows that NEAT1 is correlated with the process of carcinogenesis, including proliferation, invasion, survival, drug resistance, and metastasis. NEAT1 is considered to be a biomarker and a novel therapeutic target for the diagnosis and prognosis of different cancer types. The mechanisms by which NEAT1 plays a critical role in cancers are mainly via interactions with miRNAs. NEAT1-miRNA regulatory networks play significant roles in tumorigenesis, which has attracted much attention from researchers around the world. In this review, we summarize the interaction of NEAT1 with miRNAs in the regulation of protein-coding genes in cancer. A better understanding of the NEAT1-miRNA interactions in cancer will help develop new diagnostic biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Hui Zhou
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Yongxiang Wang
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Zhongtao Liu
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Zijian Zhang
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Li Xiong
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Yu Wen
- />Department of General SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| |
Collapse
|
12
|
+HOXA10-AS Promotes Malignant Phenotypes of Gastric Cancer via Upregulating HOXA10. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:1846687. [PMID: 35222681 PMCID: PMC8866012 DOI: 10.1155/2022/1846687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/16/2021] [Accepted: 12/29/2021] [Indexed: 12/07/2022]
Abstract
Objective To study the role of long noncoding RNA HOXA10-AS in gastric cancer (GC) and its underlying mechanism which is one of the most common and fetal malignancies. Long noncoding RNA HOXA10-AS is highly expressed and acts in an oncogenic role in cancers. However, its roles in GC are still unknown. Methods The expression of HOXA10-AS and HOXA10 in GC tissues from the TCGA database was analyzed. Western blot and qRT-PCR assays were applied to examine the expression of HOXA10-AS and HOXA10. Cell proliferation was evaluated with CCK-8 and EdU incorporation assays. Cell apoptosis was analyzed by flow cytometry. Migratory and invasive capacities were evaluated with wound healing and transwell assays. Results HOXA10-AS and HOXA10 were upregulated in GC, and their expressions were positively correlated. Knockdown of HOXA10-AS inhibited HOXA10 expression in GC cells. Furthermore, knockdown of HOXA10-AS restrained GC cell proliferation, migration, and invasion but promoted apoptosis. In addition, overexpression of HOXA10-AS promoted malignant phenotypes of GC cells, but all these effects could be reversed by knockdown of HOXA10. Conclusion HOXA10-AS promoted GC cell proliferation, migration and invasion and enhanced apoptosis via upregulating HOXA10. Our study implies a novel regulatory mechanism of malignant phenotypes and provides potential therapeutic targets for GC.
Collapse
|
13
|
Fernandes M, Marques H, Teixeira AL, Medeiros R. ceRNA Network of lncRNA/miRNA as Circulating Prognostic Biomarkers in Non-Hodgkin Lymphomas: Bioinformatic Analysis and Assessment of Their Prognostic Value in an NHL Cohort. Int J Mol Sci 2021; 23:ijms23010201. [PMID: 35008626 PMCID: PMC8745130 DOI: 10.3390/ijms23010201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
Research has been focusing on identifying novel biomarkers to better stratify non-Hodgkin lymphoma patients based on prognosis. Studies have demonstrated that lncRNAs act as miRNA sponges, creating ceRNA networks to regulate mRNA expression, and its deregulation is associated with lymphoma development. This study aimed to identify novel circulating prognostic biomarkers based on miRNA/lncRNA-associated ceRNA network for NHL. Herein, bioinformatic analysis was performed to construct ceRNA networks for hsa-miR-150-5p and hsa-miR335-5p. Then, the prognostic value of the miRNA–lncRNA pairs’ plasma levels was assessed in a cohort of 113 NHL patients. Bioinformatic analysis identified MALAT1 and NEAT1 as hsa-miR-150-5p and has-miR-335-5p sponges, respectively. Plasma hsa-miR-150-5p/MALAT1 and hsa-miR335-5p/NEAT1 levels were significantly associated with more aggressive and advanced disease. The overall survival and progression-free survival analysis indicated that hsa-miR-150-5p/MALAT1 and hsa-miR335-5p/NEAT1 pairs’ plasma levels were remarkably associated with NHL patients’ prognosis, being independent prognostic factors in a multivariate Cox analysis. Low levels of hsa-miR-150-5p and hsa-miR-335-5p combined with high levels of the respective lncRNA pair were associated with poor prognosis of NHL patients. Overall, the analysis of ceRNA network expression levels may be a useful prognostic biomarker for NHL patients and could identify patients who could benefit from more intensive treatments.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cohort Studies
- Computational Biology
- Disease-Free Survival
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- Humans
- Lymphoma, Non-Hodgkin/blood
- Lymphoma, Non-Hodgkin/genetics
- MicroRNAs/blood
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Prognosis
- RNA, Long Noncoding/blood
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Risk Factors
Collapse
Affiliation(s)
- Mara Fernandes
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (A.L.T.)
- Research Department of the Portuguese League against Cancer Regional Nucleus of the North (LPCC-NRN), 4200-177 Porto, Portugal
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
| | - Herlander Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
- Department of Oncology, Hospital de Braga, 4710-243 Braga, Portugal
- CINTESIS, Center for Health Technology and Services Research, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (A.L.T.)
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-513 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (A.L.T.)
- Research Department of the Portuguese League against Cancer Regional Nucleus of the North (LPCC-NRN), 4200-177 Porto, Portugal
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-513 Porto, Portugal
- Biomedical Research Center (CEBIMED), Faculty of Health Sciences of Fernando Pessoa University (UFP), 4249-004 Porto, Portugal
- Correspondence: ; Tel.: +351-225-084-000 (ext. 5414)
| |
Collapse
|
14
|
Liu Y, Ding W, Yu W, Zhang Y, Ao X, Wang J. Long non-coding RNAs: Biogenesis, functions, and clinical significance in gastric cancer. Mol Ther Oncolytics 2021; 23:458-476. [PMID: 34901389 PMCID: PMC8637188 DOI: 10.1016/j.omto.2021.11.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is one of the most prevalent malignant tumor types and the third leading cause of cancer-related death worldwide. Its morbidity and mortality are very high due to a lack of understanding about its pathogenesis and the slow development of novel therapeutic strategies. Long non-coding RNAs (lncRNAs) are a class of non-coding RNAs with a length of more than 200 nt. They play crucial roles in a wide spectrum of physiological and pathological processes by regulating the expression of genes involved in proliferation, differentiation, apoptosis, cell cycle, invasion, metastasis, DNA damage, and carcinogenesis. The aberrant expression of lncRNAs has been found in various cancer types. A growing amount of evidence demonstrates that lncRNAs are involved in many aspects of GC pathogenesis, including its occurrence, metastasis, and recurrence, indicating their potential role as novel biomarkers in the diagnosis, prognosis, and therapeutic targets of GC. This review systematically summarizes the biogenesis, biological properties, and functions of lncRNAs and highlights their critical role and clinical significance in GC. This information may contribute to the development of better diagnostics and treatments for GC.
Collapse
Affiliation(s)
- Ying Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266021, China
| | - Wei Ding
- Department of Comprehensive Internal Medicine, Affiliated Hospital, Qingdao University, Qingdao 266003, China
| | - Wanpeng Yu
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266021, China
| | - Xiang Ao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Jianxun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
- School of Basic Medical Sciences, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| |
Collapse
|
15
|
Rao X, Liu X, Liu N, Zhang Y, Zhang Z, Zhou L, Han G, Cen R, Shi N, Zhu H, Gong H, Huang C, Ji Q, Li Q. Long noncoding RNA NEAT1 promotes tumorigenesis in H. pylori gastric cancer by sponging miR-30a to regulate COX-2/BCL9 pathway. Helicobacter 2021; 26:e12847. [PMID: 34396632 DOI: 10.1111/hel.12847] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) is a carcinogenic factor for gastric cancer. Our previous study demonstrated that H. pylori decreased the expression of micro-RNA (miRNA)-30a to promote the tumorigenesis of gastric cancer. However, the upstream regulatory molecules of miR-30a are not well elucidated. In this study, we found the long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) may sponge miR-30a to regulate COX-2/BCL9 pathway. METHODS The expression of NEAT1 was detected in gastric cancer tissues and tumor-adjacent tissues by fluorescence in situ hybridization (FISH) analysis and RT-qPCR. LncRNA-miRNA interaction networks were constructed using the RNAhybrid and starBase v.2.0. and then validated using a dual-luciferase reporter assay. The effects of NEAT1 dysregulation on the proliferative, migratory, and invasive abilities of H. pylori filtrate-infected gastric cancer cells were observed by cell counting kit-8 (CCK-8), colony formation, wound healing test, and transwell assays. Western blot and RT-qPCR were performed to detect protein and RNA expression. Immunohistochemistry (IHC) was carried out to analyze the localization and expression of COX-2 and BCL9. RESULTS FISH and RT-qPCR demonstrated that the expression of NEAT1 was up-regulated in gastric cancer tissues, especially in H. pylori-infected gastric cancer tissues, and the expression of NEAT1 was negatively correlated with miR-30a (miR-30a-3p and miR-30a-5p). The upregulation of NEAT1 enhanced proliferation, migration, and invasion of H. pylori filtrate-infected gastric cancer cells, while the downregulation of NEAT1 decreased these abilities, and miR-30a could reverse the effect of NEAT1 on these abilities. The dual-luciferase reporter assay identified that NEAT1 directly targeted miR-30a (miR-30a-3p and miR-30a-5p). Because miR-30a (miR-30a-3p and miR-30a-5p) negatively regulates the expression of downstream COX-2 and BCL9, NEAT1 was identified to upregulate indirectly the expression of COX-2 and BCL9. IHC showed that the expression of COX-2 and BCL9 was increased in H. pylori gastric cancer tissues. CONCLUSION The study demonstrated that lncRNA NEAT1 may act as a promoter of tumorigenesis in H. pylori gastric cancer, by sponging miR-30a (miR-30a-3p and miR-30a-5p) to regulate the COX-2/BCL9 pathway.
