51
|
Chen Y, Yang L, Yin D, Feng X, Jie J, Yao D, Chen J. Role of Long Noncoding RNA Regulator of Reprogramming in Colon Cancer Progression via Epidermal Growth Factor Receptor Signaling. Technol Cancer Res Treat 2022; 21:15330338221114707. [PMID: 35946134 PMCID: PMC9373180 DOI: 10.1177/15330338221114707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/05/2022] [Accepted: 07/04/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Long intergenic noncoding RNA regulator of reprogramming (linc-ROR) is a novel long noncoding RNA that exhibits significant effects on cancer progression. This research presented that linc-ROR had a crucial part in promoting biological characteristics associated with worse prognosis in colon cancer. Method: Bioinformatics analysis was performed to predict signaling pathways related to linc-ROR. In addition, western blot, quantitative reverse transcription-polymerase chain reaction, RNA-pulldown, cell proliferation assays, colony formation assays, wound healing assays, and transwell assays were applied to detect the role and regulation of particular molecules. Results: Our results showed that the knockdown of linc-ROR reduced cell invasion, proliferative ability, and migration in colon cancer. Further evaluation verified that downregulating linc-ROR inhibited the activation of epidermal growth factor receptor (EGFR) signaling. In addition, cbl-b, a kind of E3 ubiquitin ligase that increases the degradation of EGFR, was found to be a potential linc-ROR target. Conclusions: Based on our findings, it was presented that linc-ROR served a role as a tumor-promoting factor via repressing the ubiquitination and degradation of EGFR signaling, which indicated that it could be a possible prognostic marker and therapeutic target for colon cancer.
Collapse
Affiliation(s)
- Ying Chen
- Department of Oncology, Nantong City No. 1 People's Hospital and
Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Li Yang
- Department of Oncology, Nantong City No. 1 People's Hospital and
Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Dian Yin
- Department of Oncology, Nantong City No. 1 People's Hospital and
Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiu Feng
- Department of Oncology, Nantong City No. 1 People's Hospital and
Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jing Jie
- Department of Oncology, Nantong City No. 1 People's Hospital and
Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - DengFu Yao
- Research Center of Clinical Medicine, The Affiliated Hospital of Nantong
University, Nantong, Jiangsu, China
| | - JianRong Chen
- Department of Oncology, Nantong City No. 1 People's Hospital and
Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
52
|
Han X, Mo J, Yang Y, Wang Y, Lu H. Crucial Roles of LncRNAs-Mediated Autophagy in Breast Cancer. Int J Med Sci 2022; 19:1082-1092. [PMID: 35813295 PMCID: PMC9254371 DOI: 10.7150/ijms.72621] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/09/2022] [Indexed: 11/05/2022] Open
Abstract
Breast cancer remains a worldwide public health issue. LncRNA and autophagy respectively or simultaneously, get involved in cellular and molecular processes of many different cancers, including genesis, metastasis, and deterioration of breast cancer and other malignant tumors. In this review, relevant studies have been summarized, and we have found that lncRNA-mediated autophagy in luminal A breast cancer, luminal B breast cancer, HER-2 positive breast cancer, and basal-like breast cancer may play an important role in mediating drug resistance sensitivity. LncRNAs target genes and affect different signaling pathways to a complex network, which attenuates the occurrence and development of primary breast cancer by coordinating autophagy. Abnormal expression of LncRNA may lead to dysregulation of autophagy, resulting in tumor genesis, expansion, and resistance to anti-tumor therapy. Targeting specific lncRNAs for autophagy regulation may conduct as a bio-marker for reliable diagnosis and prognosis treatment of breast cancer or provide a promising therapeutic strategy.
Collapse
Affiliation(s)
- Xinwei Han
- Tai Zhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China.,Cytotherapy Laboratory, Shenzhen People's Hospital, 1017, Dongmen North Road, Luohu, Shenzhen, 518020, China
| | - Jinggang Mo
- Tai Zhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China
| | - Yingmei Yang
- Tai Zhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China
| | - Yichao Wang
- Tai Zhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China
| | - Hongsheng Lu
- Tai Zhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China
| |
Collapse
|
53
|
Sideris N, Dama P, Bayraktar S, Stiff T, Castellano L. LncRNAs in breast cancer: a link to future approaches. Cancer Gene Ther 2022; 29:1866-1877. [PMID: 35788171 PMCID: PMC9750866 DOI: 10.1038/s41417-022-00487-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/06/2022] [Accepted: 05/27/2022] [Indexed: 01/25/2023]
Abstract
Breast cancer affects millions of women each year. Despite recent advances in targeted treatments breast cancer remains a significant threat to women's health. In recent years the development of high-throughput sequencing technologies has advanced the field of transcriptomics shedding light on the role of non-coding RNAs (ncRNAs), including long ncRNAs (lncRNAs), in human cellular function and disease. LncRNAs are classified as transcripts longer than 200nt with no coding potential. These transcripts constitute a diverse group of regulatory molecules essential to the modulation of crucial cellular processes, which dysregulation of leads to disease. LncRNAs exert their regulatory functions through their sequences and by forming complex secondary and tertiary structures that interact with other transcripts, chromatin and/or proteins. Numerous studies have provided evidence of the involvement of LncRNAs in tumor development and disease progression. They possess multiple characteristics that make them novel therapeutic and diagnostic targets. Indeed, the discovery of a novel mechanism by which lncRNAs associated with proteins can induce the formation of phase-separated droplets broadens our understanding of the spatiotemporal control of cellular processes and opens up developing a new treatment. Nevertheless, the role and the molecular mechanisms of many lncRNAs in the regulation of cellular processes and cancer still remain elusive. This is due to the absence of a thorough characterization of the regulatory role of their loci and the functional impact of their aberrations in cancer biology. Here, we present some of the latest advances concerning the role of LncRNAs in breast cancer.
Collapse
Affiliation(s)
- Nikolaos Sideris
- grid.12082.390000 0004 1936 7590Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG UK
| | - Paola Dama
- grid.12082.390000 0004 1936 7590Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG UK
| | - Salih Bayraktar
- grid.12082.390000 0004 1936 7590Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG UK
| | - Thomas Stiff
- grid.12082.390000 0004 1936 7590Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG UK
| | - Leandro Castellano
- grid.12082.390000 0004 1936 7590Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QG UK ,grid.7445.20000 0001 2113 8111Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ UK
| |
Collapse
|
54
|
Selem NA, Youness RA, Gad MZ. What is beyond LncRNAs in breast cancer: A special focus on colon cancer-associated Transcript-1 (CCAT-1). Noncoding RNA Res 2021; 6:174-186. [PMID: 34938928 PMCID: PMC8666458 DOI: 10.1016/j.ncrna.2021.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNAs (LncRNAs) play a vital role in the process of malignant transformation. In breast cancer (BC), lncRNAs field is currently under intensive investigations. Yet, the role of lncRNAs as promising diagnostic and/or prognostic biomarkers and as therapeutic target/tool among BC patients still needs a special focus from the biomedical scientists. In BC, triple negative breast cancer patients (TNBC) are the unlucky group as they are always represented with the worst prognosis and the highest mortality rates. For that reason, a special focus on TNBC and associated lncRNAs was addressed in this review. Colon cancer-associated transcript 1 (CCAT-1) is a newly discovered oncogenic lncRNA that has been emerged as a vital biomarker for diagnosis, prognosis and therapeutic interventions in multiple malignancies and showed differential expression among TNBC patients. In this review, the authors shed the light onto the general role of lncRNAs in BC and the specific functional activities, molecular mechanisms, competing endogenous ncRNA role of CCAT-1 in TNBC.
Collapse
Affiliation(s)
- Noha A. Selem
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Egypt
| | - Rana A. Youness
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Egypt
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, 11586, Cairo, Egypt
| | - Mohamed Z. Gad
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Egypt
| |
Collapse
|
55
|
Xiong G, Pan S, Jin J, Wang X, He R, Peng F, Li X, Wang M, Zheng J, Zhu F, Qin R. Long Noncoding Competing Endogenous RNA Networks in Pancreatic Cancer. Front Oncol 2021; 11:765216. [PMID: 34760707 PMCID: PMC8573238 DOI: 10.3389/fonc.2021.765216] [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: 08/26/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) is a highly malignant disease characterized by insidious onset, rapid progress, and poor therapeutic effects. The molecular mechanisms associated with PC initiation and progression are largely insufficient, hampering the exploitation of novel diagnostic biomarkers and development of efficient therapeutic strategies. Emerging evidence recently reveals that noncoding RNAs (ncRNAs), including long ncRNAs (lncRNAs) and microRNAs (miRNAs), extensively participate in PC pathogenesis. Specifically, lncRNAs can function as competing endogenous RNAs (ceRNAs), competitively sequestering miRNAs, therefore modulating the expression levels of their downstream target genes. Such complex lncRNA/miRNA/mRNA networks, namely, ceRNA networks, play crucial roles in the biological processes of PC by regulating cell growth and survival, epithelial-mesenchymal transition and metastasis, cancer stem cell maintenance, metabolism, autophagy, chemoresistance, and angiogenesis. In this review, the emerging knowledge on the lncRNA-associated ceRNA networks involved in PC initiation and progression will be summarized, and the potentials of the competitive crosstalk as diagnostic, prognostic, and therapeutic targets will be comprehensively discussed.
Collapse
Affiliation(s)
- Guangbing Xiong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shutao Pan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jikuan Jin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiang Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruizhi He
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Peng
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Li
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianwei Zheng
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Zhu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
56
|
Mao J, Zhang Q, Cong YS. Human endogenous retroviruses in development and disease. Comput Struct Biotechnol J 2021; 19:5978-5986. [PMID: 34849202 PMCID: PMC8604659 DOI: 10.1016/j.csbj.2021.10.037] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Human endogenous retroviruses (HERVs) represent ∼8% of human genome, deriving from exogenous retroviral infections of germ line cells occurred millions of years ago and being inherited by the offspring in a Mendelian fashion. Most of HERVs are nonprotein-coding because of the accumulation of mutations, insertions, deletions, and/or truncations. It has been long thought that HERVs were "junk DNA". However, it is now known that HERVs are involved in various biological processes through encoding proteins, acting as promoters/enhancers, or lncRNAs to affect human health and disease. In this review, we summarized recent findings about HERVs, with implications in embryonic development, pluripotency, cancer, aging, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jian Mao
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University School of Basic Medical Sciences, Hangzhou, China
| | - Qian Zhang
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University School of Basic Medical Sciences, Hangzhou, China
| | - Yu-Sheng Cong
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University School of Basic Medical Sciences, Hangzhou, China
| |
Collapse
|
57
|
Taheri Bajgan E, Gholipour A, Faghihi M, Mowla SJ, Malakootian M. Linc-ROR has a Potential ceRNA Activity for OCT4A by Sequestering miR-335-5p in the HEK293T Cell Line. Biochem Genet 2021; 60:1007-1024. [PMID: 34669056 DOI: 10.1007/s10528-021-10140-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/07/2021] [Indexed: 12/26/2022]
Abstract
Linc-ROR has a regulatory role in reprogramming, and the core stem cell transcription factors, OCT4, SOX2, and NANOG, regulate its expression. MicroRNAs (miRNAs) are also a critical constituent of pivotal posttranscriptional regulatory pathways. One of such interactions is a competing endogenous RNA interaction that connects small and long non-coding RNAs with coding transcripts. Here, we aimed to investigate the existence of such associations between OCT4A, Linc-ROR, hsa-miR-335-5p, and hsa-miR-544. Bioinformatic analysis was performed to evaluate the expression status of OCT4A, Linc-ROR, miR-335, and miR-544 throughout differentiation as well as in various differentiated cells. The complete lengths of OCT4A and Linc-ROR, and OCT4A 3'-UTR were cloned in the luciferase reporter vector, and the precursors of miR-335 and miR-544 were cloned in expression vectors. Following the overexpression of miR-335 and miR-544 in the 5637 cell line, the endogenous expression of OCT4A and Linc-ROR was evaluated. Afterward, the expression vectors of miRNAs and the reporter vectors of OCT4A/Linc-ROR were co-transfected in the HEK293T cell line. Via the Dual-Luciferase assay, the effect of the overexpression of miRNAs on their two possible targets (Linc-ROR and OCT4A) was investigated. The bioinformatic analysis demonstrated a relatively similar expression pattern for OCT4A and Linc-ROR, while miR-335 showed a different expression status. Both miR-335 and miR-544 inhibited the endogenous expression of OCT4A. The Dual-Luciferase assay likewise confirmed the inhibitory effect of miR-335 and miR-544 on OCT4A expression. In contrast, the miR-335 inhibitory effect was reversed in the presence of Linc-ROR, resulting in the upregulation of OCT4A. Such evidence suggests that Linc-ROR may compete with OCT4A to interact with miR-335.
