1
|
Zhang C, Qin Y, Tang Y, Gu M, Li Z, Xu H. MEG3 in hematologic malignancies: from the role of disease biomarker to therapeutic target. Pharmacogenet Genomics 2024; 34:209-216. [PMID: 38743429 DOI: 10.1097/fpc.0000000000000534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Maternally expressed gene 3 ( MEG3 ) is a noncoding RNA that is known as a tumor suppressor in solid cancers. Recently, a line of studies has emphasized its potential role in hematological malignancies in terms of tumorigenesis, metastasis, and drug resistance. Similar to solid cancers, MEG3 can regulate various cancer hallmarks via sponging miRNA, transcriptional, or posttranslational regulation mechanisms, but may regulate different key elements. In contrast with solid cancers, in some subtypes of leukemia, MEG3 has been found to be upregulated and oncogenic. In this review, we systematically describe the role and underlying mechanisms of MEG3 in multiple types of hematological malignancies. Particularly, we highlight the role of MEG3 in drug resistance and as a novel therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | - Heng Xu
- Department of Laboratory Medicine/Research Center of Clinical Laboratory Medicine
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Lee JY, Bhandare RR, Boddu SHS, Shaik AB, Saktivel LP, Gupta G, Negi P, Barakat M, Singh SK, Dua K, Chellappan DK. Molecular mechanisms underlying the regulation of tumour suppressor genes in lung cancer. Biomed Pharmacother 2024; 173:116275. [PMID: 38394846 DOI: 10.1016/j.biopha.2024.116275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Tumour suppressor genes play a cardinal role in the development of a large array of human cancers, including lung cancer, which is one of the most frequently diagnosed cancers worldwide. Therefore, extensive studies have been committed to deciphering the underlying mechanisms of alterations of tumour suppressor genes in governing tumourigenesis, as well as resistance to cancer therapies. In spite of the encouraging clinical outcomes demonstrated by lung cancer patients on initial treatment, the subsequent unresponsiveness to first-line treatments manifested by virtually all the patients is inherently a contentious issue. In light of the aforementioned concerns, this review compiles the current knowledge on the molecular mechanisms of some of the tumour suppressor genes implicated in lung cancer that are either frequently mutated and/or are located on the chromosomal arms having high LOH rates (1p, 3p, 9p, 10q, 13q, and 17p). Our study identifies specific genomic loci prone to LOH, revealing a recurrent pattern in lung cancer cases. These loci, including 3p14.2 (FHIT), 9p21.3 (p16INK4a), 10q23 (PTEN), 17p13 (TP53), exhibit a higher susceptibility to LOH due to environmental factors such as exposure to DNA-damaging agents (carcinogens in cigarette smoke) and genetic factors such as chromosomal instability, genetic mutations, DNA replication errors, and genetic predisposition. Furthermore, this review summarizes the current treatment landscape and advancements for lung cancers, including the challenges and endeavours to overcome it. This review envisages inspired researchers to embark on a journey of discovery to add to the list of what was known in hopes of prompting the development of effective therapeutic strategies for lung cancer.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Sciences, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Richie R Bhandare
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates.
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates
| | - Afzal B Shaik
- St. Mary's College of Pharmacy, St. Mary's Group of Institutions Guntur, Affiliated to Jawaharlal Nehru Technological University Kakinada, Chebrolu, Guntur, Andhra Pradesh 522212, India; Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Lakshmana Prabu Saktivel
- Department of Pharmaceutical Technology, University College of Engineering (BIT Campus), Anna University, Tiruchirappalli 620024, India
| | - Gaurav Gupta
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Al-Jurf, P.O. Box 346, Ajman, United Arab Emirates; School of Pharmacy, Suresh Gyan Vihar University, Jaipur, Rajasthan 302017, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University, PO Box 9, Solan, Himachal Pradesh 173229, India
| | - Muna Barakat
- Department of Clinical Pharmacy & Therapeutics, Applied Science Private University, Amman-11937, Jordan
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara 144411, India; Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Kamal Dua
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Sydney 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
3
|
Hussain MS, Majami AA, Ali H, Gupta G, Almalki WH, Alzarea SI, Kazmi I, Syed RU, Khalifa NE, Bin Break MK, Khan R, Altwaijry N, Sharma R. The complex role of MEG3: An emerging long non-coding RNA in breast cancer. Pathol Res Pract 2023; 251:154850. [PMID: 37839358 DOI: 10.1016/j.prp.2023.154850] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/24/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023]
Abstract
MEG3, a significant long non-coding RNA (lncRNA), substantially functions in diverse biological processes, particularly breast cancer (BC) development. Within the imprinting DLK-MEG3 region on human chromosomal region 14q32.3, MEG3 spans 35 kb and encompasses ten exons. It exerts regulatory effects through intricate interactions with miRNAs, proteins, and epigenetic modifications. MEG3's multifaceted function in BC is evident in gene expression modulation, osteogenic tissue differentiation, and involvement in bone-related conditions. Its role as a tumor suppressor is highlighted by its influence on miR-182 and miRNA-29 expression in BC. Additionally, MEG3 is implicated in acute myocardial infarction and endothelial cell function, emphasising cell-specific regulatory mechanisms. MEG3's impact on gene activity encompasses transcriptional and post-translational adjustments, including DNA methylation, histone modifications, and interactions with transcription factors. MEG3 dysregulation is linked to unfavourable outcomes and drug resistance. Notably, higher MEG3 expression is associated with enhanced survival in BC patients. Overcoming challenges such as unravelling context-specific interactions, understanding epigenetic control, and translating findings into clinical applications is imperative. Prospective endeavours involve elucidating underlying mechanisms, exploring epigenetic alterations, and advancing MEG3-based diagnostic and therapeutic approaches. A comprehensive investigation into broader signaling networks and rigorous clinical trials are pivotal. Rigorous validation through functional and molecular analyses will shed light on MEG3's intricate contribution to BC progression.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017, Jaipur, Rajasthan, India
| | - Abdullah A Majami
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Haider Ali
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan.
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India; School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, 302017, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; Medical and Diagnostic Research Centre, University of Hail, Hail 55473, Saudi Arabia
| | - Nasrin E Khalifa
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; Medical and Diagnostic Research Centre, University of Hail, Hail 55473, Saudi Arabia; Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, 11115, Sudan
| | - Mohammed Khaled Bin Break
- Medical and Diagnostic Research Centre, University of Hail, Hail 55473, Saudi Arabia; Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| | - Ruqaiyah Khan
- Department of Basic Health Sciences, Deanship of Preparatory Year for the Health Colleges, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Najla Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint, Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Rahul Sharma
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017, Jaipur, Rajasthan, India
| |
Collapse
|
4
|
Hussain MS, Afzal O, Gupta G, Altamimi ASA, Almalki WH, Alzarea SI, Kazmi I, Fuloria NK, Sekar M, Meenakshi DU, Thangavelu L, Sharma A. Long non-coding RNAs in lung cancer: Unraveling the molecular modulators of MAPK signaling. Pathol Res Pract 2023; 249:154738. [PMID: 37595448 DOI: 10.1016/j.prp.2023.154738] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023]
Abstract
Lung cancer (LC) continues to pose a significant global medical burden, necessitating a comprehensive understanding of its molecular foundations to establish effective treatment strategies. The mitogen-activated protein kinase (MAPK) signaling system has been scientifically associated with LC growth; however, the intricate regulatory mechanisms governing this system remain unknown. Long non-coding RNAs (lncRNAs) are emerging as crucial regulators of diverse cellular activities, including cancer growth. LncRNAs have been implicated in LC, which can function as oncogenes or tumor suppressors, and their dysregulation has been linked to cancer cell death, metastasis, spread, and proliferation. Due to their involvement in critical pathophysiological processes, lncRNAs are gaining attention as potential candidates for anti-cancer treatments. This article aims to elucidate the regulatory role of lncRNAs in MAPK signaling in LC. We provide a comprehensive review of the key components of the MAPK pathway and their relevance in LC, focusing on aberrant signaling processes associated with disease progression. By examining recent research and experimental findings, this article examines the molecular mechanisms through which lncRNAs influence MAPK signaling in lung cancer, ultimately contributing to tumor development.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017 Jaipur, Rajasthan, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | | | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mahendran Sekar
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | | | - Lakshmi Thangavelu
- Center for Global Health Research , Saveetha Medical College , Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Ajay Sharma
- Delhi Pharmaceutical Science and Research University, Pushp Vihar Sector-3, MB Road, New Delhi 110017, India.
| |
Collapse
|
5
|
Hu Q, Huang T. Regulation of the Cell Cycle by ncRNAs Affects the Efficiency of CDK4/6 Inhibition. Int J Mol Sci 2023; 24:ijms24108939. [PMID: 37240281 DOI: 10.3390/ijms24108939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Cyclin-dependent kinases (CDKs) regulate cell division at multiple levels. Aberrant proliferation induced by abnormal cell cycle is a hallmark of cancer. Over the past few decades, several drugs that inhibit CDK activity have been created to stop the development of cancer cells. The third generation of selective CDK4/6 inhibition has proceeded into clinical trials for a range of cancers and is quickly becoming the backbone of contemporary cancer therapy. Non-coding RNAs, or ncRNAs, do not encode proteins. Many studies have demonstrated the involvement of ncRNAs in the regulation of the cell cycle and their abnormal expression in cancer. By interacting with important cell cycle regulators, preclinical studies have demonstrated that ncRNAs may decrease or increase the treatment outcome of CDK4/6 inhibition. As a result, cell cycle-associated ncRNAs may act as predictors of CDK4/6 inhibition efficacy and perhaps present novel candidates for tumor therapy and diagnosis.