Collapse
Affiliation(s)
- Xiwu Rao
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan Liu
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ningning Liu
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Zhang
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhaozhou Zhang
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihong Zhou
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gang Han
- Department of Gastrointestinal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Cen
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nuolin Shi
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huirong Zhu
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hangjun Gong
- Department of Gastrointestinal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qing Ji
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
Homayoonfal M, Asemi Z, Yousefi B. Targeting long non coding RNA by natural products: Implications for cancer therapy. Crit Rev Food Sci Nutr 2021:1-29. [PMID: 34783279 DOI: 10.1080/10408398.2021.2001785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In spite of achieving substantial progress in its therapeutic strategies, cancer-associated prevalence and mortality are persistently rising globally. However, most malignant cancers either cannot be adequately diagnosed at the primary phase or resist against multiple treatments such as chemotherapy, surgery, radiotherapy as well as targeting therapy. In recent decades, overwhelming evidences have provided more convincing words on the undeniable roles of long non-coding RNAs (lncRNAs) in incidence and development of various cancer types. Recently, phytochemical and nutraceutical compounds have received a great deal of attention due to their inhibitory and stimulatory effects on oncogenic and tumor suppressor lncRNAs respectively that finally may lead to attenuate various processes of cancer cells such as growth, proliferation, metastasis and invasion. Therefore, application of phytochemicals with anticancer characteristics can be considered as an innovative approach for treating cancer and increasing the sensitivity of cancer cells to standard prevailing therapies. The purpose of this review was to investigate the effect of various phytochemicals on regulation of lncRNAs in different human cancer and evaluate their capabilities for cancer treatment and prevention.
Collapse
Affiliation(s)
- Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
The Role of Long Non-coding RNA, Nuclear Enriched Abundant Transcript 1 (NEAT1) in Cancer and Other Pathologies. Biochem Genet 2021; 60:843-867. [PMID: 34689290 DOI: 10.1007/s10528-021-10138-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/23/2021] [Indexed: 02/08/2023]
Abstract
Nuclear enriched abundant transcript 1 (NEAT1), consisting of two kinds of lncRNAs of 3.7 kB NEAT1-1 and 23 kB NEAT1-2, can be highly expressed in organs and tissues such as the ovary, prostate, colon, and pancreas, and is involved in paraspeckle formation and mRNA editing and gene expression. Therefore, NEAT1 is a potential biomarker for the treatment of a variety of diseases, which may be caused by two factors (isoforms of NEAT1 and NEAT1 sponging miRNA as ceRNA). However, there is still much confusion about the mechanism and downstream effector between the abnormal expression of NEAT1 and various diseases. This review summarizes recent research progress on NEAT1 in cancer and other pathologies and provides a more reliable theoretical basis for the treatment of related diseases.
Collapse
|
18
|
Li K, Yao T, Zhang Y, Li W, Wang Z. NEAT1 as a competing endogenous RNA in tumorigenesis of various cancers: Role, mechanism and therapeutic potential. Int J Biol Sci 2021; 17:3428-3440. [PMID: 34512157 PMCID: PMC8416723 DOI: 10.7150/ijbs.62728] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/24/2021] [Indexed: 12/24/2022] Open
Abstract
The nuclear paraspeckle assembly transcript 1 (NEAT1) is a long non-coding RNA (lncRNA) that is upregulated in a variety of human cancer types. Increasing evidence has shown that the elevation of NEAT1 in cancer cells promotes cell growth, migration, and invasion and inhibits cell apoptosis. It is also known that lncRNAs act as a competing endogenous RNA (ceRNA) by sponging microRNAs (miRNAs) to alter the expression levels of their target genes in the development of cancers. Therefore, it is important to understand the molecular mechanisms underlying this observation. In this review, specific emphasis was placed on NEAT1's role in tumor development. We also summarize and discuss the feedback roles of NEAT1/miRNA/target network in the progression of various cancers. As our understanding of the role of NEAT1 during tumorigenesis improves, its therapeutic potential as a biomarker and/or target for cancer also becomes clearer.
Collapse
Affiliation(s)
- Kun Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Tongyue Yao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Yu Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Wen Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Ziqiang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China.,Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| |
Collapse
|
19
|
Research updates on the clinical implication of long noncoding RNA in digestive system cancers and chemoresistance. 3 Biotech 2021; 11:423. [PMID: 34603923 DOI: 10.1007/s13205-021-02971-w] [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: 03/20/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are implicated in various biological processes, such as cell proliferation, differentiation, apoptosis, migration, and invasion. They are also key players in various biological pathways. LncRNA was considered as 'translational noise' before 1980s. It has been reported that lncRNAs are aberrantly expressed in different cancers, either as oncogene or tumor suppressor gene. Therefore, more and more lncRNAs are recognized as potential diagnostic biomarkers and/or therapeutic targets. As competitive endogenous RNA, lncRNAs can interact with microRNA to alter the expression of target genes, which may have extensive clinical implications in cancers, including diagnosis, treatment, prognosis, and chemoresistance. This review comprehensively summarizes the functions and clinical relevance of lncRNAs in digestive system cancers, especially as a potential tool to overcome chemoresistance.
Collapse
|
20
|
Zhao Y, Wang Z, Gao M, Wang X, Feng H, Cui Y, Tian X. lncRNA MALAT1 regulated ATAD2 to facilitate retinoblastoma progression via miR-655-3p. Open Med (Wars) 2021; 16:931-943. [PMID: 34222668 PMCID: PMC8231467 DOI: 10.1515/med-2021-0290] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 04/03/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was reported as an oncogene in many tumors including retinoblastoma (RB). This research mainly focused on the functions and mechanism of MALAT1 in RB. MALAT1 was upregulated in RB tissues and cells, and it served as a competing endogenous RNA (ceRNA) and inhibited miRNA-655-3p (miR-655-3p) expression, which eventually regulated the expression of miR-655-3p downstream target ATPase Family AAA Domain Containing 2 (ATAD2). The level of ATAD2 significantly increased, while that of miR-655-3p remarkably decreased in RB tissues and cells. MALAT1 depletion inhibited cell proliferation, metastasis, and epithelial-mesenchymal transition (EMT), but promoted apoptosis in vitro and blocked xenograft tumor growth in vivo. MALAT1 exerted its oncogenic functions in RB by regulating miR-655-3p/ATAD2 axis.
Collapse
Affiliation(s)
- Yuxin Zhao
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Zhaoxia Wang
- Department of Pediatric, Weihai Central Hospital, Weihai, Shandong, China
| | - Meili Gao
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Xuehong Wang
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Hui Feng
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Yuanyuan Cui
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| | - Xia Tian
- Department of Ophthalmology, Weihai Central Hospital, No. 3, Mishandongluxi, Wendeng District, Weihai, 264400, Shandong, China
| |
Collapse
|
21
|
Wang K, Wang B, Wang Z, Yang R. Alginic acid inhibits non-small cell lung cancer-induced angiogenesis via activating miR-506 expression. J Nat Med 2021; 75:553-564. [PMID: 33666835 DOI: 10.1007/s11418-021-01493-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/05/2021] [Indexed: 12/24/2022]
Abstract
Angiogenesis is a key event in non-small cell lung cancer progression. Alginic acid (AA), a kind of naturally occurring polyuronic acid, is generally enriched in edible brown algae. Recent studies have uncovered its anti-anaphylactic and anti-inflammatory properties. However, the effects of AA on human malignancies remain unknown. Herein, efficient inhibition of AA on NSCLC-induced angiogenesis was observed with tube formation and xenograft models. Subsequent results indicated that AA downregulated the expression of VEGF-A, a key angiogenesis-inducing cytokine. In addition, AA downregulated STAT3, a transcriptional inducer of VEGF-A and increased non-coding RNA miR-506 expression, respectively. Furthermore, miR-506 directly modulated STAT3 relying on base pairing the 3'-UTR in STAT3 mRNA. We also found that abrogation of miR-506 abolished the inhibitory effect of AA on VEGF-A expression and NSCLC-induced angiogenesis. Finally, xenografts experiments also showed that oral administration of AA could significantly attenuate NSCLC angiogenesis, indicated by decreased micro-vessel density (MVD) and the MVD marker CD31 expression in xenografts tissues. Correspondingly, AA treatment also downregulated VEGF-A, STAT3 and increased miR-506 expression in xenografts samples, respectively. Taken together, these results suggested that AA could suppress NSCLC-induced angiogenesis via miR-506/STAT3/VEGF-A axis. .