Collapse
Affiliation(s)
- Elham Taheri Bajgan
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Akram Gholipour
- Department of Biology, Islamic Azad University Tehran Science and Research Branch, Tehran, Iran
| | - Mohammadali Faghihi
- Center for Therapeutic Innovation and Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, FL, 33136, USA
| | - Seyed Javad Mowla
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahshid Malakootian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
58
|
Tian JH, Liu SH, Yu CY, Wu LG, Wang LB. The Role of Non-Coding RNAs in Breast Cancer Drug Resistance. Front Oncol 2021; 11:702082. [PMID: 34589423 PMCID: PMC8473733 DOI: 10.3389/fonc.2021.702082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is one of the commonly occurring malignancies in females worldwide. Despite significant advances in therapeutics, the mortality and morbidity of BC still lead to low survival and poor prognosis due to the drug resistance. There are certain chemotherapeutic, endocrine, and target medicines often used for BC patients, including anthracyclines, taxanes, docetaxel, cisplatin, and fluorouracil. The drug resistance mechanisms of these medicines are complicated and have not been fully elucidated. It was reported that non-coding RNAs (ncRNAs), such as micro RNAs (miRNA), long-chain non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) performed key roles in regulating tumor development and mediating therapy resistance. However, the mechanism of these ncRNAs in BC chemotherapeutic, endocrine, and targeted drug resistance was different. This review aims to reveal the mechanism and potential functions of ncRNAs in BC drug resistance and to highlight the ncRNAs as a novel target for achieving improved treatment outcomes for BC patients.
Collapse
Affiliation(s)
- Jin-Hai Tian
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| | - Shi-Hai Liu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chuan-Yang Yu
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| | - Li-Gang Wu
- Department of Oncology, General Hospital of Ningxia Medical University, Yingchuan, China
| | - Li-Bin Wang
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
59
|
Jin H, Du W, Huang W, Yan J, Tang Q, Chen Y, Zou Z. lncRNA and breast cancer: Progress from identifying mechanisms to challenges and opportunities of clinical treatment. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:613-637. [PMID: 34589282 PMCID: PMC8463317 DOI: 10.1016/j.omtn.2021.08.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Breast cancer is a malignant tumor that has a high mortality rate and mostly occurs in women. Although significant progress has been made in the implementation of personalized treatment strategies for molecular subtypes in breast cancer, the therapeutic response is often not satisfactory. Studies have reported that long non-coding RNAs (lncRNAs) are abnormally expressed in breast cancer and closely related to the occurrence and development of breast cancer. In addition, the high tissue and cell-type specificity makes lncRNAs particularly attractive as diagnostic biomarkers, prognostic factors, and specific therapeutic targets. Therefore, an in-depth understanding of the regulatory mechanisms of lncRNAs in breast cancer is essential for developing new treatment strategies. In this review, we systematically elucidate the general characteristics, potential mechanisms, and targeted therapy of lncRNAs and discuss the emerging functions of lncRNAs in breast cancer. Additionally, we also highlight the advantages and challenges of using lncRNAs as biomarkers for diagnosis or therapeutic targets for drug resistance in breast cancer and present future perspectives in clinical practice.
Collapse
Affiliation(s)
- Huan Jin
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China.,MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Wei Du
- Department of Neurosurgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Wentao Huang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Jiajing Yan
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Qing Tang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yibing Chen
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
60
|
Ouban A. Expression of SALL4 stemness marker in laryngeal squamous cell carcinomas (LSCCs) and its clinical significance. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1972349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Abderrahman Ouban
- Department of Pathology, Alfaisal University College of Medicine, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
61
|
Lu C, Wei D, Zhang Y, Wang P, Zhang W. Long Non-Coding RNAs as Potential Diagnostic and Prognostic Biomarkers in Breast Cancer: Progress and Prospects. Front Oncol 2021; 11:710538. [PMID: 34527584 PMCID: PMC8436618 DOI: 10.3389/fonc.2021.710538] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
Breast cancer is the most common malignancy among women worldwide, excluding non-melanoma skin cancer. It is now well understood that breast cancer is a heterogeneous entity that exhibits distinctive histological and biological features, treatment responses and prognostic patterns. Therefore, the identification of novel ideal diagnostic and prognostic biomarkers is of utmost importance. Long non-coding RNAs (lncRNAs) are commonly defined as transcripts longer than 200 nucleotides that lack coding potential. Extensive research has shown that lncRNAs are involved in multiple human cancers, including breast cancer. LncRNAs with dysregulated expression can act as oncogenes or tumor-suppressor genes to regulate malignant transformation processes, such as proliferation, invasion, migration and drug resistance. Intriguingly, the expression profiles of lncRNAs tend to be highly cell-type-specific, tissue-specific, disease-specific or developmental stage-specific, which makes them suitable biomarkers for breast cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Cuicui Lu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Duncan Wei
- Department of Pharmacy, The First Affiliated Hospital of Medical College of Shantou University, Shantou, China
| | - Yahui Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Peng Wang
- Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Wen Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
62
|
Luo D, Yang L, Yu L, Chen Y, Huang Z, Liu H. Clinicopathological and prognostic significance of long non-coding RNA-ROR in cancer patients: A systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e26535. [PMID: 34232190 PMCID: PMC8270596 DOI: 10.1097/md.0000000000026535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 06/16/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Accumulating studies have focused on the clinicopathological and prognostic roles of large intergenic noncoding RNA regulator of reprogramming (lincRNA-ROR) in cancer patients. However, the results were controversial and unconvincing. Thus, we performed a meta-analysis to assess the associations between lincRNA-ROR expression and survival and clinicopathological characteristics of cancer patients. METHODS Hazard ratios for overall survival and disease-free survival with their 95% confidence intervals were used to evaluate the role of lincRNA-ROR expression in the prognosis of cancer patients. Risk ratios with their 95% confidence intervals were applied to assess the relationship between lincRNA-ROR expression and clinicopathological parameters. RESULTS A total of 18 articles with 1441 patients were enrolled. Our results indicated that high lincRNA-ROR expression was significant associated with tumor size, TNM stage, clinical stage, lymph metastasis, metastasis and vessel invasion of cancer patients. There were no correlations between high lincRNA-ROR expression and age, gender, infiltration depth, differentiation, serum CA19-9 and serum CEA of cancer patients. In addition, high lincRNA-ROR expression was associated with shorter Overall survival and disease-free survival on both univariate and multivariate analyses. Meanwhile, there were no obvious publication bias in our meta-analysis. CONCLUSIONS LincRNA-ROR expression was associated with the clinicopathological features and outcome of cancer patients, which suggested that lincRNA-ROR might serve as a potential biomarker for cancer prognosis. ETHICAL APPROVAL Since this study is on the basis of published articles, ethical approval and informed consent of patients are not required.
Collapse
Affiliation(s)
- Deqing Luo
- Department of Orthopaedic Surgery, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, Fujian Province, China
| | - Limin Yang
- Department of Pathology, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, Fujian Province, China
| | - Le Yu
- Department of Pathology, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, Fujian Province, China
| | - Yijin Chen
- Department of Pathology, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, Fujian Province, China
| | - Zunxian Huang
- Department of Orthopaedic Surgery, Quanzhou Orthopedic-Traumatological Hospital of Fujian Chinese Medical University, Quanzhou, Fujian Province, China
| | - Hui Liu
- Department of Orthopaedic Surgery, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, Fujian Province, China
| |
Collapse
|
63
|
Khojasteh Poor F, Keivan M, Ramazii M, Ghaedrahmati F, Anbiyaiee A, Panahandeh S, Khoshnam SE, Farzaneh M. Mini review: The FDA-approved prescription drugs that target the MAPK signaling pathway in women with breast cancer. Breast Dis 2021; 40:51-62. [PMID: 33896802 DOI: 10.3233/bd-201063] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer (BC) is the most common cancer and the prevalent type of malignancy among women. Multiple risk factors, including genetic changes, biological age, dense breast tissue, and obesity are associated with BC. The mitogen-activated protein kinases (MAPK) signaling pathway has a pivotal role in regulating biological functions such as cell proliferation, differentiation, apoptosis, and survival. It has become evident that the MAPK pathway is associated with tumorigenesis and may promote breast cancer development. The MAPK/RAS/RAF cascade is closely associated with breast cancer. RAS signaling can enhance BC cell growth and progression. B-Raf is an important kinase and a potent RAF isoform involved in breast tumor initiation and differentiation. Depending on the reasons for cancer, there are different strategies for treatment of women with BC. Till now, several FDA-approved treatments have been investigated that inhibit the MAPK pathway and reduce metastatic progression in breast cancer. The most common breast cancer drugs that regulate or inhibit the MAPK pathway may include Farnesyltransferase inhibitors (FTIs), Sorafenib, Vemurafenib, PLX8394, Dabrafenib, Ulixertinib, Simvastatin, Alisertib, and Teriflunomide. In this review, we will discuss the roles of the MAPK/RAS/RAF/MEK/ERK pathway in BC and summarize the FDA-approved prescription drugs that target the MAPK signaling pathway in women with BC.