Collapse
Affiliation(s)
- Qingyi Hu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
6
|
Seal RL, Tweedie S, Bruford EA. A standardised nomenclature for long non-coding RNAs. IUBMB Life 2023; 75:380-389. [PMID: 35880706 PMCID: PMC9877250 DOI: 10.1002/iub.2663] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/08/2022] [Indexed: 01/29/2023]
Abstract
The HUGO Gene Nomenclature Committee (HGNC) is the sole group with the authority to approve symbols for human genes, including long non-coding RNA (lncRNA) genes. Use of approved symbols ensures that publications and biomedical databases are easily searchable and reduces the risks of confusion that can be caused by using the same symbol to refer to different genes or using many different symbols for the same gene. Here, we describe how the HGNC names lncRNA genes and review the nomenclature of the seven lncRNA genes most mentioned in the scientific literature.
Collapse
Affiliation(s)
- Ruth L. Seal
- HUGO Gene Nomenclature Committee, European Molecular Biology LaboratoryEuropean Bioinformatics Institute, Wellcome Genome CampusHinxtonUK
- Department of HaematologyUniversity of Cambridge School of Clinical MedicineCambridgeUK
| | - Susan Tweedie
- HUGO Gene Nomenclature Committee, European Molecular Biology LaboratoryEuropean Bioinformatics Institute, Wellcome Genome CampusHinxtonUK
| | - Elspeth A. Bruford
- HUGO Gene Nomenclature Committee, European Molecular Biology LaboratoryEuropean Bioinformatics Institute, Wellcome Genome CampusHinxtonUK
- Department of HaematologyUniversity of Cambridge School of Clinical MedicineCambridgeUK
| |
Collapse
|
7
|
Elhelaly Elsherbeny M, Ramadan Elsergany A, Gamil Shaker O. Association of lncRNA MEG3 Rs7158663 Polymorphism and Serum Expression with Colorectal Cancer in Egyptian Patients. Rep Biochem Mol Biol 2023; 12:102-111. [PMID: 37724147 PMCID: PMC10505460 DOI: 10.52547/rbmb.12.1.102] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/16/2023] [Indexed: 09/20/2023]
Abstract
Background Colorectal cancer (CRC) is considered the third most common cancer around the world and second in terms of mortality. A significant aspect in its development is genetics. The risk of CRC and other clinicopathologic characteristics were investigated in this work in relation to the long non-coding RNA (lncRNA) MEG3 rs7158663 polymorphism, MEG3 expression, in an Egyptian population. Methods 160 CRC patients and 160 healthy controls were enrolled in this case-control study. The lncRNA MEG3 rs7158663 was examined using TaqMan Real-time PCR. RT-PCR was used to assess the levels of serum MEG3 expression. Results A significant higher expression of 'A' allele (risk allele) and A/A genotype in CRC cases vs. control subjects (P <0.001) Participants with A/A genotype had 4.8 times higher odds to exhibit CRC. Serum MEG3 gene expression was generally low in CRC patients, and it was considerably lower in those with the rs7158663 AA genotype than those with the GG genotype (P <0.001). It was found that CRC patients with the rs7158663 GA genotype had lower serum MEG3 expression levels than those with the GG genotype (P <0.001). Conclusions MEG3 low expression and MEG3 rs7158663 (AA) were associated with CRC risk in Egyptian patients and may serve as a diagnostic and prognostic marker for CRC patients.
Collapse
Affiliation(s)
- Mona Elhelaly Elsherbeny
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Alyaa Ramadan Elsergany
- Internal Medicine Department, Medical oncology unit, Oncology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Olfat Gamil Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
8
|
Zeng M, Zhang T, Lin Y, Lin Y, Wu Z. The Common LncRNAs of Neuroinflammation-Related Diseases. Mol Pharmacol 2023; 103:113-131. [PMID: 36456192 DOI: 10.1124/molpharm.122.000530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 12/04/2022] Open
Abstract
Spatio-temporal specific long noncoding RNAs (lncRNAs) play important regulatory roles not only in the growth and development of the brain but also in the occurrence and development of neurologic diseases. Generally, the occurrence of neurologic diseases is accompanied by neuroinflammation. Elucidation of the regulatory mechanisms of lncRNAs on neuroinflammation is helpful for the clinical treatment of neurologic diseases. This paper focuses on recent findings on the regulatory effect of lncRNAs on neuroinflammatory diseases and selects 10 lncRNAs that have been intensively studied to analyze their mechanism action. The clinical treatment status of lncRNAs as drug targets is also reviewed. SIGNIFICANCE STATEMENT: Gene therapies such as clustered regularly interspaced short palindrome repeats technology, antisense RNA technology, and RNAi technology are gradually applied in clinical treatment, and the development of technology is based on a large number of basic research investigations. This paper focuses on the mechanisms of lncRNAs regulation of neuroinflammation, elucidates the beneficial or harmful effects of lncRNAs in neurosystemic diseases, and provides theoretical bases for lncRNAs as drug targets.
Collapse
Affiliation(s)
- Meixing Zeng
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| | - Ting Zhang
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| | - Yan Lin
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| | - Yongluan Lin
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| | - Zhuomin Wu
- The First Affiliated Hospital of Shantou University Medical College (M.Z., Y.L., Z.W.) and The Second Affiliated Hospital of Shantou University Medical College (Y.L.), Shantou, Guangdong, China, and The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, China(T.Z.)
| |
Collapse
|
9
|
Zhang Z, Shi S, Li J, Costa M. Long Non-Coding RNA MEG3 in Metal Carcinogenesis. TOXICS 2023; 11:toxics11020157. [PMID: 36851033 PMCID: PMC9962265 DOI: 10.3390/toxics11020157] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 06/06/2023]
Abstract
Most transcripts from human genomes are non-coding RNAs (ncRNAs) that are not translated into proteins. ncRNAs are divided into long (lncRNAs) and small non-coding RNAs (sncRNAs). LncRNAs regulate their target genes both transcriptionally and post-transcriptionally through interactions with proteins, RNAs, and DNAs. Maternally expressed gene 3 (MEG3), a lncRNA, functions as a tumor suppressor. MEG3 regulates cell proliferation, cell cycle, apoptosis, hypoxia, autophagy, and many other processes involved in tumor development. MEG3 is downregulated in various cancer cell lines and primary human cancers. Heavy metals, such as hexavalent chromium (Cr(VI)), arsenic, nickel, and cadmium, are confirmed human carcinogens. The exposure of cells to these metals causes a variety of cancers. Among them, lung cancer is the one that can be induced by exposure to all of these metals. In vitro studies have demonstrated that the chronic exposure of normal human bronchial epithelial cells (BEAS-2B) to these metals can cause malignant cell transformation. Metal-transformed cells have the capability to cause an increase in cell proliferation, resistance to apoptosis, elevated migration and invasion, and properties of cancer stem-like cells. Studies have revealed that MEG is downregulated in Cr(VI)-transformed cells, nickel-transformed cells, and cadmium (Cd)-transformed cells. The forced expression of MEG3 reduces the migration and invasion of Cr(VI)-transformed cells through the downregulation of the neuronal precursor of developmentally downregulated protein 9 (NEDD9). MEG3 suppresses the malignant cell transformation of nickel-transformed cells. The overexpression of MEG3 decreases Bcl-xL, causing reduced apoptosis resistance in Cd-transformed cells. This paper reviews the current knowledge of lncRNA MEG3 in metal carcinogenesis.
Collapse
|
10
|
Xu J, Wang X, Zhu C, Wang K. A review of current evidence about lncRNA MEG3: A tumor suppressor in multiple cancers. Front Cell Dev Biol 2022; 10:997633. [PMID: 36544907 PMCID: PMC9760833 DOI: 10.3389/fcell.2022.997633] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNA (lncRNA) maternally expressed gene 3 (MEG3) is a lncRNA located at the DLK1-MEG3 site of human chromosome 14q32.3. The expression of MEG3 in various tumors is substantially lower than that in normal adjacent tissues, and deletion of MEG3 expression is involved in the occurrence of many tumors. The high expression of MEG3 could inhibit the occurrence and development of tumors through several mechanisms, which has become a research hotspot in recent years. As a member of tumor suppressor lncRNAs, MEG3 is expected to be a new target for tumor diagnosis and treatment. This review discusses the molecular mechanisms of MEG3 in different tumors and future challenges for the diagnosis and treatment of cancers through MEG3.