Collapse
Affiliation(s)
- Keping Wang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing Medical University Affiliated Brain Hospital (Thoracic Academy), No.215, Guangzhou Road, Gulou District, Nanjing, 210029, People's Republic of China
| | - Bin Wang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing Medical University Affiliated Brain Hospital (Thoracic Academy), No.215, Guangzhou Road, Gulou District, Nanjing, 210029, People's Republic of China
| | - Zunqiao Wang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing Medical University Affiliated Brain Hospital (Thoracic Academy), No.215, Guangzhou Road, Gulou District, Nanjing, 210029, People's Republic of China
| | - Rusong Yang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing Medical University Affiliated Brain Hospital (Thoracic Academy), No.215, Guangzhou Road, Gulou District, Nanjing, 210029, People's Republic of China.
| |
Collapse
|
22
|
Ji X, Yan Y, Ma N, He G, Wang K, Zhang Y, Yin J, Song C, Wang P, Ye H, Dai L, Zhang J, Wang K. Variant of SNPs at lncRNA NEAT1 contributes to gastric cancer susceptibility in Chinese Han population. Int J Clin Oncol 2021; 26:694-700. [PMID: 33464437 DOI: 10.1007/s10147-020-01852-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The long noncoding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1) has been implicated in many tumors risk including gastric cancer. However, the association of single nucleotide polymorphisms (SNPs) at NEAT1 with gastric cancer risk has not been studied. The aim of this study was to investigate the association between SNPs in NEAT1 and gastric cancer susceptibility. METHODS In this study, four SNPs in lncRNA NEAT1 were selected for genotyping in 484 gastric cancer patients and 484 controls in Chinese Han population. Quantitative real-time PCR (qRT-PCR) was conducted to evaluate the potential function of rs3825071. Attributable risk percentage (ARP) and population attributable risk percentage (PARP) were used to assess the epidemiological effect. RESULTS In the dominant model (GG), the genotypes AG + AA of rs3825071 and rs7943779 were associated with an increased risk of gastric cancer (OR = 1.72, 95%CI = 1.27-2.32 and OR = 1.63, 95%CI = 1.19-2.22). Individuals harboring ≥ 3 risk alleles have higher risk of gastric cancer (OR = 1.88, 95% CI = 1.26-2.80, P = 0.002). ARP and PARP associated with gastric cancer were 42.53% and 10.88% for rs3825071, and were 33.78% and 6.26% for rs7943779, respectively. Furthermore, compared with the genotype GG of rs3825071, the genotypes AG and AA had higher expression of NEAT1. CONCLUSIONS We found that the genetic variations in NEAT1 were significantly associated with risk of gastric cancer. The G > A variant of rs3825071 may confer gastric cancer susceptibility by changed biological effects to increase the expression of NEAT1.
Collapse
Affiliation(s)
- Xuanke Ji
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Yali Yan
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Nan Ma
- Henan Academy of Medical Sciences, Zhengzhou, 450003, Henan Province, China
| | - Gui He
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Kunyan Wang
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Yuehua Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Jingjing Yin
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Chunhua Song
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Peng Wang
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Hua Ye
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Liping Dai
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Jianying Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Kaijuan Wang
- Department of Epidemiology, College of Public Health, Zhengzhou University, No 100 Kexue Avenue, Zhengzhou City, 450001, China.
- Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou University, Zhengzhou, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
23
|
Guo T, Wang W, Ji Y, Zhang M, Xu G, Lin S. LncRNA PROX1-AS1 Facilitates Gastric Cancer Progression via miR-877-5p/PD-L1 Axis. Cancer Manag Res 2021; 13:2669-2680. [PMID: 33776485 PMCID: PMC7989960 DOI: 10.2147/cmar.s275352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction Growing evidences imply that multiple long non-coding RNAs (lncRNAs) play a significant role in the treatment of cancer. Therefore, it is of great significance to discover new biomarkers or therapeutic targets of gastric cancer (GC). However, the potential molecular mechanism of lncPROX1-AS1 in GC remains unknown. The objective of current study is to investigate the effect of PROX1-AS1 in GC. Methods Thus, we detect that PROX1-AS1 is over-expressed in tissues and cell lines of GC using qRT-PCR analysis. CCK-8, colony formation, flow cytometry, wounding healing and transwell analyses were performed to explore the effect of PROX1-AS1 on GC malignant behaviors. Results It is further disclosed that silencing of PROX1-AS1 represses cell proliferation, migration, and invasion, whereas promotes cell apoptosis in GC. Bioinformatics analysis suggests that miR-877-5p is negatively regulated by PROX1-AS1 and ectopic of miR-877-5p alleviates the malignant behaviors of GC. Subsequently, miR-877-5p suppresses the activity of PD-L1-3ʹ UTR. At last, rescue assays demonstrated that the GC progression is suppressed by sh-PROX1-AS1 and facilitated on account of miR-877-5p inhibitors and then is retrieved by sh-PD-L1. Discussion Our findings reveal that PROX1-AS1 exerts its role via miR-877-5p/PD-L1 axis in the GC progression, suggesting that PROX1-AS1 may represent a new therapeutic target for the diagnosis and treatment of GC patients.
Collapse
Affiliation(s)
- TianWei Guo
- Department of Pathology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, People's Republic of China
| | - Wei Wang
- Department of Pathology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, People's Republic of China
| | - YueXia Ji
- Department of Pathology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, People's Republic of China
| | - Min Zhang
- Department of Pathology, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - GuoYing Xu
- School of Medical Technology, Jiangsu College of Nursing, Huai'an, Jiangsu, People's Republic of China
| | - Sen Lin
- The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huai'an, Jiangsu, People's Republic of China
| |
Collapse
|
24
|
Guo L, Tang Y, Wang Y, Xu H. Prognostic Value of lncRNA NEAT1 as a New Biomarker in Digestive System Tumors: a Systematic Study and Meta-analysis. Expert Rev Mol Diagn 2021; 21:91-99. [PMID: 33550874 DOI: 10.1080/14737159.2021.1874921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Nuclear paraspeckle assembly transcript 1 (NEAT1), a newly found lncRNA, is found abnormally expressed in digestive system tumors. This meta-analysis aims to evaluate the effect of NEAT1 on digestive system tumors. METHODS An analysis was conducted to investigate NEAT1 expression in digestive system tumors from the PubMed, Embase, and Web of Science databases. The relationship between NEAT1 expression and patient overall survival (OS) and clinicopathology was evaluated by correlation analysis with the pooled hazard ratio (HR), 95% confidence interval (CI), and odds ratio (OR). RESULTS A total of 12 published studies were enrolled in this meta-analysis. The NEAT1 overexpression was significantly associated with poor OS (HR = 1.64, 95% CI:1.41-1.91, p < 0.05), lymphatic metastasis (OR = 2.70, 95% CI: 2.02-3.61, p < 0.05), distal metastasis (OR = 3.01, 95% CI: 1.97-4.59, p < 0.05) and advanced tumor stage (OR = 3.04, 95% CI: 2.32-3.99, p < 0.05). However, digestive system tumor patients with high NEAT1 expression was not related to the patients' age (OR = 0.91, 95% CI: 0.65-1.26, p = 0.561), gender (OR = 1.04, 95% CI: 0.81-1.33, p = 0.761), tumor size (OR = 1.84, 95% CI: 0.88-3.88, p = 0.106), and tumor differentiation (OR = 0.86, 95% CI: 0.51-1.44, p = 0.570). CONCLUSION Collectively, NEAT1 can be used as a potential biomarker to predict the prognosis of patients with digestive system tumors, which is worth verifying in clinical practice.
Collapse
Affiliation(s)
- Lu Guo
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Tang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yukai Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Hong Xu
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
25
|
Ashrafizadeh M, Gholami MH, Mirzaei S, Zabolian A, Haddadi A, Farahani MV, Kashani SH, Hushmandi K, Najafi M, Zarrabi A, Ahn KS, Khan H. Dual relationship between long non-coding RNAs and STAT3 signaling in different cancers: New insight to proliferation and metastasis. Life Sci 2021; 270:119006. [PMID: 33421521 DOI: 10.1016/j.lfs.2020.119006] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022]
Abstract
Uncontrolled growth and metastasis of cancer cells is an increasing challenge for overcoming cancer, and improving survival of patients. Complicated signaling networks account for proliferation and invasion of cancer cells that need to be elucidated for providing effective cancer therapy, and minimizing their malignancy. Long non-coding RNAs (lncRNAs) are RNA molecules with a length of more than 200 nucleotides. They participate in cellular events, and their dysregulation in a common phenomenon in different cancers. Noteworthy, lncRNAs can regulate different molecular pathways, and signal transducer and activator of transcription 3 (STAT3) is one of them. STAT3 is a tumor-promoting factors in cancers due to its role in cancer proliferation (cell cycle progression and apoptosis inhibition) and metastasis (EMT induction). LncRNAs can function as upstream mediators of STAT3 pathway, reducing/enhancing its expression. This dual relationship is of importance in affecting proliferation and metastasis of cancer cells. The response of cancer cells to therapy such as chemotherapy and radiotherapy is regulated by lncRNA/STAT3 axis. Tumor-promoting lncRNAs including NEAT1, SNHG3 and H19 induces STAT3 expression, while tumor-suppressing lncRNAs such as MEG3, PTCSC3 and NKILA down-regulate STAT3 expression. Noteworthy, upstream mediators of STAT3 such as microRNAs can be regulated by lncRNAs. These complicated signaling networks are mechanistically described in the current review.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
| | | | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirabbas Haddadi
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | | | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| |
Collapse
|
26
|
Non-coding RNAs underlying chemoresistance in gastric cancer. Cell Oncol (Dordr) 2020; 43:961-988. [PMID: 32495294 DOI: 10.1007/s13402-020-00528-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 04/17/2020] [Accepted: 04/24/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is a major health issue in the Western world. Current clinical imperatives for this disease include the identification of more effective biomarkers to detect GC at early stages and enhance the prevention and treatment of metastatic and chemoresistant GC. The advent of non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long-non coding RNAs (lncRNAs), has led to a better understanding of the mechanisms by which GC cells acquire features of therapy resistance. ncRNAs play critical roles in normal physiology, but their dysregulation has been detected in a variety of cancers, including GC. A subset of ncRNAs is GC-specific, implying their potential application as biomarkers and/or therapeutic targets. Hence, evaluating the specific functions of ncRNAs will help to expand novel treatment options for GC. CONCLUSIONS In this review, we summarize some of the well-known ncRNAs that play a role in the development and progression of GC. We also review the application of such ncRNAs in clinical diagnostics and trials as potential biomarkers. Obviously, a deeper understanding of the biology and function of ncRNAs underlying chemoresistance can broaden horizons toward the development of personalized therapy against GC.