Collapse
Affiliation(s)
- Fatemeh Khojasteh Poor
- Department of Obstetrics and Gynecology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mona Keivan
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Ramazii
- Kerman University of Medical Sciences, University of Kerman, Kerman, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Samira Panahandeh
- School of Health, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
64
|
Meng X, Zhang Z, Chen L, Wang X, Zhang Q, Liu S. Silencing of the Long Non-Coding RNA TTN-AS1 Attenuates the Malignant Progression of Osteosarcoma Cells by Regulating the miR-16-1-3p/TFAP4 Axis. Front Oncol 2021; 11:652835. [PMID: 34141611 PMCID: PMC8204018 DOI: 10.3389/fonc.2021.652835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/14/2021] [Indexed: 11/27/2022] Open
Abstract
Objectives Osteosarcoma (OS) is a type of bone malignancy. This study attempted to explore the effect of long non-coding RNA TTN-AS1 (TTN-AS1) on OS and to determine its molecular mechanisms. Methods The expression of TTN-AS1, microRNA-16-1-3p (miR-16-1-3p), and transcription factor activating enhancer binding protein 4 (TFAP4) in OS was assessed using qRT-PCR. The OS cell proliferation, migration, and invasion were measured using 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), wound-healing, and transwell assays. N-cadherin and MMP-2 protein level was determined with western blot. Interactions between TTN-AS1 and miR-16-1-3p or TFAP4 and miR-16-1-3p were confirmed using the dual-luciferase reporter assay. Additionally, an OS xenograft tumor model was constructed to assess the effect of TTN-AS1 on tumor growth. Results TTN-AS1 and TFAP4 expression was increased in OS, while miR-16-1-3p expression was decreased. TTN-AS1 silencing restrained OS cell proliferation, migration, invasion, N-cadherin and MMP-2 protein expression, and hindered tumor growth. MiR-16-1-3p overexpression retarded the malignant behavior of OS cells. TTN-AS1 played a carcinostatic role by down-regulating miR-16-1-3p in the OS cells. Moreover, miR-16-1-3p inhibition or TFAP4 elevation weakened the suppressive effect of TTN-AS1 silencing on OS cell tumor progression. Conclusion TTN-AS1 promoted the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of OS cells via mediating the miR-16-1-3p/TFAP4 axis. TTN-AS1 may be a critical target for improving OS.
Collapse
Affiliation(s)
- Xianghai Meng
- Trauma Center, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhenjun Zhang
- Trauma Center, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lin Chen
- Department of Burn Reconstructive Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xi Wang
- Department of Spine Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qingguo Zhang
- Department of Spine Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuheng Liu
- Department of Spine Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
65
|
Zampedri C, Martínez-Flores WA, Melendez-Zajgla J. The Use of Zebrafish Xenotransplant Assays to Analyze the Role of lncRNAs in Breast Cancer. Front Oncol 2021; 11:687594. [PMID: 34123857 PMCID: PMC8190406 DOI: 10.3389/fonc.2021.687594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 12/19/2022] Open
Abstract
Breast cancer represents a great challenge since it is the first cause of death by cancer in women worldwide. LncRNAs are a newly described class of non-coding RNAs that participate in cancer progression. Their use as cancer markers and possible therapeutic targets has recently gained strength. Animal xenotransplants allows for in vivo monitoring of disease development, molecular elucidation of pathogenesis and the design of new therapeutic strategies. Nevertheless, the cost and complexities of mice husbandry makes medium to high throughput assays difficult. Zebrafishes (Danio rerio) represent a novel model for these assays, given the ease with which xenotransplantation trials can be performed and the economic and experimental advantages it offers. In this review we propose the use of xenotransplants in zebrafish to study the role of breast cancer lncRNAs using low to medium high throughput assays.
Collapse
Affiliation(s)
- Cecilia Zampedri
- Functional Genomics Laboratories, Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| | | | - Jorge Melendez-Zajgla
- Functional Genomics Laboratories, Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| |
Collapse
|
66
|
Schwerdtfeger M, Desiderio V, Kobold S, Regad T, Zappavigna S, Caraglia M. Long non-coding RNAs in cancer stem cells. Transl Oncol 2021; 14:101134. [PMID: 34051619 PMCID: PMC8176362 DOI: 10.1016/j.tranon.2021.101134] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 04/29/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Long non coding RNAs are involved in the regulation of multiple cellular processes. Cancer stemness and escape from immunological anti-cancer mechanisms are important mechanisms of resistance to anti-cancer agents and are pivotal in controlling cancer development and metastases. Long non coding RNAs have deep effects on the immune-modulation and on the control of cancer stem cells. Several pathways involved in immunological escape and cancer stemness are modulated by long non coding RNAs. Targeting long non coding RNAs is a potential new strategy to control tumor development and metastases.
In recent years, it has been evidenced that the human transcriptome includes several types of non-coding RNAs (ncRNAs) that are mainly involved in the regulation of different cellular processes. Among ncRNAs, long-non-coding RNAs (lncRNAs) are defined as longer than 200 nucleotides and have been shown to be involved in several physiological and pathological events, including immune system regulation and cancer. Cancer stem cells (CSCs) are defined as a population of cancer cells that possess characteristics, such as resistance to standard treatments, cancer initiation, ability to undergo epithelial-to-mesenchymal transition, and the ability to invade, spread, and generate metastases. The cancer microenvironment, together with genetic and epigenetic factors, is fundamental for CSC maintenance and tumor growth and progression. Unsurprisingly, lncRNAs have been involved in both CSC biology and cancer progression, prognosis and recurrence. Here we review the most recent literature on IncRNAs involvement in CSC biology and function.
Collapse
Affiliation(s)
- Melanie Schwerdtfeger
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy; Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany, Member of the German Center for Lung Research (DZL); German Center for Translational Cancer Research (DKTK), Partner site Munich, Munich, Germany
| | - Tarik Regad
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Silvia Zappavigna
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
67
|
Hou P, Lin T, Meng S, Shi M, Chen F, Jiang T, Li Z, Li M, Chu S, Zheng J, Bai J. Long noncoding RNA SH3PXD2A-AS1 promotes colorectal cancer progression by regulating p53-mediated gene transcription. Int J Biol Sci 2021; 17:1979-1994. [PMID: 34131400 PMCID: PMC8193262 DOI: 10.7150/ijbs.58422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play key roles in various human cancers. We aimed to determine the key lncRNAs mediating colorectal cancer (CRC) progression. We identified some lncRNAs aberrantly expressed in CRC tissues by using lncRNA microarrays and demonstrated that SH3PXD2A-AS1 was one of the most highly overexpressed lncRNAs in CRC. We further aimed to explore the roles and possible molecular mechanisms of SH3PXD2A-AS1 in CRC. RNA ISH revealed that SH3PXD2A-AS1 was overexpressed in CRC compared with adjacent normal colon tissues and indicated poor prognosis in CRC. Functional analyses showed that SH3PXD2A-AS1 enhanced cell proliferation, angiogenesis, and metastasis. Mechanistically, SH3PXD2A-AS1 can directly interact with p53 protein and regulate p53-mediated gene transcription in CRC. We provided mechanistic insights into the regulation of SH3PXD2A-AS1 on p53-mediated gene transcription and suggested its potential as a new prognostic biomarker and target for the clinical management of CRC.
Collapse
Affiliation(s)
- Pingfu Hou
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tian Lin
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sen Meng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meilin Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fang Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tao Jiang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhongwei Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
68
|
Zhang Y, Wu W, Sun Q, Ye L, Zhou D, Wang W. linc‑ROR facilitates hepatocellular carcinoma resistance to doxorubicin by regulating TWIST1‑mediated epithelial‑mesenchymal transition. Mol Med Rep 2021; 23:340. [PMID: 33760121 PMCID: PMC7974311 DOI: 10.3892/mmr.2021.11979] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/22/2020] [Indexed: 01/28/2023] Open
Abstract
Long non‑coding RNAs are associated with cancer progression. Long intergenic non‑protein coding RNA (linc)‑regulator of reprogramming (ROR) enhances tumor development in hepatocellular carcinoma (HCC). However, the effect of chemoresistance and its underlying mechanisms in HCC are not completely understood. The present study aimed to identify the effect of ROR on sensitivity to doxorubicin (DOX) in HCC cells. In the present study, Cell Counting Kit‑8 and EdU assays were performed to assess cell viability and proliferation, respectively. In addition, E‑cadherin and vimentin protein expression levels were assessed via western blotting and immunofluorescence.The results of the present study demonstrated that HCC cells with high linc‑ROR expression levels were more resistant to DOX, and linc‑ROR knockdown increased HCC cell DOX sensitivity compared with the control group. The results indicated that compared with the NC siRNA group, linc‑ROR knockdown notably suppressed epithelial‑mesenchymal transition by downregulating twist family bHLH transcription factor 1 (TWIST1) expression. TWIST1 knockdown displayed a similar effect on HCC cell DOX sensitivity to linc‑ROR knockdown. Moreover, linc‑ROR knockdown‑induced HCC cell DOX sensitivity was inhibited by TWIST1 overexpression. The present study provided evidence that linc‑ROR promoted HCC resistance to DOX by inducing EMT via interacting with TWIST1. Therefore, linc‑ROR might serve as a therapeutic target for reducing DOX resistance in HCC.
Collapse
Affiliation(s)
- Yuanbiao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Weiding Wu
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Qiang Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Longyun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Dongkai Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
69
|
Liu L, Zhang X, Ding H, Liu X, Cao D, Liu Y, Liu J, Lin C, Zhang N, Wang G, Hou J, Huang B, Zhang Y, Lu J. Arginine and lysine methylation of MRPS23 promotes breast cancer metastasis through regulating OXPHOS. Oncogene 2021; 40:3548-3563. [PMID: 33927350 DOI: 10.1038/s41388-021-01785-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) is a vital regulator of tumor metastasis. However, the mechanisms governing OXPHOS to facilitate tumor metastasis remain unclear. In this study, we discovered that arginine 21(R21) and lysine 108 (K108) of mitochondrial ribosomal protein S23 (MRPS23) was methylated by the protein arginine methyltransferase 7 (PRMT7) and SET-domain-containing protein 6 (SETD6), respectively. R21 methylation accelerated the poly-ubiquitin-dependent degradation of MRPS23 to a low level. The MRPS23 degradation inhibited OXPHOS with elevated mtROS level, which consequently increased breast cancer cell invasion and metastasis. In contrast, K108 methylation increased MRPS23 stability, and K108 methylation coordinated with R21 methylation to maintain a low level of MRPS23, which was in favor of supporting breast cancer cell survival through regulating OXPHOS. Consistently, R21 and K108 methylation was correlated with malignant breast carcinoma. Significantly, our findings unveil a unique mechanism of controlling OXPHOS by arginine and lysine methylation and point to the impact of the PRMT7-SETD6-MRPS23 axis during breast cancer metastasis.
Collapse
Affiliation(s)
- Lingxia Liu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Xiliu Zhang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Huayi Ding
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Xin Liu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Donghui Cao
- Division of Clinical Research, First Hospital of Jilin University, Changchun, China
| | - Yingqi Liu
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Jiwei Liu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Cong Lin
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Na Zhang
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Guannan Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Jingyao Hou
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Baiqu Huang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Yu Zhang
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China.
| | - Jun Lu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China.
| |
Collapse
|
70
|
Interaction between LINC-ROR and Stemness State in Gastric Cancer Cells with Helicobacter pylori Infection. IRANIAN BIOMEDICAL JOURNAL 2021. [PMID: 33745265 PMCID: PMC8183384 DOI: 10.52547/ibj.25.3.157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
71
|
Wang MQ, Zhu WJ, Gao P. New insights into long non-coding RNAs in breast cancer: Biological functions and therapeutic prospects. Exp Mol Pathol 2021; 120:104640. [PMID: 33878314 DOI: 10.1016/j.yexmp.2021.104640] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/24/2021] [Accepted: 04/16/2021] [Indexed: 11/29/2022]
Abstract
Breast cancer (BC) has become one of the most common malignant tumors in the world, seriously endangering women's health and life. However, the underlying molecular mechanisms of BC remain unclear. Over the past decade, long non-coding RNAs (lncRNAs) were gradually discovered and appreciated to play pivotal regulatory role in the progression of BC. It has been demonstrated that lncRNAs are implicated in regulating plenty of biological phenomena including cell proliferation, apoptosis, invasion and metastasis by interacting with DNA, RNA or proteins. In addition to these, the function of lncRNAs in tumor resistance has increasingly attracted more attention. In this review, we summarized the emerging impact of lncRNAs on the occurrence and progression of human BC, specifically focusing on the functions and mechanisms of them, with the aim of exploring the potential value of lncRNAs as oncogenic drivers or tumor suppressors. Furthermore, the potential clinical application of lncRNAs as diagnostic biomarkers and therapeutic targets in BC was also discussed.