Collapse
Affiliation(s)
- Jie Xu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chunming Zhu
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, China,*Correspondence: Chunming Zhu, ; Kefeng Wang,
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China,*Correspondence: Chunming Zhu, ; Kefeng Wang,
| |
Collapse
|
11
|
Hu Q, Ma H, Chen H, Zhang Z, Xue Q. LncRNA in tumorigenesis of non-small-cell lung cancer: From bench to bedside. Cell Death Dis 2022; 8:359. [PMID: 35963868 PMCID: PMC9376075 DOI: 10.1038/s41420-022-01157-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022]
Abstract
Lung cancer has been one of the leading causes of cancer-related death worldwide, and non-small-cell lung cancer (NSCLC) accounts for the majority of lung cancer morbidity, yet the pathogenesis of NSCLC has not been fully elucidated. Recently, long-chain non-coding RNA (lncRNA) has attracted widespread attention. LncRNA is a type of non-coding RNA whose transcript length exceeds 200 nucleotides. After constant research, academics updated their understanding of lncRNA, especially its role in the biological processes of cancer cells, including epigenetic regulation, cell proliferation, and cell differentiation. Notably, examination of lncRNAs could serve as potential hallmarks for clinicopathological features, long-term prognosis, and drug sensitivity. Therefore, it is necessary to explore the functions of lncRNA in NSCLC and innovate potential strategies against NSCLC based on lncRNA-related research. Herein, we reviewed the functions of lncRNA in the occurrence, diagnosis, treatment, and prognosis of NSCLC, which not only help promote a comprehensive view of lncRNA in NSCLC, but also shed light on the potential of lncRNA-based diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Qin Hu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Medical School of Nantong University, Nantong, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Medical School of Nantong University, Nantong, China
| | - Hongyu Chen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Medical School of Nantong University, Nantong, China
| | - Zhouwei Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Medical School of Nantong University, Nantong, China
| | - Qun Xue
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| |
Collapse
|
12
|
Mirzazadeh S, Sarani H, Nakhaee A, Hashemi SM, Taheri M, Hashemi M, Bahari G. Association between PAX8AS1 (rs4848320 C > T, rs1110839 G > T, and rs6726151 T > G) and MEG3 (rs7158663) gene polymorphisms and non-Hodgkin lymphoma risk. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 41:1174-1186. [PMID: 35938744 DOI: 10.1080/15257770.2022.2104870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 06/06/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
Long non-coding RNA (lncRNA) PAX8 antisense RNA 1 (PAX8AS1) and Maternal-expressed gene 3 (MEG3) contribute to the pathogenesis of various malignancies including non-Hodgkin lymphoma (NHL). In this study, we aimed to examine the possible association of polymorphisms of PAX8 and MEG3 and the risk NHL. A total of 175 patients and 175 healthy subjects were genotyped by PCR-RFLP and Tetra-Arms PCR assays. Results demonstrated rs4848320 C > T and rs6726151 T > G of PAX8AS1 and rs7158663 of MEG3 play a potential role in the susceptibility of NHL and PAX8AS1 rs1110839 T > G variant was associated with decreased risk of NHL. Haplotype analysis of rs1110839, rs4848320, and rs6726151 demonstrated GCG haplotype is associated with increased risk of lymphoma and TTG, TTT, and GTT haplotypes are related to decreased lymphoma susceptibility.
Collapse
Affiliation(s)
- Samaneh Mirzazadeh
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hosna Sarani
- Children and Adolescent Health Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Alireza Nakhaee
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Seyed-Mehdi Hashemi
- Department of Internal Medicine, Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohsen Taheri
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Gholamreza Bahari
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
- Children and Adolescent Health Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
13
|
Zhang Z, Li J, Yan B, Tu H, Huang C, Costa M. Loss of MEG3 and upregulation of miR-145 play an important role in the invasion and migration of Cr(VI)-transformed cells. Heliyon 2022; 8:e10086. [PMID: 36046536 PMCID: PMC9421329 DOI: 10.1016/j.heliyon.2022.e10086] [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: 02/04/2022] [Revised: 04/25/2022] [Accepted: 07/21/2022] [Indexed: 11/30/2022] Open
Abstract
Chronic exposure of human bronchial epithelial BEAS-2B cells to hexavalent chromium (Cr(VI)) causes malignant cell transformation. These transformed cells exhibit increases in migration and invasion. Neuronal precursor of developmentally downregulated protein 9 (NEDD9) is upregulated in Cr(VI)-transformed cells compared to that of passage-matched normal BEAS-2B cells. Knockdown of NEDD9 by its shRNA reduced invasion and migration of Cr(VI)-transformed cells. Maternally expressed gene 3 (MEG3), a long noncoding RNA, was lost and microRNA 145 (miR-145) was upregulated in Cr(VI)-transformed cells. MEG3 was bound to miR-145 and this binding reduced its activity. Overexpression of MEG3 or inhibition of miR-145 decreased invasion and migration of Cr(VI)-transformed cells. Overexpression of MEG3 was able to decrease miR-145 level and NEDD9 protein level in Cr(VI)-transformed cells. Ectopic expression of MEG3 was also shown to reduce β-catenin activation. Inhibition of miR-145 in Cr(VI)-transformed cells decreased Slug, an important transcription factor that regulates epithelial-to-mesenchymal transition (EMT). Inhibition of miR-145 was found to increase MEG3 in Cr(VI)-transformed cells. Further studies showed that mutation of MEG3 at the binding site for miR-145 did not change NEDD9 and failed to decrease invasion and migration. The present study demonstrated that loss of MEG3 and upregulation of miR-145 elevated NEDD9, resulting in activation of β-catenin and further upregulation of EMT, leading to increased invasion and migration of Cr(VI)-transformed cells.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Jingxia Li
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Bo Yan
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Huailu Tu
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Chao Huang
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| | - Max Costa
- Department of Environmental Medicine, New York University Grossman School of Medicine, 341 East 25 Street, New York, New York, 10010, USA
| |
Collapse
|
14
|
Pan T, Ding H, Jin L, Zhang S, Wu D, Pan W, Dong M, Ma X, Chen Z. DNMT1-mediated demethylation of lncRNA MEG3 promoter suppressed breast cancer progression by repressing Notch1 signaling pathway. Cell Cycle 2022; 21:2323-2337. [PMID: 35822955 DOI: 10.1080/15384101.2022.2094662] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Breast carcinoma is one of the common causes of cancer-related mortality in women. Maternally expressed gene 3 (MEG3), a lncRNA located at 14q32, can be involved in carcinogenesis. In this study, we discovered that MEG3 was downregulated by CpG hypermethylation within its gene promoter. Functionally, treatment of breast cancer cells with the DNA methylation inhibitor 5-AzadC as well as silencing of DNA methyltransferase-1 (DNMT1) could decrease the abnormal hypermethylation of the MEG3 promoter, reverse MEG3 expression, inhibit cell proliferation and promote cell apoptosis. In addition, we found that MEG3 expression was negatively correlated with DNMT1. Mechanistically, MEG3 knockdown combined with 5-AzadC or sh-DNMT1 treatment restored the expression of Notch1 receptor, leading to the Notch1 pathway activation, and promoted the progression of epithelial mesenchymal transformation (EMT). Finally, the mice tumor model experiments showed that DNMT1 knockdown can increase MEG3 expression and inhibit tumor growth. Collectively, our findings uncovered that DNMT1-mediated MEG3 demethylation leads to MEG3 upregulation, which in turn inhibits the Notch1 pathway and EMT process in breast cancer.
Collapse
Affiliation(s)
- Tingting Pan
- Department of General Surgery, Diagnosis and Therapy Center of Thyroid and Breast, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Haiwen Ding
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Le Jin
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Shaobo Zhang
- Department of General Surgery, Hefei High-tech Cardiovascular Hospital, Hefei, Anhui, P.R. China
| | - Delin Wu
- Department of General Surgery, Diagnosis and Therapy Center of Thyroid and Breast, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Wanwan Pan
- Anhui Provincial Hospital, Wannan Medical College, Hefei, Anhui, P.R. China
| | - Menghao Dong
- Anhui Provincial Hospital, Wannan Medical College, Hefei, Anhui, P.R. China
| | - Xiaopeng Ma
- Department of General Surgery, Diagnosis and Therapy Center of Thyroid and Breast, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Zhaolin Chen
- Department of Pharmacy, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| |
Collapse
|
15
|
Long Non-Coding RNAs in Pancreatic Cancer: Biologic Functions, Mechanisms, and Clinical Significance. Cancers (Basel) 2022; 14:cancers14092115. [PMID: 35565245 PMCID: PMC9100048 DOI: 10.3390/cancers14092115] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022] Open
Abstract
Despite tremendous efforts devoted to research in pancreatic cancer (PC), the mechanism underlying the tumorigenesis and progression of PC is still not completely clear. Additionally, ideal biomarkers and satisfactory therapeutic strategies for clinical application in PC are still lacking. Accumulating evidence suggests that long non-coding RNAs (lncRNAs) might participate in the pathogenesis of diverse cancers, including PC. The abnormal expression of lncRNAs in PC is considered a vital factor during tumorigenesis that affects tumor cell proliferation, migration, invasion, apoptosis, angiogenesis, and drug resistance. With this review of relevant articles published in recent years, we aimed to summarize the biogenesis mechanism, classifications, and modes of action of lncRNAs and to review the functions and mechanisms of lncRNAs in PC. Additionally, the clinical significance of lncRNAs in PC was discussed. Finally, we pointed out the questions remaining from recent studies and anticipated that further investigations would address these gaps in knowledge in this field.