Collapse
|
27
|
NEAT1 and Paraspeckles in Cancer Development and Chemoresistance. Noncoding RNA 2020; 6:ncrna6040043. [PMID: 33143162 PMCID: PMC7712271 DOI: 10.3390/ncrna6040043] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 09/21/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Non-coding RNA were previously thought to be biologically useless molecules arising from simple transcriptional noise. These are now known to be an integral part of cellular biology and pathology. The wide range of RNA molecules have a diverse range of structures, functions, and mechanisms of action. However, structural long non-coding RNAs (lncRNAs) are a particular class of ncRNA that are proving themselves more and more important in cellular biology, as the exact structures that such RNAs form and stabilise become more understood. Nuclear Enriched Abundant Transcript 1 (NEAT1) is a specific structural RNA emerging as a critical component in the progress and development of cancer. NEAT1 forms part of multiple biological pathways, acting through a diverse group of mechanisms. The most important of these is the formation of the paraspeckle, through which it can influence the stability of a tumour to develop resistance to drugs. This review will thus cover the range of effects by which NEAT1 interacts with cancer progression in order to describe the various roles of NEAT1 in chemoresistance, as well as to identify drug targets that protein research alone could not provide.
Collapse
|
28
|
Wu D, Li H, Wang J, Li H, Xiao Q, Zhao X, Huo Z. LncRNA NEAT1 promotes gastric cancer progression via miR-1294/AKT1 axis. Open Med (Wars) 2020; 15:1028-1038. [PMID: 33336058 PMCID: PMC7718639 DOI: 10.1515/med-2020-0218] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/17/2020] [Accepted: 07/08/2020] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) were reported to promote the development of gastric cancer (GC). Nuclear-enriched abundant transcript 1 (NEAT1) played a great role in diverse cancers, but the mechanism of NEAT1 in GC remains indistinct. NEAT1 and AKT1 were distinctly up-regulated and miR-1294 was down-regulated in GC tissues and cells. Cell proliferation and metastasis were refrained but apoptosis was promoted in GC cells after knockdown of NEAT1. NEAT1 negatively regulated miR-1294 expression, and the miR-1294 inhibitor reverted the si-NEAT1-induced effect on GC cells. NEAT1 modulated AKT1 expression through miR-1294, and the si-NEAT1-induced effect was relieved by AKT1. NEAT1 affected phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway via regulating miR-1294 and AKT1. NEAT1 could modulate cell proliferation, apoptosis, and metastasis in GC cells by regulating the PI3K/AKT/mTOR signaling pathway via the miR-1294/AKT1 axis, showing the great potential for NEAT1 as a valid biomarker in the progression and treatment of GC.
Collapse
Affiliation(s)
- Dianchao Wu
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Hui Li
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Junfeng Wang
- Department of Colorectal Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Li
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Qihai Xiao
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Xiaofeng Zhao
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| | - Zhibin Huo
- Department of Surgical Oncology, Xingtai People’s Hospital, No.16, Hongxing Street, Xingtai, 054031, Hebei, China
| |
Collapse
|
29
|
Zang J, Zheng MH, Cao XL, Zhang YZ, Zhang YF, Gao XY, Cao Y, Shi M, Han H, Liang L. Adenovirus infection promotes the formation of glioma stem cells from glioblastoma cells through the TLR9/NEAT1/STAT3 pathway. Cell Commun Signal 2020; 18:135. [PMID: 32843056 PMCID: PMC7448505 DOI: 10.1186/s12964-020-00598-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Glioma stem cells (GSCs) are glioma cells with stemness and are responsible for a variety of malignant behaviors of glioma. Evidence has shown that signals from tumor microenvironment (TME) enhance stemness of glioma cells. However, identification of the signaling molecules and underlying mechanisms has not been completely elucidated. METHODS Human samples and glioma cell lines were cultured in vitro to determine the effects of adenovirus (ADV) infection by sphere formation, RT-qPCR, western blotting, FACS and immunofluorescence. For in vivo analysis, mouse intracranial tumor model was applied. Bioinformatics analysis, gene knockdown by siRNA, RT-qPCR and western blotting were applied for further mechanistic studies. RESULTS Infection of patient-derived glioma cells with ADV increases the formation of tumor spheres. ADV infection upregulated stem cell markers and in turn promoted the capacities of self-renewal and multi-lineage differentiation of the infected tumor spheres. These ADV infected tumor spheres had stronger potential to form xenograft tumors in immune-compromised mice. GSCs formation could be promoted by ADV infection via TLR9, because TLR9 was upregulated after ADV infection, and knockdown of TLR9 reduced ADV-induced GSCs. Consistently, MYD88, as well as total STAT3 and phosphorylated (p-)STAT3, were also upregulated in ADV-induced GSCs. Knockdown of MYD88 or pharmaceutical inhibition of STAT3 attenuated stemness of ADV-induced GSCs. Moreover, we found that ADV infection upregulated lncRNA NEAT1. Knockdown of NEAT1 impaired stemness of ADV-induced GSCs. Lastly, HMGB1, a damage associated molecular pattern (DAMP) that triggers TLR signaling, also upregulated stemness markers in glioma cells. CONCLUSION ADV, which has been developed as vectors for gene therapy and oncolytic virus, promotes the formation of GSCs via TLR9/NEAT1/STAT3 signaling. Video abstract.
Collapse
Affiliation(s)
- Jian Zang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China.,Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China
| | - Min-Hua Zheng
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiu-Li Cao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi-Zhe Zhang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China
| | - Yu-Fei Zhang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China
| | - Xiang-Yu Gao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China
| | - Yuan Cao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China
| | - Mei Shi
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China.
| | - Hua Han
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China. .,Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China.
| | - Liang Liang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China. .,Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #169, Xi'an, 710032, China.
| |
Collapse
|
30
|
Gao Y, Wang JW, Ren JY, Guo M, Guo CW, Ning SW, Yu S. Long noncoding RNAs in gastric cancer: From molecular dissection to clinical application. World J Gastroenterol 2020; 26:3401-3412. [PMID: 32655264 PMCID: PMC7327794 DOI: 10.3748/wjg.v26.i24.3401] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/24/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are important regulators of cell processes that are usually dysregulated in gastric cancer (GC). Based on their high specificity and ease of detection in tissues and body fluids, increasing attention has spurred the study of the roles of lncRNAs in GC patients. Thus, it is necessary to elucidate the molecular mechanisms and further explore the clinical applications of lncRNAs in GC. In this review, we summarize current knowledge to examine dysregulated lncRNAs in GC and their underlying molecular mechanisms and activities in GC, which involve microRNA sponging, mRNA stability, genetic variants, alternative splicing, transcription factor binding, and epigenetic modification. More significantly, the potential of lncRNAs as prognostic, circulating, and drug-resistant biomarkers for GC is also described. This review highlights the method of dissecting molecular mechanisms to explore the clinical application of lncRNAs in GC. Overall, this review offers assistance in using lncRNAs as novel candidates for molecular mechanisms and for the identification of revolutionary biomarkers for GC.
Collapse
Affiliation(s)
- Yue Gao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Jun-Wei Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Jia-Yi Ren
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Mian Guo
- Department of Neurosurgery, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Cheng-Wang Guo
- Department of Gastroenterology and Gastrosurgery, Gansu Wuwei Tumor Hospital, Wuwei 733000, Gansu Province, China
| | - Shang-Wei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Shan Yu
- Department of Pathology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| |
Collapse
|
31
|
STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects. BIOLOGY 2020; 9:biology9060126. [PMID: 32545648 PMCID: PMC7345582 DOI: 10.3390/biology9060126] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
Abstract
Molecular signaling pathways play a significant role in the regulation of biological mechanisms, and their abnormal expression can provide the conditions for cancer development. The signal transducer and activator of transcription 3 (STAT3) is a key member of the STAT proteins and its oncogene role in cancer has been shown. STAT3 is able to promote the proliferation and invasion of cancer cells and induces chemoresistance. Different downstream targets of STAT3 have been identified in cancer and it has also been shown that microRNA (miR), long non-coding RNA (lncRNA) and other molecular pathways are able to function as upstream mediators of STAT3 in cancer. In the present review, we focus on the role and regulation of STAT3 in gastric cancer (GC). miRs and lncRNAs are considered as potential upstream mediators of STAT3 and they are able to affect STAT3 expression in exerting their oncogene or onco-suppressor role in GC cells. Anti-tumor compounds suppress the STAT3 signaling pathway to restrict the proliferation and malignant behavior of GC cells. Other molecular pathways, such as sirtuin, stathmin and so on, can act as upstream mediators of STAT3 in GC. Notably, the components of the tumor microenvironment that are capable of targeting STAT3 in GC, such as fibroblasts and macrophages, are discussed in this review. Finally, we demonstrate that STAT3 can target oncogene factors to enhance the proliferation and metastasis of GC cells.
Collapse
|
32
|
Gao M, Liu L, Zhang D, Yang Y, Chang Z. Long Non-Coding RNA NEAT1 Serves as Sponge for miR-365a-3p to Promote Gastric Cancer Progression via Regulating ABCC4. Onco Targets Ther 2020; 13:3977-3985. [PMID: 32494153 PMCID: PMC7227816 DOI: 10.2147/ott.s245557] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Long non-coding RNA (lncRNA) was reported to be a crucial regulator in cancer. In this work, our purpose is to explore the biological roles of nuclear paraspeckle assembly transcript 1 (NEAT1) in gastric cancer (GC). Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect NEAT1 expression in GC cells and normal cells. GC cell behaviors after NEAT1 overexpression or downregulation were analyzed by Cell Counting Kit-8 assay, colony formation assay, wound-healing assay, and flow cytometry assay. Bioinformatic tools were used to analyze the significance of NEAT1 in GC. The involvement of microRNA-365a-3p (miR-365a-3p) and ATP-binding cassette subfamily C member 4 (ABCC4) in the biological roles of NEAT1 in GC progression was validated by luciferase activity reporter assay and rescue experiments. Results We found NEAT1 increased expression in both GC tissues and cells and correlated with poorer overall survival of cancer patients. We found NEAT1 overexpression promotes, while its knockdown inhibits GC cell proliferation, colony formation, invasion, and cell cycle progression in vitro. Mechanism analyses showed that NEAT1 serves as a ceRNA to upregulate ABCC4 expression via sponging miR-365a-3p. Conclusion In this study, we revealed a NEAT1/miR-365a-3p/ABCC4 triplet in GC progression, which may provide novel targeted therapy markers for GC.