Collapse
Affiliation(s)
- Meng-Qi Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, CheeLoo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - Wen-Jie Zhu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, CheeLoo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China.
| | - Peng Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, CheeLoo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
72
|
Sanchez-Lopez JM, Mandujano-Tinoco EA, Garcia-Venzor A, Lozada-Rodriguez LF, Zampedri C, Uribe-Carvajal S, Melendez-Zajgla J, Maldonado V, Lizarraga F. Integrative analysis of transcriptional profile reveals LINC00052 as a suppressor of breast cancer cell migration. Cancer Biomark 2021; 30:365-379. [PMID: 33361583 DOI: 10.3233/cbm-200337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Long-non-coding RNAs, a class of transcripts with lengths > 200 nt, play key roles in tumour progression. Previous reports revealed that LINC00052 (long intergenic non-coding RNA 00052) was strongly downregulated during breast cancer multicellular spheroids formation and suggested a role in cell migration and oxidative metabolism. OBJECTIVE To examine the function of LINC00052 in MCF-7 breast cancer cells. METHODS Loss-of-function studies were performed to evaluate LINC00052 role on MCF-7 breast cancer cells. Microarray expression assays were performed to determine genes and cellular functions modified after LINC00052 knockdown. Next, the impact of LINC00052 depletion on MCF-7 cell respiration and migration was evaluated. RESULTS 1,081 genes were differentially expressed upon LINC00052 inhibition. Gene set enrichment analysis, Gene Ontology and Key Pathway Advisor analysis showed that signalling networks related to cell migration and oxidative phosphorylation were enriched. However, whereas LINC00052 knockdown in MCF-7 cells revealed marginal difference in oxygen consumption rates when compared with control cells, LINC00052 inhibition enhanced cell migration in vitro and in vivo, as observed using a Zebrafish embryo xenotransplant model. CONCLUSION Our data show that LINC00052 modulates MCF-7 cell migration. Genome-wide microarray experiments suggest that cancer cell migration is affected by LINC00052 through cytoskeleton modulation and Notch/β-catenin/NF-κB signalling pathways.
Collapse
Affiliation(s)
- Jose Manuel Sanchez-Lopez
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico.,Postgraduate Program in Biological Sciences, Faculty of Medicine, Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edna Ayerim Mandujano-Tinoco
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico.,Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación Luís Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Alfredo Garcia-Venzor
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | | - Cecilia Zampedri
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Salvador Uribe-Carvajal
- Department of Molecular Genetics, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jorge Melendez-Zajgla
- Functional Genomics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Vilma Maldonado
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Floria Lizarraga
- Epigenetics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
73
|
Aravindhan S, Younus LA, Hadi Lafta M, Markov A, Ivanovna Enina Y, Yushchenkо NA, Thangavelu L, Mostafavi SM, Pokrovskii MV, Ahmadi M. P53 long noncoding RNA regulatory network in cancer development. Cell Biol Int 2021; 45:1583-1598. [PMID: 33760334 DOI: 10.1002/cbin.11600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/08/2021] [Accepted: 03/21/2021] [Indexed: 12/12/2022]
Abstract
The protein p53 as a transcription factor with strong tumor-suppressive activities is known to trigger apoptosis via multiple pathways and is directly involved in the recognition of DNA damage and DNA repair processes. P53 alteration is now recognized as a common event in the pathogenesis of many types of human malignancies. Deregulation of tumor suppressor p53 pathways plays an important role in the activation of cell proliferation or inactivation of apoptotic cell death during carcinogenesis and tumor progression. Mounting evidence indicates that the p53 status of tumors and also the regulatory functions of p53 may be relevant to the long noncoding RNAs (lncRNA)-dependent gene regulation programs. Besides coding genes, lncRNAs that do not encode for proteins are induced or suppressed by p53 transcriptional response and thus control cancer progression. LncRNAs also have emerged as key regulators that impinge on the p53 signaling network orchestrating global gene-expression profile. Studies have suggested that aberrant expression of lncRNAs as a molecular-genomic signature may play important roles in cancer biology. Accordingly, it is important to elucidate the mechanisms by which the crosstalk between lncRNAs and p53 occurs in the development of numerous cancers. Here, we review how several classes of lncRNAs and p53 pathways are linked together in controlling the cell cycle and apoptosis in various cancer cells in both human and mouse model systems.
Collapse
Affiliation(s)
- Surendar Aravindhan
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, chennai, India
| | - Laith A Younus
- Department of Clinical Laboratory Sciences, Faculty of Pharmacy, Jabir Ibn Hayyan Medical University, Al Najaf Al Ashraf, Najaf, Iraq
| | | | | | - Yulianna Ivanovna Enina
- Department of Propaedeutics of Dental Diseases, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Natalya A Yushchenkо
- Department of Legal Disciplines, Kazan Federal University, Kazan, Russian Federation
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | | | - Michail V Pokrovskii
- Department of Pharmacology and Clinical Pharmacology, Institute of Medicine, Belgorod State National Research University, Belgorod, Russian Federation
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
74
|
Zhu L, Zhou D, Guo T, Chen W, Ding Y, Li W, Huang Y, Huang J, Pan X. LncRNA GAS5 inhibits Invasion and Migration of Lung Cancer through influencing EMT process. J Cancer 2021; 12:3291-3298. [PMID: 33976738 PMCID: PMC8100807 DOI: 10.7150/jca.56218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/04/2021] [Indexed: 11/05/2022] Open
Abstract
Background: Lung cancer is a malignant tumor in mammary gland epithelium with high morbidity and mortality among women worldwide. Long noncoding RNA GAS5 (GAS5) has been proved to be closely related with tumor progression. However, the influence of GAS5 on lung cancer and the specific mechanism remain unclear. Methods: Cell invasion, cell migration, cell apoptosis and cell cycle were investigated after transfection with pcDNA-GAS5 and sh-GAS5. Sizes of tumors were measured by establishing transplanted tumor model in vivo. E-cadherin and N-cadherin expressions were investigated. Results: Cell invasion and migration were inhibited markedly in GAS5 overexpressed cell line. Cell cycle results indicated that the percentage of S-phase cells was increased, and G2-phase was reduced in the GAS5 overexpression cell line. Tumor size was suppressed obviously after GAS5 overexpression treatment. GAS5 markedly inhibited the expression of E-cadherin and induced the expression of N-cadherin. GAS5 overexpression significantly inhibited lung cancer cell proliferation by increasing the E-cadherin and decreasing N-cadherin. Conclusions: These findings provide novel evidence that GAS5 can be viewed as an anti-lung cancer agent through affecting EMT pathway.
Collapse
Affiliation(s)
- Lihuan Zhu
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Dongsheng Zhou
- Department of Radiology, Fujian Provincial Hospital, Fuzhou, China
| | - Tianxing Guo
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Wenshu Chen
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Yun Ding
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Wujing Li
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Yangyun Huang
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Jianyuan Huang
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Xiaojie Pan
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| |
Collapse
|
75
|
Chen X, Li Z, Yong H, Wang W, Wang D, Chu S, Li M, Hou P, Zheng J, Bai J. Trim21-mediated HIF-1α degradation attenuates aerobic glycolysis to inhibit renal cancer tumorigenesis and metastasis. Cancer Lett 2021; 508:115-126. [PMID: 33794309 DOI: 10.1016/j.canlet.2021.03.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Tripartite motif-containing 21 (Trim21) is mainly involved in antiviral responses and autoimmune diseases. Although Trim21 has been reported to have a cancer-promoting or anticancer effect in various tumours, its role in renal cell cancer (RCC) remains to be elucidated. In this study, we demonstrate that Trim21 is downregulated in primary RCC tissues. Low Trim21 expression in RCC is correlated with poor clinicopathological characteristics and short overall survival. Moreover, we illustrate that Trim21 inhibits RCC cells glycolysis through the ubiquitination-mediated degradation of HIF-1α, which inhibits the proliferation, tumorigenesis, migration, and metastasis of RCC cells in vitro and in vivo. Our findings show that Trim21 may become a promising predictive biomarker for the prognosis of patients with RCC.
Collapse
Affiliation(s)
- Xintian Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhongwei Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongmei Yong
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huaian, Jiangsu, China
| | - Wenwen Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Diandian Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Pingfu Hou
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
76
|
Wang S, Yi M, Zhang X, Zhang T, Jiang L, Cao L, Zhou Y, Fang X. Effects of CDKN2B-AS1 on cellular proliferation, invasion and AKT3 expression are attenuated by miR-424-5p in a model of ovarian endometriosis. Reprod Biomed Online 2021; 42:1057-1066. [PMID: 33820740 DOI: 10.1016/j.rbmo.2021.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 01/17/2023]
Abstract
RESEARCH QUESTION Endometriosis is a common and complicated gynaecologic disease. Long non-coding RNA CDKN2B-AS1 plays a crucial role in the development and progression of several cancers. Whether CDKN2B-AS1 contributes to endometriosis, however, remains unknown. DESIGN Cellular proliferation, invasion and DNA synthesis abilities were assessed by CCK8, transwell and 5-ethynyle-2'-deoxyuridine assays. The expression of epithelial-mesenchymal transition markers and three isoforms of AKT was detected using Western blot. Real-time polymerase chain reaction was used to determine the relative expression levels of CDKN2B-AS1 and candidate miRNAs in ectopic, eutopic endometria and normal endometrial tissues. The relationship between CDKN2B-AS1 and miRNA was determined by luciferase reporter assays. RESULTS The relative expression level of CDKN2B-AS1 was up-regulated in eutopic and ectopic endometria. In endometrial stromal cells and Ishikawa cells, CDKN2B-AS1 overexpression promoted cellular proliferation and invasion, and increased the protein expression of vimentin but decreased the expression of E-cadherin. miR-424-5p was confirmed the target of CDKN2B-AS1 through bioinformatics tools and luciferase reporter assays. In addition, the enhanced effect of cellular phenotype of CDKN2B-AS1 overexpression was significantly attenuated by miR-424-5p overexpression. Furthermore, miR-424-5p was able to directly target AKT3 through luciferase reporter assay. Mechanistically, CDKN2B-AS1 acts as a ceRNA by sponging miR-424-5p and targets AKT3. CONCLUSIONS The cellular mechanism of CDKN2B-AS1 in endometriosis was confirmed; CDKN2B-AS1 may be a potential target for ovarian endometriosis therapy.