Collapse
|
16
|
Ou A, Zhao X, Lu Z. The potential roles of p53 signaling reactivation in pancreatic cancer therapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188662. [PMID: 34861354 DOI: 10.1016/j.bbcan.2021.188662] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/25/2022]
Abstract
Globally, pancreatic cancer (PC) is a common and highly malignant gastrointestinal tumor that is characterized by an insidious onset and ready metastasis and recurrence. Over recent decades, the incidence of PC has been increasing on an annual basis; however, the pathogenesis of this condition remains enigmatic. PC is not sensitive to radio- or chemotherapy, and except for early surgical resection, there is no curative treatment regime; consequently, the prognosis for patients with PC is extremely poor. Transcription factor p53 is known to play key roles in many important biological processes in vertebrates, including normal cell growth, differentiation, cell cycle progression, senescence, apoptosis, metabolism, and DNA damage repair. However, there is a significant paucity of basic and clinical studies to describe how p53 gene mutations or protein dysfunction facilitate the occurrence, progression, invasion, and resistance to therapy, of malignancies, including PC. Herein, we describe the involvement of p53 signaling reactivation in PC treatment as well as its underlying molecular mechanisms, thereby providing useful insights for targeting p53-related signal pathways in PC therapy.
Collapse
Affiliation(s)
- Aixin Ou
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, LN, China
| | - Xiangxuan Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, LN, China
| | - Zaiming Lu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, LN, China.
| |
Collapse
|
17
|
Rahimi HR, Mojarrad M, Moghbeli M. MicroRNA-96: A therapeutic and diagnostic tumor marker. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:3-13. [PMID: 35656454 DOI: 10.22038/ijbms.2021.59604.13226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022]
Abstract
Cancer has been always considered as one of the main human health challenges worldwide. One of the main causes of cancer-related mortality is late diagnosis in the advanced stages of the disease, which reduces the therapeutic efficiency. Therefore, novel non-invasive diagnostic methods are required for the early detection of tumors and improving the quality of life and survival in cancer patients. MicroRNAs (miRNAs) have pivotal roles in various cellular processes such as cell proliferation, motility, and neoplastic transformation. Since circulating miRNAs have high stability in body fluids, they can be suggested as efficient noninvasive tumor markers. MiR-96 belongs to the miR-183-96-182 cluster that regulates cell migration and tumor progression as an oncogene or tumor suppressor by targeting various genes in solid tumors. In the present review, we have summarized all of the studies that assessed the role of miR-96 during tumor progression. This review clarifies the molecular mechanisms and target genes recruited by miR-96 to regulate tumor progression and metastasis. It was observed that miR-96 mainly affects tumorigenesis by targeting the structural proteins and FOXO transcription factors.
Collapse
Affiliation(s)
- Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Ou A, Zhao X, Lu Z. The potential roles of p53 signaling reactivation in pancreatic cancer therapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188662. [DOI: doi10.1016/j.bbcan.2021.188662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
19
|
De Martino M, Esposito F, Pallante P. Long non-coding RNAs regulating multiple proliferative pathways in cancer cell. Transl Cancer Res 2021; 10:3140-3157. [PMID: 35116622 PMCID: PMC8797882 DOI: 10.21037/tcr-21-230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/27/2021] [Indexed: 01/17/2023]
Abstract
Long non-coding RNAs (lncRNAs) belong to an extremely heterogeneous class of non-coding RNAs with a length ranging from 200 to 100,000 bp. They modulate a series of cellular pathways in both physiological and pathological context. It is no coincidence that they are expressed in an aberrant way in pathologies such as cancer, so as to deserve to be subclassified as oncogenes or tumor suppressors. These molecules are also involved in the regulation of cancer cell proliferation. Several lncRNAs are able to modulate cell growth both positively and negatively, and in this review we have focused on a small group of them, characterized by the simultaneous action on different pathways regulating cell proliferation. They have been considered in the light of their behavior in three different subtypes of proliferative pathways that we can define as (I) tumor suppressor, (II) oncogenic and (III) transcriptionally-driven. More specifically, we have characterized some lncRNAs considered oncogenes (such as H19, linc-ROR, MALAT1, HULC, HOTAIR and ANRIL), tumor suppressors (such as MEG3 and lincRNA-p21), and both oncogenes/tumor suppressors (UCA1 and TUG1) in a little more detail. As can be understood from the review, the interactions between lncRNAs and their molecular targets, only in the context of controlling cell proliferation, give rise to an intricate molecular network, the understanding of which, in the future, will certainly be of help for the treatment of molecular diseases such as cancer.
Collapse
Affiliation(s)
- Marco De Martino
- Institute of Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| | - Francesco Esposito
- Institute of Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| | - Pierlorenzo Pallante
- Institute of Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| |
Collapse
|
20
|
Non-coding RNAs and lipids mediate the function of extracellular vesicles in cancer cross-talk. Semin Cancer Biol 2021; 74:121-133. [PMID: 34033894 DOI: 10.1016/j.semcancer.2021.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 11/22/2022]
Abstract
Research on extracellular vesicles (EVs) has been expanded, especially in the field of cancer. The cargoes in EVs, especially those in small EVs such as exosomes include microRNAs (miRNAs), mRNA, proteins, and lipids, are assumed to work cooperatively in the tumor microenvironment. In 2007, it was reported that miRNAs were abundant among the non-coding RNAs present in exosomes. Since then, many studies have investigated the functions of miRNAs and have tried to apply these molecules to aid in the diagnosis of cancer. Accordingly, many reviews of non-coding RNAs in EVs have been published for miRNAs. This review focuses on relatively new cargoes, covering long noncoding (lnc) RNAs, circular RNAs, and repeat RNAs, among non-coding RNAs. These RNAs, regardless of EV or cell type, have newly emerged due to the innovation of sequencing technology. The poor conservation, low quantity, and technical difficulty in detecting these RNA types have made it difficult to elucidate their functions and expression patterns. We herein summarize a limited number of studies. Although lipids are major components of EVs, current research on EVs focuses on miRNA and protein biology, while the roles of lipids in exosomes have not drawn attention. However, several recent studies revealed that phospholipids, which are components of the EV membrane, play important roles in the intercommunication between cells and in the generation of lipid mediators. Here, we review the reported roles of these molecules, and describe their potential in cancer biology.
Collapse
|
21
|
Fenofibrate Exerts Antitumor Effects in Colon Cancer via Regulation of DNMT1 and CDKN2A. PPAR Res 2021; 2021:6663782. [PMID: 33959155 PMCID: PMC8075693 DOI: 10.1155/2021/6663782] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/25/2021] [Accepted: 04/05/2021] [Indexed: 12/24/2022] Open
Abstract
Peroxisome proliferator-activated receptor alpha (PPARA) is the molecular target of fibrates commonly used to treat dyslipidemia and diabetes. Recently, the potential role of PPARA in other pathological conditions, such as cancers, has been recognized. Here, using bioinformatics analysis, we found that PPARA was expressed at relatively low levels in pancancers, and Kaplan-Meier analyses revealed that high PPARA protein expression was correlated with better survival of patients with colon cancer. In vitro experiments showed that fenofibrate regulated cell cycle distribution, promoted apoptosis, and suppressed cell proliferation and epithelial mesenchymal transition by activating PPARA. PPARA activation inhibited DNMT1 activity and abolished methylation-mediated CDKN2A repression. Downregulation of cyclin-CDK complexes led to the restoration of CDKN2A, which caused cell cycle arrest in the G1 phase via regulation of the CDKN2A/RB/E2F pathway. Finally, we demonstrated that fenofibrate administration inhibited tumor growth and DNMT1 activity in vivo. The PPARA agonist, fenofibrate, might serve as an applicable agent for epigenetic therapy of colon cancer patients.