Collapse
Affiliation(s)
- Ming Gao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Liying Liu
- Department of Medical Record, The First People's Hospital of Zhengzhou, Zhengzhou 450004, People's Republic of China
| | - Dianbao Zhang
- Department of Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang 471003, People's Republic of China
| | - Yudan Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Zhiwei Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| |
Collapse
|
33
|
Ye F, Xu R, Ge Y, Zheng Y, Liu X, Deng P, Xu X. LINC00963 Confers Oncogenic Properties in Glioma by Regulating the miR-506/BCAT1 Axis. Cancer Manag Res 2020; 12:2339-2351. [PMID: 32273770 PMCID: PMC7108718 DOI: 10.2147/cmar.s246332] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
Background Glioma is a prevalent disease of the central nervous system with a high incidence and mortality rate. Many long noncoding RNAs (lncRNAs) have been determined to be critical regulators of glioma oncogenesis. However, the function and mechanism of LINC00963 in glioma have not been fully elucidated. Methods The expression level of RNA was determined by qRT-PCR, and the protein level was determined by Western blot analysis. A luciferase activity assay was conducted to verify the interaction between miRNA and lncRNA or the target gene. The proliferation, cell cycle distribution, invasion, and migration were evaluated by MTT, EdU, flow cytometry, wound-healing and Transwell invasion assays, respectively. In vivo tumor growth was evaluated in a xenograft nude mouse model. Results We found that LINC00963 was upregulated in glioma cells and tissues and associated with the poor prognosis of patients with glioma. Ectopic expression of LINC00963 promoted cell proliferation, cell cycle progression, migration, and invasion in vitro and tumorigenesis in vivo. Mechanistically, the results of luciferase activity and RNA pulldown assays validated that LINC00963 could act as a molecular sponge of miR-506. Reciprocal repression was found between LINC00963 and miR-506. In addition, BCAT1 was identified as a target of miR-506, and both the mRNA and protein levels of BCAT1 were reduced by miR-506. In tumor tissues, the expression of BCAT1 was negatively and positively correlated with miR-506 and LINC00963 expression, respectively. The reintroduction of BCAT1 in glioma cells abolished the tumor suppressive function of miR-506 by promoting cell viability and motility. The upregulated LINC00963 and BCAT1 were associated with the aggressive phenotypes of tumors. Conclusion Our data revealed that LINC00963 confers oncogenic function in the progression of glioma and that the LINC00963/miR-506/BCAT1 axis may be a novel mechanism and therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Feng Ye
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Ronghua Xu
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Yuanhong Ge
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Yi Zheng
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Xiaowei Liu
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Pingfu Deng
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| | - Xuejun Xu
- Department of Neurosurgery, The Second People's Hospital of Chengdu, Chengdu, Sichuan 610021, People's Republic of China
| |
Collapse
|
34
|
Liu H, Li A, Sun Z, Zhang J, Xu H. Long non-coding RNA NEAT1 promotes colorectal cancer progression by regulating miR-205-5p/VEGFA axis. Hum Cell 2020; 33:386-396. [PMID: 32065361 DOI: 10.1007/s13577-019-00301-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 11/09/2019] [Indexed: 12/15/2022]
Abstract
Long non-coding RNAs (lncRNAs) play key roles in tumorigenesis. It has been reported that the lncRNA nuclear-enriched abundant transcript 1 (NEAT1) may act as an oncogenic regulator in several cancers. However, the biological mechanism of action of NEAT1, particularly the miRNA sponge role in colorectal cancer (CRC), has not been fully elucidated. In our study, the expression of NEAT1, miR-205-5p, and vascular endothelial growth factor A (VEGFA) in CRC cell lines were assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis. Cell proliferation was detected by Cell Counting Kit-8 (CCK-8) assay. Cell migration and invasion were examined by wound healing and transwell assays, respectively. RNA-binding protein immunoprecipitation (RIP), and dual-luciferase and RNA pull-down assays were conducted to determine the correlation between miR-205-5p and NEAT1 or VEGFA. VEGFA, matrix metalloproteinase (MMP)2, and MMP9 protein and mRNA expression were measured by western blotting and RT-qPCR analysis, respectively. Our results demonstrated high expression of NEAT1 and VEGFA and low expression of miR-205-5p in CRC cell lines. The RIP and dual-luciferase assays confirmed miR-205-5p as a target of NEAT1. In addition, VEGFA was identified as a direct target of miR-205-5p. Inhibition of NEAT1 or overexpression of miR-205-5p was able to repress VEGFA expression. Moreover, downregulation of NEAT1 and VEGFA inhibited cell proliferation, migration, and invasion. NEAT1 overexpression facilitated tumor growth by modulating miR-205-5p. Taken together, lncRNA NEAT1 was found to be upregulated in CRC cell lines, promoting CRC cell proliferation, migration, and invasion through regulating the miR-205-5p/VEGFA signaling pathway. These findings suggest that NEAT1 may be a promising biomarker in CRC diagnosis and treatment.
Collapse
Affiliation(s)
- Huijie Liu
- Department of Breast Oncology, Xiang Yang Central Hospital, The Affiliated Hospital of Hubei College of Arts and Sciences, 136 Jingzhou Street, Xiangyang, 441021, Hubei, China
| | - Aiyi Li
- Disinfection Supply Center, The Second People's Hospital of Lianyungang, Lianyungang, 222023, China
| | - Zhichao Sun
- Department of Pathology, The Second People's Hospital of Lianyungang, Lianyungang, 222023, China
| | - Jingyu Zhang
- Department of Tumor-Chemotherapy, The Second People's Hospital of Lianyungang, 161 Xingfu Road, Haizhou, Lianyungang, 222023, Jiangsu, China.
| | - Hua Xu
- Department of Breast Oncology, Xiang Yang Central Hospital, The Affiliated Hospital of Hubei College of Arts and Sciences, 136 Jingzhou Street, Xiangyang, 441021, Hubei, China.
| |
Collapse
|
35
|
Zhou X, Wang X, Zhou Y, Cheng L, Zhang Y, Zhang Y. Long Noncoding RNA NEAT1 Promotes Cell Proliferation And Invasion And Suppresses Apoptosis In Hepatocellular Carcinoma By Regulating miRNA-22-3p/akt2 In Vitro And In Vivo. Onco Targets Ther 2019; 12:8991-9004. [PMID: 31802908 PMCID: PMC6827517 DOI: 10.2147/ott.s224521] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/28/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most aggressive cancers that is associated with cirrhosis and other chronic liver diseases. Although remarkable progress has been made in past decades, it is still necessary to continue exploring the pathology and development of HCC. OBJECTIVE In this study, we elucidated the effect of long noncoding RNA (lncRNA) NEAT1 on HCC development and underlying mechanisms. METHODS Clinicopathological features of HCC patients were collected and the correlations with NEAT1 expression were assessed. To determine cell activities, CCK-8, flow cytometry, invasion assays, and TUNEL assays were performed. Real-time PCR, Western blot, and luciferase reporter assays were performed to investigate the related mechanism of HCC. RESULTS The results revealed that NEAT1 expression was associated with tumor size and differentiation where NEAT1 was upregulated in both HCC tissues and cell lines. Overexpression of NEAT1 promoted proliferation and invasion while inhibited apoptosis in HCC cells, which was opposite to the effect of NEAT1 knockdown. Also, AKT2 was increased in HCC tissues. Downregulation of AKT2 was associated with reduced cell proliferation and invasion while increased apoptosis, while overexpression of AKT2 exerted opposite roles. In addition, the expression of miRNA-22-3p displayed an inverse association with NEAT1. miRNA-22-3p mimic and inhibitor suppressed and promoted HCC development, respectively. The luciferase assay revealed that both NEAT1 and AKT2 were direct target genes of miRNA-22-3p. Furthermore, knockdown and overexpression of NEAT1 suppressed and promoted tumor growth in the HCC mouse model, which were abolished by the miRNA-22-3p inhibitor and mimic, respectively. CONCLUSION In conclusion, the results demonstrate that NEAT1 promotes the development of HCC, both in vitro and in vivo, through regulating miRNA-22-3p/AKT2, and provides insight into developing a new strategy for HCC treatment.