Collapse
Affiliation(s)
- Sixue Wang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha Hunan 410000, PR China
| | - Mingyu Yi
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha Hunan 410000, PR China
| | - Xinyue Zhang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha Hunan 410000, PR China
| | - Tingting Zhang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha Hunan 410000, PR China
| | - Li Jiang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha Hunan 410000, PR China
| | - Le Cao
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha Hunan 410000, PR China
| | - Yuxin Zhou
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha Hunan 410000, PR China
| | - Xiaoling Fang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha Hunan 410000, PR China.
| |
Collapse
|
77
|
Interplay between p53 and non-coding RNAs in the regulation of EMT in breast cancer. Cell Death Dis 2021; 12:17. [PMID: 33414456 PMCID: PMC7791039 DOI: 10.1038/s41419-020-03327-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
The epithelial-mesenchymal transition (EMT) plays a pivotal role in the differentiation of vertebrates and is critically important in tumorigenesis. Using this evolutionarily conserved mechanism, cancer cells become drug-resistant and acquire the ability to escape the cytotoxic effect of anti-cancer drugs. In addition, these cells gain invasive features and increased mobility thereby promoting metastases. In this respect, the process of EMT is critical for dissemination of solid tumors including breast cancer. It has been shown that miRNAs are instrumental for the regulation of EMT, where they play both positive and negative roles often as a part of a feed-back loop. Recent studies have highlighted a novel association of p53 and EMT where the mutation status of p53 is critically important for the outcome of this process. Interestingly, p53 has been shown to mediate its effects via the miRNA-dependent mechanism that targets master-regulators of EMT, such as Zeb1/2, Snail, Slug, and Twist1. This regulation often involves interactions of miRNAs with lncRNAs. In this review, we present a detailed overview of miRNA/lncRNA-dependent mechanisms that control interplay between p53 and master-regulators of EMT and their importance for breast cancer.
Collapse
|
78
|
Bo H, Zhang W, Zhong X, Chen J, Liu Y, Cheong KL, Fan P, Tang S. LINC00467, Driven by Copy Number Amplification and DNA Demethylation, Is Associated with Oxidative Lipid Metabolism and Immune Infiltration in Breast Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4586319. [PMID: 34956437 PMCID: PMC8695024 DOI: 10.1155/2021/4586319] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/30/2021] [Accepted: 11/10/2021] [Indexed: 02/05/2023]
Abstract
Breast cancer (BRCA) is a malignant tumor with a high incidence and poor prognosis in females. However, its pathogenesis remains unclear. In this study, based on bioinformatic analysis, we found that LINC00467 was highly expressed in BRCA and was associated with tumor metastasis and poor prognosis. The genomic and epigenetic analysis showed that LINC00467 may also be regulated by copy number amplification (CNA), chromatin openness, and DNA methylation. In vitro experiments showed that it could promote the proliferation, migration, and invasion of BRCA cells. Competitive endogenous RNA (ceRNA) regulatory network analysis and weighted gene coexpression network analysis (WGCNA) suggested that LINC00467 may play a role in signaling pathways of peroxisomal lipid metabolism, immunity, and others through microRNAs (miRNAs) targeting transforming growth factor beta 2 (TGFB2). In addition, copy number amplification and high expression of LINC00467 were associated with the low infiltration of CD8+ and CD4+ T cells. In conclusion, we found that LINC00467, driven by copy number amplification and DNA demethylation, may be a potential biomarker for the diagnosis and prognosis of BRCA and a tumor promoter acting as a potential therapeutic target for BRCA as well.
Collapse
Affiliation(s)
- Hao Bo
- Department of Plastic and Esthetic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
- Key Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning Commission, Institute of Reproductive and Stem Cell Engineering, Basic Medicine College, Central South University, Changsha, Hunan 410078, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410078, China
| | - Wancong Zhang
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515000, China
- Plastic Surgery Institute of Shantou University Medical College, Guangdong 515000, China
| | - Xiaoping Zhong
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515000, China
- Plastic Surgery Institute of Shantou University Medical College, Guangdong 515000, China
| | - Jiasheng Chen
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515000, China
- Plastic Surgery Institute of Shantou University Medical College, Guangdong 515000, China
| | - Yang Liu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, China
| | - Kit-Leong Cheong
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, China
| | - Pengju Fan
- Department of Plastic and Esthetic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Shijie Tang
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515000, China
- Plastic Surgery Institute of Shantou University Medical College, Guangdong 515000, China
| |
Collapse
|
79
|
miR-205 in Breast Cancer: State of the Art. Int J Mol Sci 2020; 22:ijms22010027. [PMID: 33375067 PMCID: PMC7792793 DOI: 10.3390/ijms22010027] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
Despite its controversial roles in different cancer types, miR-205 has been mainly described as an oncosuppressive microRNA (miRNA), with some contrasting results, in breast cancer. The role of miR-205 in the occurrence or progression of breast cancer has been extensively studied since the first evidence of its aberrant expression in tumor tissues versus normal counterparts. To date, it is known that the expression of miR-205 in the different subtypes of breast cancer is decreasing from the less aggressive subtype, estrogen receptor/progesterone receptor positive breast cancer, to the more aggressive, triple negative breast cancer, influencing metastasis capability, response to therapy and patient survival. In this review, we summarize the most important discoveries that have highlighted the functional role of this miRNA in breast cancer initiation and progression, in stemness maintenance, in the tumor microenvironment, its potential role as a biomarker and its relevance in normal breast physiology—the still open questions. Finally, emerging evidence reveals the role of some lncRNAs in breast cancer progression as sponges of miR-205. Here, we also reviewed the studies in this field.
Collapse
|
80
|
McCabe EM, Rasmussen TP. lncRNA involvement in cancer stem cell function and epithelial-mesenchymal transitions. Semin Cancer Biol 2020; 75:38-48. [PMID: 33346133 DOI: 10.1016/j.semcancer.2020.12.012] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/14/2020] [Accepted: 12/12/2020] [Indexed: 02/06/2023]
Abstract
Epithelial to mesenchymal transition (EMT) is a cellular process in which cells composing epithelial tissue lose requirements for physical contact with neighboring cells and acquire mesenchymal characteristics consisting of increased migratory and invasive behaviors. EMT is a fundamental process that is required for initial and later events during embryogenesis. Cancer stem cells (CSCs) possess multipotency sufficient for their differentiation into bulk tumor cells and also have the capacity to undergo EMT. When CSCs initiate EMT programs the resulting cancerous mesenchymal cells become invasive and this migratory behavior also poises them for metastatic activity. Long noncoding RNAs (lncRNAs) are functional RNA molecules that do not encode proteins, yet regulate the expression of protein-coding genes through recruitment or sequestration of gene-regulatory proteins and microRNAs. lncRNA exhibit tissue-specific patterns of gene expression during development and specific sets of lncRNAs are also involved in various cancer types. This review considers the interplay between lncRNAs and the biogenesis of CSCs. We also review function of lncRNAs in EMT in CSCs. In addition, we discuss the utility of lncRNAs as biomarkers of cancer progression, and their potential use as therapeutic targets for treatment of cancer.
Collapse
Affiliation(s)
- Evan M McCabe
- Department of Molecular and Cell Biology, University of Connecticut, USA
| | - Theodore P Rasmussen
- Department of Molecular and Cell Biology, University of Connecticut, USA; Department of Pharmaceutical Sciences, University of Connecticut, USA; University of Connecticut Stem Cell Institute, University of Connecticut, USA.
| |
Collapse
|
81
|
Zhang X, Powell K, Li L. Breast Cancer Stem Cells: Biomarkers, Identification and Isolation Methods, Regulating Mechanisms, Cellular Origin, and Beyond. Cancers (Basel) 2020; 12:E3765. [PMID: 33327542 PMCID: PMC7765014 DOI: 10.3390/cancers12123765] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/03/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Despite recent advances in diagnosis and treatment, breast cancer (BC) is still a major cause of cancer-related mortality in women. Breast cancer stem cells (BCSCs) are a small but significant subpopulation of heterogeneous breast cancer cells demonstrating strong self-renewal and proliferation properties. Accumulating evidence has proved that BCSCs are the driving force behind BC tumor initiation, progression, metastasis, drug resistance, and recurrence. As a heterogeneous disease, BC contains a full spectrum of different BC subtypes, and different subtypes of BC further exhibit distinct subtypes and proportions of BCSCs, which correspond to different treatment responses and disease-specific outcomes. This review summarized the current knowledge of BCSC biomarkers and their clinical relevance, the methods for the identification and isolation of BCSCs, and the mechanisms regulating BCSCs. We also discussed the cellular origin of BCSCs and the current advances in single-cell lineage tracing and transcriptomics and their potential in identifying the origin and lineage development of BCSCs.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH 43210, USA;
| | | | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH 43210, USA;
| |
Collapse
|
82
|
Unveiling the ups and downs of miR-205 in physiology and cancer: transcriptional and post-transcriptional mechanisms. Cell Death Dis 2020; 11:980. [PMID: 33191398 PMCID: PMC7667162 DOI: 10.1038/s41419-020-03192-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022]
Abstract
miR-205 plays important roles in the physiology of epithelia by regulating a variety of pathways that govern differentiation and morphogenesis. Its aberrant expression is frequently found in human cancers, where it was reported to act either as tumor-suppressor or oncogene depending on the specific tumor context and target genes. miR-205 expression and function in different cell types or processes are the result of the complex balance among transcription, processing and stability of the microRNA. In this review, we summarize the principal mechanisms that regulate miR-205 expression at the transcriptional and post-transcriptional level, with particular focus on the transcriptional relationship with its host gene. Elucidating the mechanisms and factors regulating miR-205 expression in different biological contexts represents a fundamental step for a better understanding of the contribution of such pivotal microRNA to epithelial cell function in physiology and disease, and for the development of modulation strategies for future application in cancer therapy.
Collapse
|
83
|
Maloney SM, Hoover CA, Morejon-Lasso LV, Prosperi JR. Mechanisms of Taxane Resistance. Cancers (Basel) 2020; 12:E3323. [PMID: 33182737 PMCID: PMC7697134 DOI: 10.3390/cancers12113323] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The taxane family of chemotherapy drugs has been used to treat a variety of mostly epithelial-derived tumors and remain the first-line treatment for some cancers. Despite the improved survival time and reduction of tumor size observed in some patients, many have no response to the drugs or develop resistance over time. Taxane resistance is multi-faceted and involves multiple pathways in proliferation, apoptosis, metabolism, and the transport of foreign substances. In this review, we dive deeper into hypothesized resistance mechanisms from research during the last decade, with a focus on the cancer types that use taxanes as first-line treatment but frequently develop resistance to them. Furthermore, we will discuss current clinical inhibitors and those yet to be approved that target key pathways or proteins and aim to reverse resistance in combination with taxanes or individually. Lastly, we will highlight taxane response biomarkers, specific genes with monitored expression and correlated with response to taxanes, mentioning those currently being used and those that should be adopted. The future directions of taxanes involve more personalized approaches to treatment by tailoring drug-inhibitor combinations or alternatives depending on levels of resistance biomarkers. We hope that this review will identify gaps in knowledge surrounding taxane resistance that future research or clinical trials can overcome.