Collapse
|
22
|
Zhu M, Wang F, Mi H, Li L, Wang J, Han M, Gu Y. Long noncoding RNA MEG3 suppresses cell proliferation, migration and invasion, induces apoptosis and paclitaxel-resistance via miR-4513/PBLD axis in breast cancer cells. Cell Cycle 2020; 19:3277-3288. [PMID: 33121324 DOI: 10.1080/15384101.2020.1839700] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Breast cancer remains a general-threat event in the health of women. Currently, increasing records indicate that long non-coding RNA maternally expressed 3 (MEG3) plays a central role in breast cancer. The current research focused on the function of MEG3 in paclitaxel (PTX)-resistance and human breast cancer growth. Levels of MEG3, microRNA (miR)-4513, and phenazine biosynthesis-like domain-containing protein (PBLD) were evaluated using quantitative real-time polymerase chain reaction (qRT-PCR) or western blot assays. 3-(4.5-dimethylghiazol-2-yl)-2,5-diphenyltetrazolium Bromide (MTT) assay was performed to examine the IC50 of PTX and cell proliferation in breast cancer cells. In addition, cell apoptosis was determined utilizing flow cytometry. Transwell was conducted to assay cell migration and invasion in MCF-7 and MDA-MB-231 cells. The interaction between miR-4513 and MEG3 or PBLD was expounded via dual-luciferase reporter assay. Levels of MEG3 and PBLD were decreased, but miR-4513 level was triggered in breast cancer tissues and cell lines. Overexpression of MEG3 could reinforce cell apoptosis, impede proliferation, migration, invasion, and the IC50 of PTX in breast cancer cells. Moreover, the impact of miR-4513 inhibitor on cell progression and PTX-resistance was overturned by MEG3 deficiency. Interestingly, miR-4513 mimic could abolish the role of PBLD upregulation in cell behaviors and PTX-resistance in MCF-7 and MDA-MB-231 cells. Finally, the expression of PBLD was co-modulated by miR-4513 and MEG3 in vitro. MEG3/miR-4513/PBLD axis modulated PTX-resistance and the development of breast cancer cells, which might provide a promising therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Mingzhi Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Fang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Hailong Mi
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Lin Li
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Jing Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Mingli Han
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Yuanting Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| |
Collapse
|
23
|
Li H, Wang P, Liu J, Liu W, Wu X, Ding J, Kang J, Li J, Lu J, Pan G. Hypermethylation of lncRNA MEG3 impairs chemosensitivity of breast cancer cells. J Clin Lab Anal 2020; 34:e23369. [PMID: 32618397 PMCID: PMC7521317 DOI: 10.1002/jcla.23369] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/13/2020] [Accepted: 04/02/2020] [Indexed: 01/01/2023] Open
Abstract
Background Chemoresistance posed a barrier to successful treatment of breast cancer (BC), and lncRNA MEG3 has been documented to implicate in BC development. However, whether MEG3 methylation, which led to low MEG3 expression, was relevant to BC progression and chemoresistance remained uncertain. Methods In the aggregate, 374 pairs of tumor tissues and adjacent normal tissues were collected from pathologically confirmed BC patients, and four BC cell lines, including MDA‐MB‐231, Bcap‐37, MCF‐7, and SK‐BR‐3, were purchased. Moreover, methylation‐specific polymerase chain reaction (PCR) was adopted to evaluate the methylation status of BC tissues and cell lines, and chemo‐tolerance of BC cell lines was assessed by performing MTT assay. Concurrently, transwell assay and scratch assay were carried out to estimate the migratory and invasive capability of BC cell lines. Results Methylated MEG3, lowly expressed MEG3, large tumor size (≥2 cm), advanced TNM grade and lymphatic metastasis were potentially symbolic of poor prognosis among BC patients (P < .05). Besides, MDA‐MB‐231 cell line exhibited the strongest resistance against paclitaxel, adriamycin, and vinorelbine (P < .05), while MCF‐7 cell line seemed more sensitive against these drugs than any other BC cell line (P < .05). Furthermore, pcDNA3.1‐MEG3 and 5‐Aza‐dC markedly sensitized MDA‐MB‐231 and MCF‐7 cell lines against the drug treatments (P < .05). Simultaneously, proliferation and metastasis of the BC cell lines were slowed down under the force of pcDNA3.1‐MEG3 and 5‐Aza‐dC (P < .05). Conclusion Preventing methylation of MEG3 might matter in lessening BC chemoresistance, owing to its hindering proliferation and metastasis of BC cells.
Collapse
Affiliation(s)
- Hongchang Li
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Puhua Wang
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Jiazhe Liu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Weiyan Liu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Xubo Wu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Junbin Ding
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Jie Kang
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Jindong Li
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Jingfeng Lu
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Gaofeng Pan
- Department of General Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Poulet C, Njock MS, Moermans C, Louis E, Louis R, Malaise M, Guiot J. Exosomal Long Non-Coding RNAs in Lung Diseases. Int J Mol Sci 2020; 21:E3580. [PMID: 32438606 PMCID: PMC7279016 DOI: 10.3390/ijms21103580] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Within the non-coding genome landscape, long non-coding RNAs (lncRNAs) and their secretion within exosomes are a window that could further explain the regulation, the sustaining, and the spread of lung diseases. We present here a compilation of the current knowledge on lncRNAs commonly found in Chronic Obstructive Pulmonary Disease (COPD), asthma, Idiopathic Pulmonary Fibrosis (IPF), or lung cancers. We built interaction networks describing the mechanisms of action for COPD, asthma, and IPF, as well as private networks for H19, MALAT1, MEG3, FENDRR, CDKN2B-AS1, TUG1, HOTAIR, and GAS5 lncRNAs in lung cancers. We identified five signaling pathways targeted by these eight lncRNAs over the lung diseases mentioned above. These lncRNAs were involved in ten treatment resistances in lung cancers, with HOTAIR being itself described in seven resistances. Besides, five of them were previously described as promising biomarkers for the diagnosis and prognosis of asthma, COPD, and lung cancers. Additionally, we describe the exosomal-based studies on H19, MALAT1, HOTAIR, GAS5, UCA1, lnc-MMP2-2, GAPLINC, TBILA, AGAP2-AS1, and SOX2-OT. This review concludes on the need for additional studies describing the lncRNA mechanisms of action and confirming their potential as biomarkers, as well as their involvement in resistance to treatment, especially in non-cancerous lung diseases.
Collapse
Affiliation(s)
- Christophe Poulet
- Department of Rheumatology, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (M.-S.N.); (M.M.)
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
| | - Makon-Sébastien Njock
- Department of Rheumatology, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (M.-S.N.); (M.M.)
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Respiratory Diseases, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| | - Catherine Moermans
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Respiratory Diseases, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| | - Edouard Louis
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Gastroenterology, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| | - Renaud Louis
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Respiratory Diseases, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| | - Michel Malaise
- Department of Rheumatology, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (M.-S.N.); (M.M.)
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
| | - Julien Guiot
- Fibropôle Research Group, University Hospital of Liège (CHULiege), 4000 Liège, Belgium; (E.L.); (R.L.)
- GIGA-I3 Research Group, GIGA Institute, University of Liège (ULiege) and University Hospital of Liège (CHULiege), 4000 Liège, Belgium;
- Department of Respiratory Diseases, University Hospital of Liège (CHULiege), 4000 Liège, Belgium
| |
Collapse
|
25
|
LncRNAs Act as a Link between Chronic Liver Disease and Hepatocellular Carcinoma. Int J Mol Sci 2020; 21:ijms21082883. [PMID: 32326098 PMCID: PMC7216144 DOI: 10.3390/ijms21082883] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are emerging as important contributors to the biological processes underlying the pathophysiology of various human diseases, including hepatocellular carcinoma (HCC). However, the involvement of these molecules in chronic liver diseases, such as nonalcoholic fatty liver disease (NAFLD) and viral hepatitis, has only recently been considered in scientific research. While extensive studies on the pathogenesis of the development of HCC from hepatic fibrosis have been conducted, their regulatory molecular mechanisms are still only partially understood. The underlying mechanisms related to lncRNAs leading to HCC from chronic liver diseases and cirrhosis have not yet been entirely elucidated. Therefore, elucidating the functional roles of lncRNAs in chronic liver disease and HCC can contribute to a better understanding of the molecular mechanisms, and may help in developing novel diagnostic biomarkers and therapeutic targets for HCC, as well as in preventing the progression of chronic liver disease to HCC. Here, we comprehensively review and briefly summarize some lncRNAs that participate in both hepatic fibrosis and HCC.
Collapse
|
26
|
Fan X, Huang H, Ji Z, Mao Q. Long non-coding RNA MEG3 functions as a competing endogenous RNA of miR-93 to regulate bladder cancer progression via PI3K/AKT/mTOR pathway. Transl Cancer Res 2020; 9:1678-1688. [PMID: 35117516 PMCID: PMC8798796 DOI: 10.21037/tcr.2020.01.70] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/03/2020] [Indexed: 01/21/2023]
Abstract
BACKGROUND Maternally expressed gene 3 (MEG3) is a long non-coding RNA (lncRNA) and involved in progression of various human tumors. However, its underlying regulatory mechanism in tumorigenesis of bladder cancer (BC) remains unclear. To demonstrate effects of MEG3 on BC cell proliferation and elaborate its regulatory mechanism in BC. METHODS Aberrant expressions of MEG3 and miR-93-5p were induced by cell transfection. The mRNA and protein expression were analyzed using qRT-PCR and western blot. Cell proliferation was examined by CCK-8 assay and EdU staining. The targeted regulation effect of MEG3 on miR-93-5p was confirmed by luciferase reporter assay. The number of LC3 punctated cells was detected by immunofluorescence. Xeno-graft mouse model was constructed for in vivo validation. RESULTS MEG3 was down-regulated with increased expression of miR-93-5p in BC cells and tissues. Luciferase reporter assay showed that miR-93-5p was a direct target of MEG3 and was negatively regulated by MEG3. MEG3 overexpression inhibited cell proliferation and the expression of proliferation-, apoptosis- and autophagy-related proteins. The activation of PI3K/AKT/mTOR pathway was also suppressed with elevated cell apoptosis. miR-93-5p overexpression counteracted these results. In vivo experiments, we confirmed that miR-93-5p overexpression reversed the MEG3 overexpression-mediated suppression on tumor growth and protein expression. CONCLUSIONS lncRNA MEG3 could function as a competing endogenous RNA of miR-93 to regulate the tumorigenesis of BC via PI3K/AKT/mTOR pathway. The present research provided a new perspective to understanding the pathogenic mechanism of BC, and an effective therapeutic target for BC.