Collapse
Affiliation(s)
- Xichang Zhou
- Department of Intervention, Xuzhou Central Hospital, Xuzhou Medical University, XuZhou221009, People’s Republic of China
| | - Xiang Wang
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou Medical University, XuZhou221009, People’s Republic of China
| | - Yizhou Zhou
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou Medical University, XuZhou221009, People’s Republic of China
| | - Long Cheng
- Department of Intervention, Xuzhou Central Hospital, Xuzhou Medical University, XuZhou221009, People’s Republic of China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou Medical University, XuZhou221009, People’s Republic of China
| | - Yangmei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou Medical University, XuZhou221009, People’s Republic of China
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou215006, People’s Republic of China
| |
Collapse
|
36
|
Hu J, Ni G, Mao L, Xue X, Zhang J, Wu W, Zhang S, Zhao H, Ding L, Wang L. LINC00565 promotes proliferation and inhibits apoptosis of gastric cancer by targeting miR-665/AKT3 axis. Onco Targets Ther 2019; 12:7865-7875. [PMID: 31576144 PMCID: PMC6768015 DOI: 10.2147/ott.s189471] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 05/28/2019] [Indexed: 12/13/2022] Open
Abstract
Background Numerous studies have shown that long noncoding RNA (lncRNA) is involved in gastric cancer (GC). A relevant microarray containing gastric cancer-related lncRNAs was downloaded from The Cancer Genome Atlas database. Methods qRT-PCR was used to analyze LINC00565 and AKT3 expression in tumor tissues and cell lines. Proliferative, colony formation and apoptotic abilities of GC cells after transfection of sh-LINC00565 were determined by CCK-8, colony formation assay and flow cytometry, respectively. RIP was enrolled to detect the interaction between LINC00565, AKT3 and miR-665. Dual luciferase assay was used to confirm the relation between miR-665 and LINC00565 and AKT3. Results Expression level of LINC00565 in GC tissue was highly expressed in GC, which was negatively correlated to prognosis of GC patients. The results showed that knockdown of LINC00565 decreased proliferative and colony formation abilities, and induced apoptosis of GC cells. Pearson analysis showed that LINC00565 was positively correlated with AKT3. Besides, AKT3 was significantly up-regulated in GC. In addition, knockdown of LINC00565 down-regulated AKT3. In order to explore the mechanism, we found that miR-665 could bind to LINC00565 by bioinformatics. Dual-luciferase reporter gene assay and RIP assay both verified the binding relationship between miR-665 and AKT3. Finally, rescue experiments were carried out to explore whether AKT3 could reverse the anti-cancer effect of low-level LINC00565 on GC development. Conclusion In summary, the expression of LINC00565 is upregulated in GC. LINC00565 can be used as the sponge of miR-665 to up-regulate the expression of AKT3, thus promoting the progression of GC.
Collapse
Affiliation(s)
- Jianghong Hu
- Department of Gastroenterology, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Guohua Ni
- Department of Oncology, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Ling Mao
- Department of Oncology, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Xianglong Xue
- Department of Gastroenterology, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Jijie Zhang
- Department of Oncology, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Weixia Wu
- Department of Oncology, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Shaoru Zhang
- Central Laboratory, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Hong Zhao
- Central Laboratory, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Lifang Ding
- Department of Oncology, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| | - Lihui Wang
- Central Laboratory, Danyang People's Hospital of Jiangsu Province and Danyang Hospital Affiliated to Nantong University, Danyang, Jiangsu 212300, People's Republic of China
| |
Collapse
|
37
|
Wang J, Ding Y, Wu Y, Wang X. Identification of the complex regulatory relationships related to gastric cancer from lncRNA‐miRNA‐mRNA network. J Cell Biochem 2019; 121:876-887. [DOI: 10.1002/jcb.29332] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Jie Wang
- Laboratory of Media Design and Software Technology Jiangnan University Wuxi Jiangsu China
| | - Yanrui Ding
- Laboratory of Media Design and Software Technology Jiangnan University Wuxi Jiangsu China
- Key Laboratory of Industrial Biotechnology Jiangnan University Wuxi Jiangsu China
| | - Yanyan Wu
- Laboratory of Media Design and Software Technology Jiangnan University Wuxi Jiangsu China
| | - Xiaxia Wang
- Laboratory of Media Design and Software Technology Jiangnan University Wuxi Jiangsu China
| |
Collapse
|
38
|
Ding F, Lai J, Gao Y, Wang G, Shang J, Zhang D, Zheng S. NEAT1/miR-23a-3p/KLF3: a novel regulatory axis in melanoma cancer progression. Cancer Cell Int 2019; 19:217. [PMID: 31462890 PMCID: PMC6706883 DOI: 10.1186/s12935-019-0927-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Melanoma is an extremely aggressive malignant skin tumor with high mortality. Many types of long noncoding RNAs and microRNAs have been reported to be associated with the oncogenesis of melanoma. However, a novel lncRNA-NEAT has not been thoroughly investigated in melanoma cancer. The purposes of this study were to investigate the underlying molecular mechanism in a novel couple of lnc-NEAT1 and miR-23a-3p, as well as the function role of KLF3 in the regulation of melanoma cancer. METHODS 28 groups of tumor tissues and normal tissues were obtained from melanoma cancer patients. We performed a series of experiments and analysis, including RT-qPCR, western blots, CCK-8 assay, and migration/invasion assay, to investigate the expressions of NEAT1, miR-23a-5p and KLF3, cell viabilities, and tumor growth in vivo. RESULTS In this study, we observed that the expression of NEAT1 was significantly upregulated in melanoma tissues, which remarkedly promoted the cells' proliferation, cell migration, and invasion in melanoma cell lines. Besides, NEAT1 could directly bind to miR-23a-3p, which was found to reverse the effect caused by NEAT1. MiR-23a-3p was discovered to bind to 3'UTR of KLF3, which reduced KLF3 expression. In addition, the overexpression of KLF3 could lower the effects of miR-23a-3p caused on melanoma cancer cell development. CONCLUSION Our results demonstrated that NEAT1 could sponge miR-23a-3p and functions via the expression of KLF3. This axis of NEAT1/miR-23a-5p/KLF3 could together regulate melanoma cancer proliferation. This might provide a new therapeutic strategy for melanoma skin cancer.Trial registration HBTCM38574839, registered 12 October 2012.
Collapse
Affiliation(s)
- Fei Ding
- Department of Dermatology, Zhoukou Central Hospital, Zhoukou, 466000 Henan China
| | - Jindong Lai
- Department of Dermatology, Suining First People’s Hospital, Suining, 629000 Sichuan China
| | - Yang Gao
- Department of Dermatology, Affiliated Hospital of Hebei Academy of Traditional Chinese Medicine, Shijiazhuang, 050000 Hebei China
| | - Genhui Wang
- Department of Dermatology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, 050000 Hebei China
| | - Jingwen Shang
- Department of Dermatology, Zhoukou Central Hospital, Zhoukou, 466000 Henan China
| | - Daojun Zhang
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 400040 China
| | - Shumao Zheng
- Department of Dermatology, Hebei Academy of Chinese Medicine, Shijiazhuang, 050000 Hebei China
| |
Collapse
|
39
|
Pang Y, Wu J, Li X, Wang C, Wang M, Liu J, Yang G. NEAT1/miR‑124/STAT3 feedback loop promotes breast cancer progression. Int J Oncol 2019; 55:745-754. [PMID: 31322202 DOI: 10.3892/ijo.2019.4841] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/25/2019] [Indexed: 11/05/2022] Open
Abstract
The long non‑coding RNA nuclear enriched abundant transcript 1 (NEAT1) has important roles in the regulation of multiple cell functions, such as proliferation, apoptosis and migration. However, the mechanism by which NEAT1 regulates breast cancer progression is not well elucidated. In the present study, NEAT1 and microRNA‑124 (miR‑124) levels were detected by reverse transcription‑quantitative PCR in breast cancer tissues and cell lines. STAT3 protein levels were detected by western blot analysis. Cell proliferation and cell cycle distribution were determined using MTT and colony formation assays, and flow cytometry, respectively. The results demonstrated that NEAT1 and STAT3 expression levels were increased in breast cancer tissues compared with normal breast tissues, whereas miR‑124 expression was significantly decreased. Functional analyses revealed that NEAT1 promoted cell proliferation and cell cycle progression in breast cancer cells. Additionally, NEAT1 and STAT3 expression levels were negatively correlated with miR‑124 levels in breast cancer tissues. A direct interaction between miR‑124, and NEAT1 and STAT3, was predicted by bioinformatics analysis and confirmed using a luciferase activity assay. NEAT1 overexpression markedly increased STAT3 protein expression levels, and this effect was reversed by miR‑124 overexpression in breast cancer cells. Furthermore, miR‑124 overexpression partially attenuated the effects of NEAT1 on breast cancer cell proliferation and cell cycle progression. The inhibitory effects of miR‑124 overexpression on the proliferation rate and cell cycle progression were abolished by STAT3 overexpression. In turn, STAT3 silencing inhibited NEAT1 transcription in breast cancer cells. In summary, the present findings revealed that NEAT1 and STAT3 formed a feedback loop via sponging miR‑124 to promote breast cancer progression.
Collapse
Affiliation(s)
- Yamei Pang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jie Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiang Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Cuicui Wang
- Department of Hematology, Zoucheng People's Hospital, Zoucheng, Shandong 273500, P.R. China
| | - Meng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jian Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ganghua Yang
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
40
|
Zhou R, Wu Z, Deng X, Chen H. The long non-coding RNA OLC8 enhances gastric cancer by interaction with IL-11. J Clin Lab Anal 2019; 33:e22962. [PMID: 31273847 PMCID: PMC6805327 DOI: 10.1002/jcla.22962] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/02/2019] [Accepted: 06/05/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The gastric cancer (GC) represents a common malignancy especially in China. Long non-coding RNAs (lncRNAs) are critically involved in various types of cancer. However, the underlying mechanisms of OLC8 in gastric cancer are still largely unknown. METHODS The lncRNA profiling was used to identify novel lncRNAs associated with GC. The expression of OLC8 was quantified using qRT-PCR. Migration and viability assays were performed to evaluate the in vitro effects. Xenograft tumor models were conducted to investigate the in vivo oncogenic potential. RNA-seq was used to identify IL-11 as OLC8 binding partner. RESULTS In current study, we have identified a novel lncRNA termed OLC8. OLC8 was significantly overexpressed in gastric cancer specimens and cell lines. In vitro experiments showed that OLC8 facilitated migration and viability of MKN1 and AGS cells. As expected, in vivo experiments also confirmed an oncogenic role for OLC8. Mechanistic study indicated that OLC8 associated with IL-11 transcripts. The OLC8-IL-11 binding greatly impaired the degradation of IL-11 mRNAs. Not surprisingly, enhanced expression of IL-11 could increase STAT3 activation to favor gastric cancer development. CONCLUSIONS Our current research has identified OLC8 as a novel oncogenic lncRNA in IL-11/STAT3 signaling, and OLC8 may constitute a potential target for gastric cancer intervention.