Collapse
Affiliation(s)
- Sara M. Maloney
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
| | - Camden A. Hoover
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Lorena V. Morejon-Lasso
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Jenifer R. Prosperi
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| |
Collapse
|
84
|
Tang H, Han X, Feng Y, Hao Y. linc00968 inhibits the tumorigenesis and metastasis of lung adenocarcinoma via serving as a ceRNA against miR-9-5p and increasing CPEB3. Aging (Albany NY) 2020; 12:22582-22598. [PMID: 33159015 PMCID: PMC7746359 DOI: 10.18632/aging.103833] [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: 04/10/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Increasing evidence confirms that long noncoding RNAs (lncRNAs) exert vital functions in multiple biological process among malignant cancers. In the current study, we uncovered that linc00968 was downregulated in lung adenocarcinoma (LUAD). Furthermore, the low level of linc00968 was correlated with worse prognosis in patients with LUAD. Upregulation of linc00968 restrained the growth and metastatic phenotypes of LUAD cell in vitro and in vivo. Using bioinformation methods and luciferase reporter assay, we identified that linc00968 acted as a competing endogenous RNA (ceRNA) via sponging miR-9-5p to modulate the level of Cytoplasmic Polyadenylation Element Binding Protein 3 (CPEB3) in LUAD. In addition, LUAD cell migration, colony formation and epithelial-mesenchymal transition (EMT) process were suppressed by linc00968 while these aggressive traits were reversed by miR-142-5p or CPEB3 silencing. Altogether, our work disclosed that linc00968 played a critical role in LUAD and linc00968/miR-9-5p/CPEB3 regulatory axis might be a potential treatment target in LUAD.
Collapse
Affiliation(s)
- Huaping Tang
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Xiaolei Han
- Health Office, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Yan Feng
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Yueqin Hao
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China
| |
Collapse
|
85
|
Mi J, Han Y, Zhang J, Hao X, Xing M, Shang C. Long noncoding RNA LINC01410 promotes the tumorigenesis of neuroblastoma cells by sponging microRNA-506-3p and modulating WEE1. Cancer Med 2020; 9:8133-8143. [PMID: 32886453 PMCID: PMC7643657 DOI: 10.1002/cam4.3398] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/24/2020] [Accepted: 08/02/2020] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Neuroblastoma (NBL) is an extra-cranial solid tumor in children. This study was attempted to investigate the regulatory mechanism of long noncoding RNA LINC01410 (LINC01410) on NBL. METHODS The expression of LINC01410, miR-506-3p, and WEE1 in NBL was evaluated by quantitative real time polymerase chain reaction. The proliferation and colony formation ability of NBL cells were analyzed by MTT and colony formation assay. Flow cytometry assay was executed to measure the apoptosis and cell cycle. Dual-luciferase reporter assay was used to detect the targeted relationships among LINC01410, miR-506-3p, and WEE1. Additionally, the role of LINC01410 on NBL in vivo was evaluated according to a tumor xenograft model. RESULTS The expression of LINC01410 and WEE1 was enhanced and miR-506-3p was inhibited in NBL. LINC01410 knockdown attenuated the cell proliferation, colony formation ability, and inhibited tumor growth. Moreover, LINC01410 silencing facilitated the apoptosis and arrested the cell cycle. LINC01410 interacted with miR-506-3p to elevate the WEE1 expression in NBL. Additionally, miR-506-3p inhibition or WEE1 overexpression weakened the reduction effects of sh-LINC01410 on cell proliferation, colony formation ability, apoptosis, and cell cycle. CONCLUSIONS Knockdown of LINC01410 inhibited the development of NBL by miR-506-3p/WEE1 axis in vitro, which could serve as a potential therapeutic target for NBL therapy.
Collapse
Affiliation(s)
- Jie Mi
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdao CityShandong ProvinceChina
| | - Yang Han
- Department of Pediatric StomatologicalStomatological Hospital of Qingdao CityQingdao CityShandong ProvinceChina
| | - Jin Zhang
- Department of RespiratoryQingdao Women and Children's HospitalQingdao CityShandong ProvinceChina
| | - Xiwei Hao
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdao CityShandong ProvinceChina
| | - Maoqing Xing
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdao CityShandong ProvinceChina
| | - Cong Shang
- Department of Pediatric SurgeryThe Affiliated Hospital of Qingdao UniversityQingdao CityShandong ProvinceChina
| |
Collapse
|
86
|
Wang S, Chen W, Yu H, Song Z, Li Q, Shen X, Wu Y, Zhu L, Ma Q, Xing D. lncRNA ROR Promotes Gastric Cancer Drug Resistance. Cancer Control 2020; 27:1073274820904694. [PMID: 32019330 PMCID: PMC7003177 DOI: 10.1177/1073274820904694] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Gastric cancer is one of the most common malignant tumors worldwide, and for resectable tumors, the most effective treatment is surgery with chemotherapy in neoadjuvant or adjuvant setting. However, the majority of patients fail to achieve the ideal initial response and/or develop resistance to chemotherapy. It was reported that long noncoding RNA regulator of reprogramming (ROR) is highly associated with the progression of gastric cancer. However, the role ROR in multidrug resistance (MDR) remains unclear. METHODS The messenger RNA levels of 63 specimens of patients with gastric cancer were determined by real-time polymerase chain reaction analysis and were correlated with drug resistance and survival of patients. To determine the cellular functions of ROR, we generated gastric cancer MDR cells. The effect of ROR depletion on multidrug resistance-associated protein 1 (MRP1) expression and cell apoptosis were examined by immunoblotting analyses, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and flow cytometry. RESULTS We found that ROR expression levels are positively associated with increased MDR and poor prognosis of patients with gastric cancer. Regulator of reprogramming expression is increased in gastric cancer cells resistant to adriamycin (ADR) and vincristine (VCR). Depletion of ROR reduced MRP1 expression and increased apoptosis of drug-resistant gastric cancer cells in response to ADR and VCR treatment. CONCLUSIONS We demonstrated that ROR expression promotes MRP1 expression and MDR of gastric cancer cells and is correlated with increased MDR and poor prognosis of patients with gastric cancer. Our finding highlighted the potential of targeting ROR to improve the efficacy of chemotherapy.
Collapse
Affiliation(s)
- Shuai Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China.,Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong, China
| | - Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Hualong Yu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhengming Song
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Qian Li
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Xin Shen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Lei Zhu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Qingxia Ma
- Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Innovative Drug Research and transformation platform, Qingdao Cancer Institute, Qingdao, Shandong, China.,School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
87
|
Wang Y, Dong T, Wang P, Li S, Wu G, Zhou J, Wang Z. LINC00922 regulates epithelial-mesenchymal transition, invasive and migratory capacities in breast cancer through promoting NKD2 methylation. Cell Signal 2020; 77:109808. [PMID: 33045317 DOI: 10.1016/j.cellsig.2020.109808] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer ranks as the major reason for mortality in women populations, accounting for 23% of all cancer deaths. One in every three Asian women encounters the risk of this cancer in their lifetime. Long intergenic non-coding RNAs (lincRNAs) have emerged as tumor promoters and suppressors. The molecular mechanism of breast cancer remains elusive. Therefore, the current study aimed to explore the role lincRNA LINC00922 plays in the development of breast cancer. Breast cancer tissues and adjacent tissues were obtained from 109 patients with breast cancer. The RNA extraction and quantification and immunohistochemical staining characterized the high expression of LINC00922 and low expression of NKD2 in breast cancer tissues in comparison to its adjacent counterparts. Furthermore, the ectopic expression and knockdown experiments were conducted to figure out the in vivo and in vitro effects of LINC00922 on breast cancer progression. The ectopically expressed LINC00922 activated the Wnt signaling pathway, promoted epithelial-mesenchymal transition, cell proliferative, invasive and migratory capacities, tumor growth and metastasis. Additionally, the RIP and ChIP assay identified that LINC00922 recruited DNMT1, DNMT3A and DNMT3B proteins in the promoter region of NKD2 to promote NKD2 promoter methylation, thus reducing the NKD2 expression. Moreover, the Wnt signaling pathway was activated subsequent to NKD2 silencing, which was reversed by LINC00922 silencing. Lastly, the anti-oncogenic effects of LINC00922 inhibition was antagonized after NKD2 knocked down. The current study provides evidence that LINC00922 acts as a tumor promoter by promoting NKD2 methylation. Hopefully, it provides a novel potential gene target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yan Wang
- Department of Thyroid and Breast Surgery, The First People's Hospital of Lianyungang, Lianyungang 222061, PR China
| | - Tianfu Dong
- Department of Thyroid and Breast Surgery, The First People's Hospital of Lianyungang, Lianyungang 222061, PR China
| | - Peishun Wang
- Department of Thyroid and Breast Surgery, The First People's Hospital of Lianyungang, Lianyungang 222061, PR China
| | - Shuqin Li
- Department of Thyroid and Breast Surgery, The First People's Hospital of Lianyungang, Lianyungang 222061, PR China
| | - Geng Wu
- Department of Stomatology, The First People's Hospital of Lianyungang, Lianyungang 222061, PR China
| | - Jun Zhou
- Department of Thyroid and Breast Surgery, The First People's Hospital of Lianyungang, Lianyungang 222061, PR China
| | - Zhiqi Wang
- Department of Head and Neck Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China.
| |
Collapse
|
88
|
Mohebi M, Sattari A, Ghafouri-Fard S, Modarressi MH, Kholghi-Oskooei V, Taheri M. Expression profiling revealed up-regulation of three lncRNAs in breast cancer samples. Exp Mol Pathol 2020; 117:104544. [PMID: 32976818 DOI: 10.1016/j.yexmp.2020.104544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 09/12/2020] [Accepted: 09/19/2020] [Indexed: 01/31/2023]
Abstract
Long non-coding RNAs (lncRNAs) have been vastly investigated for their critical roles in the pathogenesis of breast cancer. Yet, the expression pattern and clinical significance of three lncRNAs namely CTBP1AS2, LINC-ROR and SPRY4-IT1 in breast cancer are not completely clarified. In the present investigation, we assessed expression of these lncRNAs in breast cancer tissues and paired non-cancerous specimens from the same patients using quantitative real time PCR. Notably, expression of CTBP1AS2, LINC-ROR and SPRY4-IT1 were upregulated in breast cancer tissues compared with non-cancerous tissues (ER = 17.62, P value<0.000; ER = 4.62, P value = 0.001 and ER = 3.47, P value = 0.005, respectively). Relative expression of LINC-ROR in tumoral tissues compared with non-tumoral tissues was associated with a history of hormone replacement therapy (P = 0.04). Expression levels of CTBP1AS2, LINC-ROR and SPRY4-IT1 were significantly correlated with each other in both tumoral and non-tumoral tissues. The strongest correlations were detected between CTBP1AS2/ LINC-ROR and CTBP1AS2/ SPRY4-IT1 pairs in non-tumoral tissues. CTBP1AS2 and SPRY4-IT1 had the best sensitivity (80%) and specificity (64%) values, respectively. Based on AUC values, the best diagnostic power belonged to CTBP1AS2. The current study potentiates CTBP1AS2, LINC-ROR and SPRY4-IT1 as putative contributors in the pathogenesis of breast cancer and suggests these lncRNAs as candidates for functional analysis in this kind of cancer.