Collapse
Affiliation(s)
- Xinrong Fan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Houfeng Huang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Quanzong Mao
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
27
|
Ji L, Ma L. MEG3 is restored by schisandrin A and represses tumor growth in choriocarcinoma cells. J Biochem Mol Toxicol 2020; 34:e22455. [PMID: 32057180 DOI: 10.1002/jbt.22455] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/17/2019] [Accepted: 01/21/2020] [Indexed: 01/01/2023]
Abstract
Schisandrin A (SchA) has been reported as a multidrug resistance-reversing agent; however, its antitumor effects have been rarely reported. Consequently, we attempted to explore whether SchA per se possesses an antitumor property in choriocarcinoma JEG-3 and BeWo cells and its potential mechanisms. JEG-3, BeWo, and HTR-8/SVneo cells were stimulated with SchA at different concentrations (10-100 μM), and cellular viability was evaluated with Cell Counting Kit-8. After stimulation with SchA, proliferation, apoptosis, migration, and invasion were detected by bromodeoxyuridine assay, Annexin V-fluorescein isothiocyanate/propidium iodide (Annexin V-FITC/PI) method, and a Transwell system, in JEG-3 cells transfected with short hairpin-RNA for maternally expressed 3. Western blot was performed to quantify protein. MEG3 was examined by a quantitative reverse transcription-polymerase chain reaction. MEG3 was downregulated in choriocarcinoma tissues. SchA diminished cellular viability, decreased proliferative activity, inhibited migratory and invasive behaviors, and repressed phosphorylation of regulators of phosphatidylinositol 3 kinase/protein kinase B/nuclear factor κB (PI3K/AKT/NF-κB) signaling cascade in gestational choriocarcinoma cells. MEG3 was upregulated by SchA in JEG-3 and BeWo cells. SchA exhibited little suppressive effects in JEG-3 cells lacking MEG3. Besides, the phosphorylation of transducers was evoked in MEG3-silenced JEG-3 cells despite stimulation with SchA. SchA administration repressed the growth of JEG-3 and BeWo cells by upregulating MEG3. Besides, SchA blocked PI3K/AKT/NF-κB signal cascade by elevating MEG3.
Collapse
Affiliation(s)
- Li Ji
- Department of Obstetrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| | - Li Ma
- Department of Obstetrics, Jining No. 1 People's Hospital, Jining, Shandong, China
| |
Collapse
|
28
|
Qin L, Zhong M, Adah D, Qin L, Chen X, Ma C, Fu Q, Zhu X, Li Z, Wang N, Chen Y. A novel tumour suppressor lncRNA F630028O10Rik inhibits lung cancer angiogenesis by regulating miR-223-3p. J Cell Mol Med 2020; 24:3549-3559. [PMID: 32052546 PMCID: PMC7131933 DOI: 10.1111/jcmm.15044] [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: 07/18/2019] [Revised: 11/28/2019] [Accepted: 12/29/2019] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the world's leading cause of cancer‐related morbidity and mortality despite advances in surgery, chemotherapy and immunotherapy; thus, there is an urgent need to find new molecules to develop novel treatment strategies. Although ncRNAs were found to account for 98% transcripts, the number of lncRNAs with distinct function in lung cancer is extremely limited. We previously demonstrated that Plasmodium infection inhibits tumour growth and metastasis, but the exact mechanisms involved have not been fully understood. In this study, we carried out RNA sequencing (RNA‐Seq) of tumour tissues isolated from LLC tumour‐bearing mice treated with either Plasmodium yoelli (Py)‐infected red blood cells or uninfected red blood cells. We found that F630028O10Rik (abbreviated as F63) is a novel lncRNA that was significantly up‐regulated in tumours isolated from mice treated with Py‐infected red blood cells compared to the control. By using gene silencing technique, F63 was found to inhibit both tumour Vascular Endothelial Growth Factor A (VEGFA) secretion and endothelial cells clone formation, migration, invasion and tube formation. Injection of cholesterol‐modified siRNA‐F63 into mice tumour tissues produced a significant increase in tumour volume, blood vessel formation and angiogenesis 17 days after injection. We further showed that inhibiting miR‐223‐3p results in the down‐regulation of VEGFA and VEGFR2 which are vital molecules for angiogenesis. These results reveal that F63 inhibit tumour growth and progression by modulating tumour angiogenesis suggesting F63 can be a novel lncRNA with great potential as a candidate molecule for gene therapy in lung cancer.
Collapse
Affiliation(s)
- Limei Qin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Foshan University, Guangdong, China.,School of Life Science and Engineering, Foshan University, Guangdong, China
| | - Menglong Zhong
- School of Life Science and Engineering, Foshan University, Guangdong, China
| | - Dickson Adah
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Science, Guangdong, China
| | - Li Qin
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Science, Guangdong, China
| | - Xiaoping Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Science, Guangdong, China
| | - Chunquan Ma
- School of Life Science and Engineering, Foshan University, Guangdong, China
| | - Qiang Fu
- School of Life Science and Engineering, Foshan University, Guangdong, China
| | - Xiaoping Zhu
- School of Life Science and Engineering, Foshan University, Guangdong, China
| | - Zhili Li
- School of Life Science and Engineering, Foshan University, Guangdong, China
| | - Nina Wang
- School of Life Science and Engineering, Foshan University, Guangdong, China
| | - Yanfeng Chen
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Foshan University, Guangdong, China.,School of Life Science and Engineering, Foshan University, Guangdong, China
| |
Collapse
|
29
|
Wang A, Hu N, Zhang Y, Chen Y, Su C, Lv Y, Shen Y. MEG3 promotes proliferation and inhibits apoptosis in osteoarthritis chondrocytes by miR-361-5p/FOXO1 axis. BMC Med Genomics 2019; 12:201. [PMID: 31888661 PMCID: PMC6937924 DOI: 10.1186/s12920-019-0649-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022] Open
Abstract
Background This study aimed to investigate the role of long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) and related molecular mechanisms, in osteoarthritis (OA). Methods Cartilage tissues of OA patients and healthy volunteers were isolated and cultured. After transfection with the appropriate constructs, chondrocytes were classified into Blank, pcDNA3.1-NC, pcDNA3.1-MEG3, si-NC, si-MEG3, pcDNA3.1-NC + mimics NC, pcDNA3.1-MEG3 + mimics NC, pcDNA3.1-NC + miR-361-5p mimics and pcDNA3.1-MEG3 + miR-361-5p mimics groups. qRT-PCR was used to detect the expression of MEG3, miR-361-5p and FOXO1. Western blot, luciferase reporter assay, RIP, CCK-8, and flow cytometry analysis were performed to reveal the morphology, proliferation, and apoptotic status of cartilage cells. Histological analysis and immunostaining were conducted in the OA rat model. Results Expression of MEG3 and FOXO1 was significantly decreased in OA compared with the normal group, while the expression of miR-361-5p was increased. MEG3 might serve as a ceRNA of miR-361-5p in OA chondrocytes. Moreover, using western blot analyses and the CCK-8 assay, MEG3 was shown to target miR-361-5p/FOXO1, elevate cell proliferation, and impair cell apoptosis. Functional analysis in vivo showed that MEG3 suppressed degradation of the cartilage matrix. Conclusion MEG3 can contribute to cell proliferation and inhibit cell apoptosis and degradation of extracellular matrix (ECM) via the miR-361-5p/FOXO1 axis in OA chondrocytes.
Collapse
Affiliation(s)
- Anying Wang
- Doctor Student, Hebei Medical University, No. 361, Zhongshan East Road, Hebei Province, Shijiazhuang, 050017, China.,Department of Orthopedic, The Second Affiliated Hospital of Shandong First Medical University, No. 366, Taishan Street, Shandong Province, Taishan, 271000, China
| | - Naixia Hu
- ICU, The Second Affiliated Hospital of Shandong First Medical University, No. 366, Taishan Street, Shandong Province, Taishan, 271000, China
| | - Yefeng Zhang
- Department of Orthopedic, The Second Affiliated Hospital of Shandong First Medical University, No. 366, Taishan Street, Shandong Province, Taishan, 271000, China
| | - Yuanzhen Chen
- Department of Orthopedic, The Central Hospital of Taian City, No. 29, Longtan Road, Shandong Province, Taian, 271000, China
| | - Changhui Su
- Department of Orthopedic, The Second Affiliated Hospital of Shandong First Medical University, No. 366, Taishan Street, Shandong Province, Taishan, 271000, China
| | - Yao Lv
- Department of Orthopedic, The Second Affiliated Hospital of Shandong First Medical University, No. 366, Taishan Street, Shandong Province, Taishan, 271000, China
| | - Yong Shen
- Department of Orthopedic, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Hebei Province, Shijiazhuang, 050051, China.