Collapse
Affiliation(s)
- Rongjia Zhou
- Department of Gastroenterology, Panyu Central Hospital, Guangzhou, Guangdong, China
| | - Zhanbin Wu
- Department of Gastroenterology, Panyu Central Hospital, Guangzhou, Guangdong, China
| | - Xixiang Deng
- Department of Gastroenterology, Panyu Central Hospital, Guangzhou, Guangdong, China
| | - Haojun Chen
- Department of Gastroenterology, Panyu Central Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
41
|
Bure IV, Nemtsova MV, Zaletaev DV. Roles of E-cadherin and Noncoding RNAs in the Epithelial-mesenchymal Transition and Progression in Gastric Cancer. Int J Mol Sci 2019; 20:ijms20122870. [PMID: 31212809 PMCID: PMC6627057 DOI: 10.3390/ijms20122870] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/03/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023] Open
Abstract
The epithelial–mesenchymal transition (EMT) is thought to be at the root of invasive and metastatic cancer cell spreading. E-cadherin is an important player in this process, which forms the structures that establish and maintain cell–cell interactions. A partial or complete loss of E-cadherin expression in the EMT is presumably mediated by mechanisms that block the expression of E-cadherin regulators and involve the E-cadherin-associated transcription factors. The protein is involved in several oncogenic signaling pathways, such as the Wnt/β-catenin, Rho GTPase, and EGF/EGFR, whereby it plays a role in many tumors, including gastric cancer. Such noncoding transcripts as microRNAs and long noncoding RNAs—critical components of epigenetic control of gene expression in carcinogenesis—contribute to regulation of the E-cadherin function by acting directly or through numerous factors controlling transcription of its gene, and thus affecting not only cancer cell proliferation and metastasis, but also the EMT. This review focuses on the role of E-cadherin and the non-coding RNAs-mediated mechanisms of its expressional control in the EMT during stomach carcinogenesis.
Collapse
Affiliation(s)
- Irina V Bure
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia.
| | - Marina V Nemtsova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia.
- Research Centre for Medical Genetics, Moskvorechie st., 1, Moscow 115522, Russia.
| | - Dmitry V Zaletaev
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia.
- Research Centre for Medical Genetics, Moskvorechie st., 1, Moscow 115522, Russia.
| |
Collapse
|
42
|
The Association of HOTAIR with the Diagnosis and Prognosis of Gastric Cancer and Its Effect on the Proliferation of Gastric Cancer Cells. Can J Gastroenterol Hepatol 2019; 2019:3076345. [PMID: 31281803 PMCID: PMC6590613 DOI: 10.1155/2019/3076345] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/02/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are a group of noncoding RNA with the length of more than 200nt. They have been identified as important diagnostic and prognostic molecules for many cancers and play an important role in the development of cancers. However, their clinical value and roles in gastric cancer (GC) remain unclear. METHODS The expression levels of HOTAIR in 54 GC tissues and their matched adjacent nontumor tissues from GC patients and 24 normal mucosa or those with minimal gastritis as healthy controls were determined by qRT-PCR. The expression levels of HOTAIR in human GC cell lines and a normal gastric epithelium cell line were also assessed by qRT-PCR. The potential relationships between its level in GC tissues and the clinicopathological features were analyzed. Furthermore, a receiver operating characteristic (ROC) curve was constructed. Additionally, the correlation between this lncRNA and overall survival (OS) was analyzed. SiRNA transfection was used to silence the expression of HOTAIR in GC cells. And cell proliferation and cell cycle assays were employed to determine the effect of HOTAIR on GC cell growth. Western blot was performed for the detection of the P53, P21, and Bcl2 proteins. RESULTS The expression levels of HOTAIR were significantly upregulated in GC tissues and cell lines. Increased HOTAIR was associated with tumor differentiation, lymph node and distant metastasis, and clinical stage. Furthermore, the area under the ROC curve (AUC) was up to 0.8416 (95 % CI=0.7661 to 0.9170, P<0.0001). The sensitivity and specificity were 66.67 and 87.04%, respectively. The correlation between HOTAIR expression and overall survival (OS) was statistically significant. The hazard ratio was 2.681, and 95% CI of ratio was 1.370 to 5.248. In addition, knockdown of HOTAIR can inhibit GC cell growth and affect cell cycle distribution. And knockdown of HOTAIR could enhance the protein levels of P21 and P53. CONCLUSION The present study demonstrated that HOTAIR was highly expressed in GC tissues and may serve as a potential diagnostic and prognostic biomarker for GC. And HOTAIR promoted GC cell proliferation.
Collapse
|
43
|
Zhao G, Wang S, Liang X, Wang C, Peng B. Oncogenic role of long non-coding RNA SNHG12 in gastric cancer cells by targeting miR-16. Exp Ther Med 2019; 18:199-208. [PMID: 31258654 PMCID: PMC6566104 DOI: 10.3892/etm.2019.7526] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/24/2019] [Indexed: 12/15/2022] Open
Abstract
The long non-coding RNA small nucleolar RNA host gene 12 (SNHG12) has recently been reported to have an oncogenic role in gastric cancer (GC), but the molecular mechanisms remain largely elusive. In the present study, it was observed that SNHG12 was significantly upregulated in GC tissues and cell lines. High expression of SNHG12 was associated with GC progression and poor prognosis of patients. Knockdown of SNHG12 markedly inhibited the proliferation and migration of the BGC823 and HGC27 GC cell lines. MicroRNA (miR)-16 was identified as a target of SNHG12, and its expression was negatively regulated by SNHG12 in BGC823 and HGC27 cells. In addition, the expression of miR-16 was significantly decreased in GC tissues and cell lines, and inversely associated with the expression of SNHG12 in GC tissues. Furthermore, knockdown of miR-16 impaired the inhibitory effects on GC cell proliferation and migration induced by downregulation of SNHG12. In conclusion, the present study demonstrates that inhibition of SNHG12 suppresses GC cell proliferation and migration by modulation of miR-16 expression, and thus suggests that the SNHG12/miR-16 interaction may be used as a promising target for GC treatment.
Collapse
Affiliation(s)
- Guodong Zhao
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570208, P.R. China
| | - Suineng Wang
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570208, P.R. China
| | - Xianwen Liang
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570208, P.R. China
| | - Chunfang Wang
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570208, P.R. China
| | - Bo Peng
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, Hainan 570208, P.R. China
| |
Collapse
|
44
|
Fu X, Tian Y, Kuang W, Wen S, Guo W. Long non-coding RNA DLX6-AS1 silencing inhibits malignant phenotypes of gastric cancer cells. Exp Ther Med 2019; 17:4715-4722. [PMID: 31105791 DOI: 10.3892/etm.2019.7521] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 04/01/2019] [Indexed: 01/05/2023] Open
Abstract
It has been revealed that long non-coding RNAs (lncRNAs) serve a key role in various malignancies, including gastric cancer (GC). In the present study, the expression and function of lncRNA distal-less homeobox 6 antisense 1 (DLX6-AS1) in GC was investigated. The data revealed that the expression of DLX6-AS1 was significantly upregulated in GC tissues compared with adjacent paired noncancerous tissues. Furthermore, the expression of DLX6-AS1 was higher in advanced GC tissue samples (III/IV) compared with the expression in early-stage samples (I/II). Furthermore, the current study demonstrated that a high expression of DLX6-AS1 was significantly associated with advanced clinical stage, lymph node metastasis and distant metastasis. Compared with patients with a low DLX6-AS1 expression, DLX6-AS1 expression in patients with GC was associated with decreased survival. In vitro experimental data indicated that DLX6-AS1 was upregulated in GC cell lines and that the inhibition of DLX6-AS1 markedly reduced GC cell proliferation, colony formation, cell cycle progression, migration and invasion. Further investigation revealed that knockdown of DLX6-AS1 inhibited EMT in GC cells. In summary, the present study demonstrated that lncRNA DLX6-AS1 was upregulated and serves an oncogenic role in GC, indicating that DLX6-AS1 may be a novel therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Xiaodan Fu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410005, P.R. China
| | - Yifu Tian
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410005, P.R. China
| | - Weilu Kuang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410005, P.R. China
| | - Sailan Wen
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Wei Guo
- Department of Pathology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410002, P.R. China
| |
Collapse
|
45
|
Esfandi F, Taheri M, Kholghi Oskooei V, Ghafouri‐Fard S. Long noncoding RNAs expression in gastric cancer. J Cell Biochem 2019; 120:13802-13809. [DOI: 10.1002/jcb.28653] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/19/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Farbod Esfandi
- Department of Medical Genetics Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Vahid Kholghi Oskooei
- Department of Medical Genetics Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Soudeh Ghafouri‐Fard
- Department of Medical Genetics Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
46
|
Chen X, Zhang N. Downregulation of lncRNA NEAT1_2 radiosensitizes hepatocellular carcinoma cells through regulation of miR-101-3p/WEE1 axis. Cell Biol Int 2019; 43:44-55. [PMID: 30488993 DOI: 10.1002/cbin.11077] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/24/2018] [Indexed: 01/04/2023]
Abstract