Collapse
Affiliation(s)
- Mehdi Mohebi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Sattari
- 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.
| | | | - Vahid Kholghi-Oskooei
- Department of Laboratory Sciences, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Health Sciences Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
89
|
The roles of long noncoding RNAs in breast cancer metastasis. Cell Death Dis 2020; 11:749. [PMID: 32929060 PMCID: PMC7490374 DOI: 10.1038/s41419-020-02954-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most significant threat to female health. Breast cancer metastasis is the major cause of mortality in breast cancer patients. To fully unravel the molecular mechanisms that underlie the breast cancer cell metastasis is critical for developing strategies to improve survival and prognosis in breast cancer patients. Recent studies have revealed that the long noncoding RNAs (lncRNAs) are involved in breast cancer metastasis through a variety of molecule mechanisms, though the precise functional details of these lncRNAs are yet to be clarified. In the present review, we focus on the functions of lncRNAs in breast cancer invasion and metastasis, with particular emphasis on the functional properties, the regulatory factors, the therapeutic promise, as well as the future challenges in studying these lncRNA.
Collapse
|
90
|
Bi Z, Li Q, Dinglin X, Xu Y, You K, Hong H, Hu Q, Zhang W, Li C, Tan Y, Xie N, Ren W, Li C, Liu Y, Hu H, Xu X, Yao H. Nanoparticles (NPs)-Meditated LncRNA AFAP1-AS1 Silencing to Block Wnt/ β-Catenin Signaling Pathway for Synergistic Reversal of Radioresistance and Effective Cancer Radiotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000915. [PMID: 32999837 PMCID: PMC7509644 DOI: 10.1002/advs.202000915] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/17/2020] [Indexed: 05/28/2023]
Abstract
Resistance to radiotherapy is frequently encountered in clinic, leading to poor prognosis of cancer patients. Long noncoding RNAs (lncRNAs) play important roles in the development of radioresistance due to their functions in regulating the expression of target genes at both transcriptional and posttranscriptional levels. Exploring key lncRNAs and elucidating the mechanisms contributing to radioresistance are crucial for the development of effective strategies to reverse radioresistance, which however remains challenging. Here, actin filament-associated protein 1 antisense RNA1 (lncAFAP1-AS1) is identified as a key factor in inducing radioresistance of triple-negative breast cancer (TNBC) via activating the Wnt/β-catenin signaling pathway. Considering the generation of a high concentration of reduction agent glutathione (GSH) under radiation, a reduction-responsive nanoparticle (NP) platform is engineered for effective lncAFAP1-AS1 siRNA (siAFAP1-AS1) delivery. Systemic delivery of siAFAP1-AS1 with the reduction-responsive NPs can synergistically reverse radioresistance by silencing lncAFAP1-AS1 expression and scavenging intracellular GSH, leading to a dramatically enhanced radiotherapy effect in both xenograft and metastatic TNBC tumor models. The findings indicate that lncAFAP1-AS1 can be used to predict the outcome of TNBC radiotherapy and combination of systemic siAFAP1-AS1 delivery with radiotherapy can be applied for the treatment of recurrent TNBC patients.
Collapse
Affiliation(s)
- Zhuofei Bi
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
- RNA Biomedical InstituteSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Qingjian Li
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Xiaoxiao Dinglin
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
- Breast Tumor CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Ying Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
- RNA Biomedical InstituteSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
| | - Kaiyun You
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Huangming Hong
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Qian Hu
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Wei Zhang
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Chenchen Li
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Yujie Tan
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Ning Xie
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Wei Ren
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Chuping Li
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Yimin Liu
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Hai Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
- RNA Biomedical InstituteSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
- RNA Biomedical InstituteSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
- RNA Biomedical InstituteSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
- Department of OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
- Breast Tumor CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120P. R. China
| |
Collapse
|
91
|
Toraih EA, El-Wazir A, Ageeli EA, Hussein MH, Eltoukhy MM, Killackey MT, Kandil E, Fawzy MS. Unleash multifunctional role of long noncoding RNAs biomarker panel in breast cancer: a predictor classification model. Epigenomics 2020; 12:1215-1237. [PMID: 32812439 DOI: 10.2217/epi-2019-0291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aim: We aimed to explore the circulating expression profile of nine lncRNAs (MALAT1, HOTAIR, PVT1, H19, ROR, GAS5, ANRIL, BANCR, MIAT) in breast cancer (BC) patients relative to normal and risky individuals. Methods: Serum relative expressions of the specified long non-coding RNAs were quantified in 155 consecutive women, using quantitative reverse-transcription PCR. Random Forest (RF) and decision tree were also applied. Results: Significant MALAT1 upregulation and GAS5 downregulation could discriminate risky women from healthy controls. Overexpression of the other genes showed good diagnostic performances. Lower GAS5 levels were associated with metastasis and recurrence. RF model revealed a better performance when combining gene expression patterns with risk factors. Conclusion: The studied panel could be utilized as diagnostic/prognostic biomarkers in BC, providing promising epigenetic-based therapeutic targets.
Collapse
Affiliation(s)
- Eman A Toraih
- Department of Histology & Cell Biology, Genetics Unit, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt.,Department of Surgery, Tulane University, School of Medicine, New Orleans, LA 70112, USA
| | - Aya El-Wazir
- Department of Histology & Cell Biology, Genetics Unit, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Essam Al Ageeli
- Department of Clinical Biochemistry (Medical Genetics), Faculty of Medicine, Jazan University, Jazan 82911, Saudi Arabia
| | - Mohammad H Hussein
- Department of Surgery, Tulane University, School of Medicine, New Orleans, LA 70112, USA
| | - Mohamed M Eltoukhy
- College of Computing and Information Technology, Khulais, University of Jeddah, Jeddah 21959, Saudi Arabia.,Department of Computer Science, Faculty of Computers and Informatics, Suez Canal University, Ismailia 41522, Egypt
| | - Mary T Killackey
- Department of Surgery, Tulane University, School of Medicine, New Orleans, LA 70112, USA
| | - Emad Kandil
- Department of Surgery, Division of Endocrine & Oncologic Surgery, Tulane University, School of Medicine, New Orleans, LA 70112, USA
| | - Manal S Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt.,Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 1321, Saudi Arabia
| |
Collapse
|
92
|
Peng L, Jiang J, Tang B, Nice EC, Zhang YY, Xie N. Managing therapeutic resistance in breast cancer: from the lncRNAs perspective. Theranostics 2020; 10:10360-10377. [PMID: 32929354 PMCID: PMC7482807 DOI: 10.7150/thno.49922] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/04/2020] [Indexed: 02/05/2023] Open
Abstract
Breast cancer (BC) is the most common female malignancy and the second leading cause of cancer-related death worldwide. In spite of significant advances in clinical management, the mortality of BC continues to increase due to the frequent occurrence of treatment resistance. Intensive studies have been conducted to elucidate the molecular mechanisms underlying BC therapeutic resistance, including increased drug efflux, altered drug targets, activated bypass signaling pathways, maintenance of cancer stemness, and deregulated immune response. Emerging evidence suggests that long noncoding RNAs (lncRNAs) are intimately involved in BC therapy resistance through multiple modes of action. Therefore, an in-depth understanding of the implication of lncRNAs in resistance to clinical therapies may improve the clinical outcome of BC patients. Here, we highlight the role and underlying mechanisms of lncRNAs in regulating BC treatment resistance with an emphasis on lncRNAs-mediated resistance in different clinical scenarios, and discuss the potential of lncRNAs as novel biomarkers or therapeutic targets to improve BC therapy response.
Collapse
Affiliation(s)
- Liyuan Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Jingwen Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Bo Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Yuan-Yuan Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, P.R. China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| |
Collapse
|
93
|
Chen W, Yang J, Fang H, Li L, Sun J. Relevance Function of Linc-ROR in the Pathogenesis of Cancer. Front Cell Dev Biol 2020; 8:696. [PMID: 32850817 PMCID: PMC7432147 DOI: 10.3389/fcell.2020.00696] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/09/2020] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are the key components of non-coding RNAs (ncRNAs) with a length of 200 nucleotides. They are transcribed from the so-called “dark matter” of the genome. Increasing evidence have shown that lncRNAs play an important role in the pathophysiology of human diseases, particularly in the development and progression of tumors. Linc-ROR, as a new intergenic non-protein coding RNA, has been considered to be a pivotal regulatory factor that affects the occurrence and development of human tumors, including breast cancer (BC), colorectal cancer (CRC), pancreatic cancer (PC), hepatocellular carcinoma (HCC), and so on. Dysregulation of Linc-ROR has been closely related to advanced clinicopathological factors predicting a poor prognosis. Because linc-ROR can regulate cell proliferation, apoptosis, migration, and invasion, it can thus be used as a potential biomarker for patients with tumors and has potential clinical significance as a therapeutic target. This article reviewed the role of linc-ROR in the development of tumors, its related molecular mechanisms, and clinical values.
Collapse
Affiliation(s)
- Wenjian Chen
- Anhui Provincial Children's Hospital, Affiliated to Anhui Medical University, Hefei, China
| | - Junfa Yang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.,School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hui Fang
- Department of Pharmacology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Lei Li
- The Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun Sun
- Anhui Provincial Children's Hospital, Affiliated to Anhui Medical University, Hefei, China
| |
Collapse
|
94
|
Sun DE, Ye SY. Emerging Roles of Long Noncoding RNA Regulator of Reprogramming in Cancer Treatment. Cancer Manag Res 2020; 12:6103-6112. [PMID: 32765105 PMCID: PMC7382586 DOI: 10.2147/cmar.s253042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/09/2020] [Indexed: 12/17/2022] Open
Abstract
Despite numerous advances in cancer treatment, the global prevalence and cancer-related mortality remain high. Understanding tumor initiation and progression mechanisms are critical as it will lead to the development of interventions for improving the prognosis of cancer patients. The roles of long noncoding RNAs (lncRNAs) in cancer have attracted immense research interest. Growing evidence indicates that lncRNA regulator of reprogramming (linc-ROR), a well-studied RNA, regulates the progression of various cancers, such as lung cancer (LC), hepatocellular carcinoma (HCC), breast cancer (BC), colorectal cancer (CRC), pancreatic cancer (PC), papillary thyroid carcinoma (PTC), or esophageal squamous cell carcinoma (ESCC). linc-ROR promotes the proliferation, invasion, migration and chemoresistance of cancer cells. Herein, we reviewed current literature on the modulatory functions and mechanisms of linc-ROR in cancer development. We highlight new linc-ROR-related therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Di-Er Sun
- Clinical Laboratory, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China.,Clinical Laboratory, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China
| | - Shu-Yuan Ye
- Clinical Laboratory, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China.,Clinical Laboratory, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China
| |
Collapse
|
95
|
A Novel Long Non-Coding RNA-01488 Suppressed Metastasis and Tumorigenesis by Inducing miRNAs That Reduce Vimentin Expression and Ubiquitination of Cyclin E. Cells 2020; 9:cells9061504. [PMID: 32575745 PMCID: PMC7348830 DOI: 10.3390/cells9061504] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
Long intergenic non-coding RNAs (lincRNAs) play important roles in human cancer development, including cell differentiation, apoptosis, and tumor progression. However, their underlying mechanisms of action are largely unknown at present. In this study, we focused on a novel suppressor lincRNA that has the potential to inhibit progression of human hepatocellular carcinoma (HCC). Our experiments disclosed long intergenic non-protein coding RNA 1488 (LINC01488) as a key negative regulator of HCC. Clinically, patients with high LINC01488 expression displayed greater survival rates and better prognosis. In vitro and in vivo functional assays showed that LINC01488 overexpression leads to significant suppression of cell proliferation and metastasis in HCC. Furthermore, LINC01488 bound to cyclin E to induce its ubiquitination and reduced expression of vimentin mediated by both miR-124-3p/miR-138-5p. Our results collectively indicate that LINC01488 acts as a tumor suppressor that inhibits metastasis and tumorigenesis in HCC via the miR-124-3p/miR-138-5p/vimentin axis. Furthermore, LINC01488 interacts with and degrades cyclin E, which contributes to its anti-tumorigenic activity. In view of these findings, we propose that enhancement of LINC01488 expression could be effective as a potential therapeutic strategy for HCC.