| |
Collapse
|
30
|
Wang J, Wang N, Zhao WD, Zhao LX, Jing YG, Yang LJ, He J, Li J. RNA-Seq Analysis Identified XLOC_009190 as Potential Therapeutic Target for Lung Adenocarcinoma. Onco Targets Ther 2019; 12:11221-11229. [PMID: 31908488 PMCID: PMC6927261 DOI: 10.2147/ott.s225532] [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: 07/31/2019] [Accepted: 10/31/2019] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The abnormal regulation on the expression of lncRNAs had been linked to multiple kinds of cancers, including lung adenocarcinoma. METHODS In this study, we carried out RNA-Seq on the three tumors and their paired normal samples from Chinese patients with lung adenocarcinoma. All the transcripts were de novo assembled, among which all the possible lncRNAs were predicted by tools including PLEK, CNCI, CPC, Blastp, hmmscan, and so forth. Their expression levels, altogether with the annotated mRNAs, were quantified. The weighted correlation network analysis and analysis of differential expression were carried out to explain the biological function of these novel lncRNAs. RESULTS The weighted correlation network analysis showed that the lncRNAs, which were highly correlated with protein-coding genes, participated in various pathways, including PI3K kinase pathways. These lncRNAs were important regulators in biological processes. Next, the differentially expressed lncRNAs were identified, including four known lncRNAs and one novel lncRNA (XLOC_009190). The cis-regulation of this novel lncRNA might act on MGST1, which protected cells by conjugation and glutathione peroxidase functions. The trans-regulation of this lncRNA was investigated by its correlated mRNAs. The results showed that it possibly played a role in transmembrane receptors like G protein-coupled receptors and potassium channels. CONCLUSION We proposed the potential biological function of XLOC_009190, but further experiments are needed to elucidate its roles and its potential to be the therapeutic target.
Collapse
Affiliation(s)
- Jing Wang
- Department of Surgery, School of Clinical Medicine, Dali University, Dali, Yunnan671000, People’s Republic of China
| | - Ning Wang
- Department of Surgery, School of Clinical Medicine, Dali University, Dali, Yunnan671000, People’s Republic of China
| | - Wei-dong Zhao
- The First Affiliated Hospital of Dali University, Dali, Yunnan671000, People’s Republic of China
| | - Li-xian Zhao
- The First Affiliated Hospital of Dali University, Dali, Yunnan671000, People’s Republic of China
| | - Yong-guang Jing
- Department of Human Anatomy, School of Basic Medical Sciences, Dali University, Dali, Yunnan671000, People’s Republic of China
| | - Li-jie Yang
- The First Affiliated Hospital of Dali University, Dali, Yunnan671000, People’s Republic of China
| | - Jie He
- The First Affiliated Hospital of Dali University, Dali, Yunnan671000, People’s Republic of China
| | - Jun Li
- Department of Surgery, School of Clinical Medicine, Dali University, Dali, Yunnan671000, People’s Republic of China
| |
Collapse
|
31
|
Yang B, Zheng C, Yu H, Zhang R, Zhao C, Cai S. Cardio-protective effects of salvianolic acid B on oxygen and glucose deprivation (OGD)-treated H9c2 cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2274-2281. [PMID: 31184214 DOI: 10.1080/21691401.2019.1621885] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The morphological feature of apoptosis is induced by oxygen and glucose deprivation (OGD) in cardiomyocytes H9c2 cells. Salvianolic acid B (Sal-B) has been studied in several pathological progresses, whereas it is still unclear whether maternally expressed gene 3 (MEG3) is an intermediate regulator during this progress. After pre-incubation with Sal-B and stimulation with OGD, viability and apoptosis of were examined in MEG3-overexpressed H9c2 cells. Cyclin D1, apoptosis-correlated proteins and regulators of signalling pathways were quantified with Western blot assay. MEG3 was detected by quantitative reverse transcription PCR (qRT-PCR). Sal-B was implicated in the enhancement of cell viability and suppression of apoptosis in OGD-treated H9c2 cells by repressing MEG3. In addition, MEG3 overexpression exerted an inhibitory effect on murine double minute 2 (MDM2) expression while aggrandized p53 expression in OGD-treated H9c2 cells which were pre-incubated with Sal-B. Furthermore, MEG3 overexpression abolished the up-regulative effect of Sal-B on phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) in OGD-treated H9c2 cells. These results indicated that cardio-protective function of Sal-B might be ascribed to its down-regulatory property on MEG3 expression which hence blocks p53 and triggers AMPK activation in OGD-treated cells.
Collapse
Affiliation(s)
- Bin Yang
- a Department of Cardiology, The Affiliated Hospital of Qingdao University , Qingdao , China
| | - Chunyan Zheng
- b Medical Consultation Center, The Affiliated Hospital of Qingdao University , Qingdao , China
| | - Haichu Yu
- a Department of Cardiology, The Affiliated Hospital of Qingdao University , Qingdao , China
| | - Rui Zhang
- a Department of Cardiology, The Affiliated Hospital of Qingdao University , Qingdao , China
| | - Cong Zhao
- a Department of Cardiology, The Affiliated Hospital of Qingdao University , Qingdao , China
| | - Shanglang Cai
- a Department of Cardiology, The Affiliated Hospital of Qingdao University , Qingdao , China
| |
Collapse
|
32
|
Wang M, Sun X, Wang H, Xin Y, Jiao W. Long non-coding RNAs in non-small cell lung cancer: functions and distinctions from other malignancies. Transl Cancer Res 2019; 8:2636-2653. [PMID: 35117021 PMCID: PMC8797712 DOI: 10.21037/tcr.2019.10.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/08/2019] [Indexed: 01/17/2023]
Abstract
Lung cancer leads to the most cancer-related death in the world. It was shown from the increasing evidences that long non-coding RNAs (lncRNAs) are emerging as molecules for diagnosis, prognosis and even therapy of lung cancer and other malignancies. The biological functions or involved signaling pathways of lncRNAs are always found to be inconsistent among different types of malignancies. However, no available literature has systemically summarized differences in the functions and underlying molecular mechanisms of lncRNAs between lung cancer and other cancers. In this review, the biological functions and molecular mechanisms of lncRNAs in lung cancer were introduced. Furthermore, their functional differences between lung cancer and other malignancies were discussed. Finally, their potential clinical applications in future lung cancer therapy were focused on.
Collapse
Affiliation(s)
- Maolong Wang
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiao Sun
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Hao Wang
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yanlu Xin
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Wenjie Jiao
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
33
|
Chi Y, Wang D, Wang J, Yu W, Yang J. Long Non-Coding RNA in the Pathogenesis of Cancers. Cells 2019; 8:cells8091015. [PMID: 31480503 PMCID: PMC6770362 DOI: 10.3390/cells8091015] [Citation(s) in RCA: 553] [Impact Index Per Article: 92.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/25/2019] [Accepted: 08/29/2019] [Indexed: 12/24/2022] Open
Abstract
The incidence and mortality rate of cancer has been quickly increasing in the past decades. At present, cancer has become the leading cause of death worldwide. Most of the cancers cannot be effectively diagnosed at the early stage. Although there are multiple therapeutic treatments, including surgery, radiotherapy, chemotherapy, and targeted drugs, their effectiveness is still limited. The overall survival rate of malignant cancers is still low. It is necessary to further study the mechanisms for malignant cancers, and explore new biomarkers and targets that are more sensitive and effective for early diagnosis, treatment, and prognosis of cancers than traditional biomarkers and methods. Long non-coding RNAs (lncRNAs) are a class of RNA transcripts with a length greater than 200 nucleotides. Generally, lncRNAs are not capable of encoding proteins or peptides. LncRNAs exert diverse biological functions by regulating gene expressions and functions at transcriptional, translational, and post-translational levels. In the past decade, it has been demonstrated that the dysregulated lncRNA profile is widely involved in the pathogenesis of many diseases, including cancer, metabolic disorders, and cardiovascular diseases. In particular, lncRNAs have been revealed to play an important role in tumor growth and metastasis. Many lncRNAs have been shown to be potential biomarkers and targets for the diagnosis and treatment of cancers. This review aims to briefly discuss the latest findings regarding the roles and mechanisms of some important lncRNAs in the pathogenesis of certain malignant cancers, including lung, breast, liver, and colorectal cancers, as well as hematological malignancies and neuroblastoma.
Collapse
Affiliation(s)
- Yujing Chi
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China
| | - Di Wang
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China
| | - Junpei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
- Key Laboratory of Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Beijing 100191, China
| | - Weidong Yu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
- Key Laboratory of Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Beijing 100191, China.
| |
Collapse
|
34
|
Liu HY, Lu SR, Guo ZH, Zhang ZS, Ye X, Du Q, Li H, Wu Q, Yu B, Zhai Q, Liu JL. lncRNA SLC16A1-AS1 as a novel prognostic biomarker in non-small cell lung cancer. J Investig Med 2019; 68:52-59. [PMID: 31371390 PMCID: PMC6996107 DOI: 10.1136/jim-2019-001080] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2019] [Indexed: 12/29/2022]
Abstract
Long non-coding RNAs (lncRNAs) have proved to act as crucial biomarkers in tumors. Novel biomarkers in non-small cell lung cancer (NSCLC) need to be investigated badly. To identify the differentially expressed lncRNAs between NSCLC tissue and adjacent tissue, microarray analysis was performed. lncRNA SLC16A1-AS1 was significantly less expressed in NSCLC tissue than that in adjacent tissue. Gain-of-function experiments was performed to determine the biological functions of SLC16A1-AS. In situhybridization and survival analysis were applied in lung cancer tissue samples to determine the prognostic role of SLC16A1-AS1. It was showed that SLC16A1-AS1 was remarkably downregulated in NSCLC tissues and cell lines. Functionally, SLC16A1-AS1 overexpression could inhibit the viability and proliferation of lung cancer cell, block the cell cycle and promote cell apoptosis in vitro which may result from reduced phosphorylation of rat sarcoma (RAS)/ proto-oncogene serine/threonine-protein kinase (RAF)/ mitogen-activated protein kinase kinase (MEK)/ extracellular regulated protein kinases (ERK) pathway caused by elevated expression of SLC16A1-AS1. Clinical sample analysis showed that SLC16A1-AS1 had a favorable impact on the overall survival and progression-free survival of patients with NSCLC. Our results suggested that SLC16A1-AS1 may act as a potential biomarker for patients with NSCLC.