Radioresistance is a major obstacle in hepatocellular carcinoma (HCC) radiotherapy. Aberrant expression of long non-coding RNA (lncRNA) has been postulated to be implicated in the development of HCC radioresistance. We investigated the role of lncRNA nuclear enriched abundant transcript 1_2 (NEAT1_2) in radioresistance of HCC and its molecular mechanism in this study. We found that NEAT1_2 and WEE1 were upregulated, and miR-101-3p was downregulated in HCC tissues, as well as HCC cell lines. Downregulation of WEE1 sensitized the radiosensitivity of HCC cells, as evidenced by decreased survival fractions of Huh7 and PLC5 cells and increased percentage of apoptotic cells. Also, knockdown of NEAT1_2 exerted a reinforcing effect on the radiosensitivity of HCC cells. In addition, WEE1 was confirmed as a direct target of miR-101-3p. Upregulation of miR-101-3p obviously decreased the mRNA and protein levels of WEE1 compared with that in the miR-NC group, while transfection of anta-miR-101-3p presented the opposite effects. In parallel, NEAT1_2 was identified to interact with miR-101-3p, and NEAT1_2 upregulated the expression of WEE1 in Huh7 cells through sponging miR-101-3p. Besides, the reinforcing effect of NEAT1_2 silencing could be attenuated by downregulation of miR-101-3p. To conclude, our results support the concept that downregulation of lncRNA NEAT1_2 radiosensitizes hepatocellular carcinoma cells through regulation of miR-101-3p/WEE1 axis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Nuclear Medicine, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330006, Jiangxi Province, P. R. China
| | - Nuobei Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330006, Jiangxi Province, P. R. China
| |
Collapse
|
47
|
Wen J, Wang H, Dong T, Gan P, Fang H, Wu S, Li J, Zhang Y, Du R, Zhu Q. STAT3-induced upregulation of lncRNA ABHD11-AS1 promotes tumour progression in papillary thyroid carcinoma by regulating miR-1301-3p/STAT3 axis and PI3K/AKT signalling pathway. Cell Prolif 2019; 52:e12569. [PMID: 30657221 PMCID: PMC6495520 DOI: 10.1111/cpr.12569] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/14/2018] [Accepted: 11/24/2018] [Indexed: 12/16/2022] Open
Abstract
Objectives Emerging evidences indicated the importance of long non‐coding RNAs (lncRNAs) in the tumorigenesis and deterioration of malignant tumours. To our knowledge, the study about lncRNAs in papillary thyroid carcinoma (PTC) is still inadequate. ABHD11‐AS1 was highly expressed in the PTC samples of The Cancer Genome Atlas database. This study focused on the biological function and mechanism of lncRNA ABHD11‐AS1 in PTC. Materials and methods qRT‐PCR analysis was used to examine the expression of ABHD11‐AS1 in PTC tissues and cell lines. The prognostic significance of ABHD11‐AS1 for the patients with PTC was analysed with Kaplan‐Meier analysis. The effects of ABHD11‐AS1 knockdown on the cell proliferation and metastasis were evaluated by in vitro functional assays and in vivo experiments. The molecular mechanism which contributed to the oncogenic role of ABHD11‐AS1 in PTC was explored by conducting mechanism experiments. Rescue assays were carried out for final demonstration. Results High expression of ABHD11‐AS1 predicted poor prognosis for patients with PTC and promoted cell proliferation and metastasis in vitro and in vivo. ABHD11‐AS1 was activated by the transcription factor STAT3. ABHD11‐AS1 positively regulated PI3K/AKT signalling pathway. ABHD11‐AS1 acted as a competitive endogenous (ce) RNA to upregulate STAT3 by sponging miR‐1301‐3p. Conclusions STAT3‐induced lncRNA ABHD11‐AS1 promoted PTC progression by regulating PI3K/AKT signalling pathway and miR‐1301‐3p/STAT3 axis.
Collapse
Affiliation(s)
- Juyi Wen
- Department of Radiation and Oncology, Navy General Hospital, Beijing, China
| | - Hongwei Wang
- Department of Neurosurgery, Navy General Hospital, Beijing, China
| | - Tingjun Dong
- TCM-Integrated Cancer Center of Southern Medical University, GuangZhou, Guangdong, China
| | - Panpan Gan
- AnHui Medical University, HeFei, Anhui, China
| | - Henghu Fang
- Department of Radiation and Oncology, Navy General Hospital, Beijing, China
| | - Sudong Wu
- Department of Radiation and Oncology, Navy General Hospital, Beijing, China
| | - Jingjiao Li
- Department of Radiation and Oncology, Navy General Hospital, Beijing, China
| | - Yuanyuan Zhang
- Department of Radiation and Oncology, Navy General Hospital, Beijing, China
| | - Rui Du
- Department of Radiation and Oncology, Navy General Hospital, Beijing, China
| | - Qi Zhu
- Department of Radiation and Oncology, Navy General Hospital, Beijing, China
| |
Collapse
|
48
|
Klec C, Prinz F, Pichler M. Involvement of the long noncoding RNA NEAT1 in carcinogenesis. Mol Oncol 2018; 13:46-60. [PMID: 30430751 PMCID: PMC6322192 DOI: 10.1002/1878-0261.12404] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/25/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022] Open
Abstract
Altered expression levels of the long noncoding RNA (lncRNA) nuclear‐enriched abundant transcript 1 (NEAT1) have been reported in different types of cancer. More than half of the NEAT1 studies in cancer have been published within the last 2 years. In this review, we discuss very recent developments and insights into NEAT1 contribution to carcinogenesis. Summarizing the literature, it becomes obvious that NEAT1 is a lncRNA highly de‐/upregulated in a variety of cancer entities, in which it primarily acts as a competing endogenous RNA (ceRNA) which sponges tumor‐suppressive microRNA (miRNA). The sponged miRNA lose their ability to degrade, silence, or hamper translation of their downstream—mostly oncogenic—target transcripts, ultimately promoting carcinogenesis. This role of NEAT1 function in tumorigenesis suggests it may be a prognostic biomarker as well as potential therapeutic target, pending the completion of further studies into the underlying mechanisms.
Collapse
Affiliation(s)
- Christiane Klec
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria
| | - Felix Prinz
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Austria.,Research Unit for Non-coding RNAs and Genome Editing, Medical University of Graz (MUG), Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
49
|
Yan H, Silva MA, Li H, Zhu L, Li P, Li X, Wang X, Gao J, Wang P, Zhang Z. Long noncoding RNA DQ786243 interacts with miR‐506 and promotes progression of ovarian cancer through targeting cAMP responsive element binding protein 1. J Cell Biochem 2018; 119:9764-9780. [PMID: 30105876 DOI: 10.1002/jcb.27295] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Huan Yan
- Department of Obstetrics and Gynecology The Third Affiliated Hospital of Zhengzhou University Zhengzhou China
- Department of Pathology and Laboratory Medicine University of Tennessee Health Science Center Memphis Tennessee
| | - Maria A. Silva
- Department of Pathology and Laboratory Medicine University of Tennessee Health Science Center Memphis Tennessee
- Department of Biotechnology Southwest Tennessee Community College Memphis Tennessee
| | - Hong Li
- Department of Obstetrics and Gynecology The Third Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Linlin Zhu
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Pengyun Li
- Department of Clinical Laboratory The Third Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Xia Li
- Department of Obstetrics and Gynecology The Third Affiliated Hospital of Zhengzhou University Zhengzhou China
- Department of Pathology and Laboratory Medicine University of Tennessee Health Science Center Memphis Tennessee
| | - Xiaofang Wang
- Department of Clinical Laboratory The Third Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Junjun Gao
- Department of Clinical Laboratory The Third Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Ping Wang
- Department of Clinical Laboratory The Third Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Zhan Zhang
- Department of Clinical Laboratory The Third Affiliated Hospital of Zhengzhou University Zhengzhou China
| |
Collapse
|
50
|
Chen B, Zhao Q, Guan L, Lv H, Bie L, Huang J, Chen XB. Long non-coding RNA NNT-AS1 sponges miR-424/E2F1 to promote the tumorigenesis and cell cycle progression of gastric cancer. J Cell Mol Med 2018; 22:4751-4759. [PMID: 30006956 PMCID: PMC6156444 DOI: 10.1111/jcmm.13726] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 05/08/2018] [Indexed: 12/20/2022] Open
Abstract
Long non‐coding RNAs (lncRNAs) have been illustrated to function as important regulators in carcinogenesis and cancer progression. However, the roles of lncRNA NNT‐AS1 in gastric cancer remain unclear. In the present study, we investigate the biological role of NNT‐AS1 in gastric cancer tumorigenesis. Results revealed that NNT‐AS1 expression level was significantly up‐regulated in GC tissue and cell lines compared with adjacent normal tissue and normal cell lines. The ectopic overexpression of NNT‐AS1 indicated the poor prognosis of GC patients. In vitro experiments validated that NNT‐AS1 knockdown suppressed the proliferation and invasion ability and induced the GC cell cycle progression arrest at G0/G1 phase. In vivo xenograft assay, NNT‐AS1 silencing decreased the tumour growth of GC cells. Bioinformatics online program predicted that miR‐424 targeted the 3′‐UTR of NNT‐AS1. Luciferase reporter assay, RNA‐immunoprecipitation (RIP) and RNA pull‐down assay validated the molecular binding within NNT‐AS1 and miR‐424, therefore jointly forming the RNA‐induced silencing complex (RISC). Moreover, E2F1 was verified to act as the target gene of NNT‐AS1/miR‐424, indicating the NNT‐AS1/miR‐424/E2F1 axis. In conclusion, our study indicates that NNT‐AS1 sponges miR‐424/E2F1 to facilitate GC tumorigenesis and cycle progress, revealing the oncogenic role of NNT‐AS1 for GC.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Gastrointestinal Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Cancer Hospital of Henan Province, Henan, China
| | - Qingfang Zhao
- Department of Gastrointestinal Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Cancer Hospital of Henan Province, Henan, China
| | - Lulu Guan
- Department of Gastrointestinal Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Cancer Hospital of Henan Province, Henan, China
| | - Huifang Lv
- Department of Gastrointestinal Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Cancer Hospital of Henan Province, Henan, China
| | - Liangyu Bie
- Department of Gastrointestinal Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Cancer Hospital of Henan Province, Henan, China
| | - Jinxi Huang
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Cancer Hospital of Henan Province, Henan, China
| | - Xiao-Bing Chen
- Department of Gastrointestinal Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Cancer Hospital of Henan Province, Henan, China
| |
Collapse
|