Collapse
|
96
|
Nguyen TM, Alchalabi S, Oluwatoyosi A, Ropri AS, Herschkowitz JI, Rosen JM. New twists on long noncoding RNAs: from mobile elements to motile cancer cells. RNA Biol 2020; 17:1535-1549. [PMID: 32522127 DOI: 10.1080/15476286.2020.1760535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The purpose of this review is to highlight several areas of lncRNA biology and cancer that we hope will provide some new insights for future research. These include the relationship of lncRNAs and the epithelial to mesenchymal transition (EMT) with a focus on transcriptional and alternative splicing mechanisms and mRNA stability through miRNAs. In addition, we highlight the potential role of enhancer e-lncRNAs, the importance of transposable elements in lncRNA biology, and finally the emerging area of using antisense oligonucleotides (ASOs) and small molecules to target lncRNAs and their therapeutic implications.
Collapse
Affiliation(s)
- Tuan M Nguyen
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School , Boston, MA, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA, USA
| | - Sumayya Alchalabi
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, SUNY , Rensselaer, NY, USA
| | - Adewunmi Oluwatoyosi
- Department of Molecular & Cellular Biology, Baylor College of Medicine , Houston, TX, USA
| | - Ali S Ropri
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, SUNY , Rensselaer, NY, USA
| | - Jason I Herschkowitz
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, SUNY , Rensselaer, NY, USA
| | - Jeffrey M Rosen
- Department of Molecular & Cellular Biology, Baylor College of Medicine , Houston, TX, USA
| |
Collapse
|
97
|
Chen X, Zhang Z, Ma Y, Su H, Xie P, Ran J. LINC02381 Promoted Cell Viability and Migration via Targeting miR-133b in Cervical Cancer Cells. Cancer Manag Res 2020; 12:3971-3979. [PMID: 32547232 PMCID: PMC7261661 DOI: 10.2147/cmar.s237285] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/19/2020] [Indexed: 11/23/2022] Open
Abstract
Background It has been proved that lncRNAs could function as CeRNA for miRNAs in tumor growth and metastasis for cervical cancer. This paper aims to identify the role of LINC02381 in cervical cancer cells. Materials and Methods RT-qPCR was utilized to measure the expression levels of LINC02381 in cervical cancer tissues and cells. MTT, colony formation assay, transwell assay, RT-qPCR, and Western blotting were performed to investigate the roles of LINC02381 in cervical cancer cells. RegRNA 2.0 was used to predict the miRNA-binding sites of LINC02381. Luciferase reporter assay and RT-qPCR were employed to confirm the sponging effect between miR-133b and LINC02381. Results This study showed that LINC02381 was up-regulated in cervical cancer cells and acted as an oncogene in the development of cervical cancer. LINC02381 promoted cell viability and metastasis via sponging miR-133b. Moreover, miR-133b could target its downstream mediator of RhoA and inhibit its expression. Conclusion Overall, our results indicated that LINC02381 functions as an oncogene in cervical cancer and could serve as a novel target for cervical cancer therapies in the future.
Collapse
Affiliation(s)
- Xiaohua Chen
- Department Of Radiation Therapy, First Hospital Of Lanzhou University, Lanzhou City, Gansu Province 730000, People's Republic of China
| | - Zhuxiang Zhang
- Department Of Radiation Therapy, First Hospital Of Lanzhou University, Lanzhou City, Gansu Province 730000, People's Republic of China
| | - Yan Ma
- Department Of Radiation Therapy, First Hospital Of Lanzhou University, Lanzhou City, Gansu Province 730000, People's Republic of China
| | - Hongxin Su
- Department Of Radiation Therapy, First Hospital Of Lanzhou University, Lanzhou City, Gansu Province 730000, People's Republic of China
| | - Peng Xie
- Department Of Radiation Therapy, First Hospital Of Lanzhou University, Lanzhou City, Gansu Province 730000, People's Republic of China
| | - Juntao Ran
- Department Of Radiation Therapy, First Hospital Of Lanzhou University, Lanzhou City, Gansu Province 730000, People's Republic of China
| |
Collapse
|
98
|
Zhou Q, Guo J, Huang W, Yu X, Xu C, Long X. Linc-ROR promotes the progression of breast cancer and decreases the sensitivity to rapamycin through miR-194-3p targeting MECP2. Mol Oncol 2020; 14:2231-2250. [PMID: 32335998 PMCID: PMC7463371 DOI: 10.1002/1878-0261.12700] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/16/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
linc‐ROR is reported to be a potential biomarker of breast cancer, but the detailed mechanism of linc‐ROR‐mediated breast cancer regulation has not been fully studied. We aimed to explore how linc‐ROR affects proliferation, metastasis, and drug sensitivity in breast cancer. Cell lines in which linc‐ROR was overexpressed or knocked down were constructed, and the cell proliferation, colony formation, cell migration, and invasion abilities of these lines were explored. A CCK‐8 assay was performed to determine the sensitivity of the breast cancer cells to rapamycin. Next‐generation sequencing was conducted to explore the detailed regulatory mechanism of linc‐ROR; differentially expressed RNAs in the linc‐ROR‐overexpressing cell line compared with the negative control were screened out, and their target genes were chosen to perform Gene Ontology analysis, Kyoto Encyclopedia of Genes and Genomes analysis, protein–protein interaction network analysis, and competing endogenous RNA (ceRNA) network analysis. The ceRNA mechanism of linc‐ROR for miR‐194‐3p, which targets MECP2, was determined through dual‐luciferase reporter assay, RT–qPCR, western blot, and rescue experiments. Finally, we found that linc‐ROR was upregulated in breast tumor tissues. linc‐ROR promoted the cell proliferation, colony formation, cell migration, and invasion of breast cancer and decreased the sensitivity of breast cancer cells to rapamycin. The overexpression of linc‐ROR triggered changes in the whole transcriptome of breast cancer cells, and a total of 85 lncRNAs, 414 microRNAs, 490 mRNAs, and 92 circRNAs were differentially expressed in the linc‐ROR‐overexpressing cell line compared with the negative control. Through a series of bioinformatic analyses, the ‘linc‐ROR/miR‐194‐3p/MECP2’ ceRNA regulatory axis was confirmed to be involved in the linc‐ROR‐mediated progression and drug sensitivity of breast cancer. In conclusion, linc‐ROR serves as an onco‐lncRNA in breast cancer and promotes the survival of breast cancer cells during rapamycin treatment by functioning as a ceRNA sponge for miR‐194‐3p, which targets MECP2.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Juan Guo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Wenjie Huang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Xiaosi Yu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Chen Xu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| | - Xinghua Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, China
| |
Collapse
|
99
|
Long Noncoding RNA HOTTIP Serves as an Independent Predictive Biomarker for the Prognosis of Patients with Clear Cell Renal Cell Carcinoma. Int J Genomics 2020; 2020:4301634. [PMID: 32566641 PMCID: PMC7255047 DOI: 10.1155/2020/4301634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
Several studies have indicated that HOXA transcript at the distal tip (HOTTIP) play important roles in the tumorigenesis and development of various cancers. We aim to investigate the expression and prognostic value of HOTTIP in clear cell renal cell carcinoma (ccRCC). A systematic review of PubMed, Embase, Medline, and Web of Science databases was performed to select eligible literatures relevant to the correlation between HOTTIP expression and clinical outcome of different cancers. The association between the HOTTIP level and overall survival (OS), lymph node metastasis (LNM), or clinical stage was subsequently analyzed. Survival analyses were performed in a large cohort of more than 500 patients with ccRCC from The Cancer Genome Atlas (TCGA) using bioinformatic methods. Seventeen studies with a total of 1594 patients with thirteen kinds of carcinomas were included in this analysis. The result showed that high HOTTIP expression could predict worse outcome in cancer patients, with the pooled hazard ratio (HR) of 2.34 (95% confidence interval (CI) 1.96–2.79, p < 0.0001). The result also showed that elevated HOTTIP expression was correlated with more LNM (OR = 2.61, 95% CI 1.91-3.58, p < 0.0001) and advanced clinical stage (OR = 3.57, 95% CI 2.58-4.93, p < 0.0001). We further validated that ccRCC patients with higher HOTTIP expression tend to have unsatisfactory outcomes both in the entire TCGA dataset and different clinical stratums, like age, grade, and stage. The tumor of those patients was associated with a larger size, easier to metastasis, advanced clinical stage, and a higher pathological grade. These findings suggested that increased HOTTIP expression might act as a novel prognostic marker for ccRCC patients.
Collapse
|
100
|
Kim H, Lee S, Shin E, Seong KM, Jin YW, Youn H, Youn B. The Emerging Roles of Exosomes as EMT Regulators in Cancer. Cells 2020; 9:cells9040861. [PMID: 32252322 PMCID: PMC7226841 DOI: 10.3390/cells9040861] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 03/28/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) causes epithelial cells to lose their polarity and adhesion property, and endows them with migratory and invasive properties to enable them to become mesenchymal stem cells. EMT occurs throughout embryonic development, during wound healing, and in various pathological processes, including tumor progression. Considerable research in the last few decades has revealed that EMT is invariably related to tumor aggressiveness and metastasis. Apart from the interactions between numerous intracellular signaling pathways known to regulate EMT, extracellular modulators in the tumor microenvironment also influence tumor cells to undergo EMT, with extracellular vesicles (EVs) receiving increasing attention as EMT inducers. EVs comprise exosomes and microvesicles that carry proteins, nucleic acids, lipids, and other small molecules to stimulate EMT in cells. Among EVs, exosomes have been investigated in many studies, and their role has been found to be significant with respect to regulating intercellular communications. In this review, we summarize recent studies on exosomes and their cargoes that induce cancer-associated EMT. Furthermore, we describe the possible applications of exosomes as promising therapeutic strategies.
Collapse
Affiliation(s)
- Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (H.K.); (S.L.); (E.S.)
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (H.K.); (S.L.); (E.S.)
| | - Eunguk Shin
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (H.K.); (S.L.); (E.S.)
| | - Ki Moon Seong
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; (K.M.S.); (Y.W.J.)
| | - Young Woo Jin
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; (K.M.S.); (Y.W.J.)
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea
- Correspondence: (H.Y.); (B.Y.); Tel.: +82-2-6935-2438 (H.Y.); +82-51-510-2264 (B.Y.); Fax: +82-2-3408-4334 (H.Y.); +82-51-581-2962 (B.Y.)
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea; (H.K.); (S.L.); (E.S.)
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea
- Correspondence: (H.Y.); (B.Y.); Tel.: +82-2-6935-2438 (H.Y.); +82-51-510-2264 (B.Y.); Fax: +82-2-3408-4334 (H.Y.); +82-51-581-2962 (B.Y.)
| |
Collapse
|