Collapse
Affiliation(s)
- Hong Yue Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sheng Rong Lu
- Department of Pharmacy, The Central Hospital of Min-Hang District, Shanghai, China
| | - Zi Han Guo
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi Sheng Zhang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuan Ye
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiong Du
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Wu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Bo Yu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Zhai
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Long Liu
- Department of Biotechnology and Pathology, School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
35
|
Jiao J, Zhang S. Long non‑coding RNA MEG‑3 suppresses gastric carcinoma cell growth, invasion and migration via EMT regulation. Mol Med Rep 2019; 20:2685-2693. [PMID: 31524253 PMCID: PMC6691256 DOI: 10.3892/mmr.2019.10515] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/29/2018] [Indexed: 12/20/2022] Open
Abstract
Gastric carcinoma is one of the most frequently diagnosed gastrointestinal tumors. Long non-coding RNAs (lncRNAs) are broadly defined as endogenous cellular non-coding RNA molecules. Studies have demonstrated that they may be associated with human cancer progression. In the present study, the role of lncRNA-maternally expressed gene 3 (MEG3) in the progression of gastric carcinoma cells was investigated in vitro and in vivo. It was demonstrated that lncRNA-MEG3 expression was downregulated in gastric carcinoma cells compared with normal gastric cells. lncRNA-MEG3 transfection increased E-cadherin expression and markedly inhibited gastric carcinoma cell growth, migration and invasion. Flow cytometric analysis revealed that lncRNA-MEG3 transfection promoted the apoptosis of gastric carcinoma cells. Western blot analysis demonstrated that lncRNA-MEG3 transfection inhibited the expression of anti-apoptotic proteins B cell lymphoma-2 (Bcl-2) and Bcl-2-like protein 2 and increased the expression of pro-apoptotic proteins caspase-3 and caspase-9 in gastric carcinoma cells. lncRNA-MEG3 transfection upregulated the expression of epithelial marker E-cadherin and inhibited the expression of mesenchymal markers vimentin and fibronectin in gastric carcinoma cells, which suggested that lncRNA-MEG3 inhibited epithelial-mesenchymal transition (EMT), which may subsequently inhibit progression in gastric carcinoma cells. The present study also revealed that lncRNA-MEG3 transfection suppressed tumor growth mainly by decreasing the expression of vascular endothelial growth factor and increasing the expression of Bcl-2 in vivo. In conclusion, these results indicated that lncRNA-MEG3 may regulate EMT-associated signaling pathways and has the potential as a therapeutic target in gastric carcinoma.
Collapse
Affiliation(s)
- Junquan Jiao
- Center for Cancer Diagnosis and Treatment, Shangluo Central Hospital, Shangluo, Shaanxi 72600, P.R. China
| | - Shaobo Zhang
- Department of General Surgery, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, P.R. China
| |
Collapse
|
36
|
Maternally expressed gene 3 (MEG3): A tumor suppressor long non coding RNA. Biomed Pharmacother 2019; 118:109129. [PMID: 31326791 DOI: 10.1016/j.biopha.2019.109129] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022] Open
Abstract
Maternally expressed gene 3 (MEG3) is a long non-coding RNA (lncRNA) located on chromosome 14q32.3. Direct sequencing experiments have shown monoallelic expression of this lncRNA. Several studies have shown down-regulation of this lncRNA in human cancers. In some cases, hypermethylation of the promoter region has been suggested as the underlying mechanism. Functional studies have shown that this lncRNA controls expression of several tumor suppressor genes and oncogenes among them are p53, RB, MYC and TGF-β. Through regulation of Wnt-β-catenin pathway, it also affects epithelial-mesenchymal transition. In vitro studies have demonstrated contribution of MEG3 in defining response to chemotherapeutic agents such as paclitaxel, cisplatin and oxaliplatin. Certain polymorphisms within MEG3 are implicated in cancer risk (rs7158663, rs4081134 and rs11160608) or therapeutic response of cancer patients (rs10132552). Taken together, this lncRNA is regarded as a putative cancer biomarker and treatment target. In the current review, several aspects of the participation of MEG3 in carcinogenesis are discussed.
Collapse
|
37
|
Xu DH, Chi GN, Zhao CH, Li DY. Retracted: Long noncoding RNA MEG3 inhibits proliferation and migration but induces autophagy by regulation of Sirt7 and PI3K/AKT/mTOR pathway in glioma cells. J Cell Biochem 2019; 120:7516-7526. [PMID: 30417553 DOI: 10.1002/jcb.28026] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 10/15/2018] [Indexed: 02/02/2023]
Abstract
Glioma is a common primary brain tumor with high mortality rate and poor prognosis. Long noncoding RNA maternally expressed gene 3 (MEG3) is a tumor suppressor in diverse cancer types. However, the role of MEG3 in glioma remains unclear. We aimed to explore the effects of MEG3 on U251 cells as well as the underlying mechanisms. U251 cells were stably transfected with different recombined plasmids to overexpress or silence MEG3. Effects of aberrantly expressed MEG3 on cell viability, migration, apoptosis, expressions of apoptosis-associated and autophagy-associated proteins, and phosphorylated levels of key kinases in the PI3K/AKT/mTOR pathway were all evaluated. Then, messenger RNA (mRNA) and protein expression of Sirt7 in cells abnormally expressing MEG3 were estimated. In addition, effects of abnormally expressed MEG3 and Sirt7 on U251 cells were determined to reveal the underlying mechanism of MEG3-associated modulation. Cell viability and migration were significantly reduced by MEG3 overexpression whereas cell apoptosis as well as Bax and cleaved caspase-3/-9 proteins were obviously induced. Beclin-1 and LC3-II/LC3-I were upregulated and p62 was downregulated in MEG3 overexpressed cells. In addition, the autophagy pharmacological inhibitor (3-methyladenine, 3-MA) affected the effect of MEG3 overexpression on cell proliferation. Furthermore, the phosphorylated levels of key kinases in the PI3K/AKT/mTOR pathway were all reduced by MEG3 overexpression. Sirt7 was positively regulated by MEG3 expression, and effects of MEG3 overexpression on U251 cells were ameliorated by Sirt7 silence. MEG3 suppressed cell proliferation and migration but promoted autophagy in U251 cells through positively regulating Sirt7, involving in the inhibition of the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Dong-Hui Xu
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Guo-Nan Chi
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Cong-Hai Zhao
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Dong-Yuan Li
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
38
|
Screening of important lncRNAs associated with the prognosis of lung adenocarcinoma, based on integrated bioinformatics analysis. Mol Med Rep 2019; 19:4067-4080. [PMID: 30896819 PMCID: PMC6471985 DOI: 10.3892/mmr.2019.10061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 12/21/2018] [Indexed: 12/15/2022] Open
Abstract
The study aimed to elucidate the mechanisms underlying the occurrence and development of lung adenocarcinoma, and to reveal long non-coding RNA (lncRNA) prognostic factors to identify patients at high risk of disease recurrence or metastasis. Based on extensive RNA sequencing data and clinical survival prognosis information from patients with lung adenocarcinoma, obtained from The Cancer Genome Atlas and the Gene Expression Omnibus databases, a co-expression network of lncRNAs with different expression levels was built using weighted correlation network analysis and MetaDE.ES. The prognostic lncRNAs were identified using the Cox proportional hazards model and Kaplan-Meier survival curves to construct a risk scoring system. The reliability of the system was confirmed in validation datasets. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was performed on the genes significantly associated with the prognostic lncRNAs using gene set enrichment analysis. A total of 58 and 1,633 differentially expressed lncRNAs and mRNAs were identified, respectively. Considering the module stability, annotation, correlation between modules and clinical factors, and the differential expression levels of lncRNAs, 32 differentially expressed lncRNAs were selected from the brown, red, blue, green and yellow modules for subsequent survival analysis. A signature-based risk scoring system involving five lncRNAs [DIAPH2 antisense RNA 1, FOXN3 antisense RNA 2, long intergenic non-protein coding RNA 652, maternally expressed 3 and RHPN1 antisense RNA 1 (head to head)] was developed. The system successfully distinguished between low- and high-risk prognostic samples. System effectiveness was further verified using two independent validation datasets. Further KEGG pathway analysis indicated that the target genes of the five prognostic lncRNAs were associated with a number of cellular processes and signaling pathways, including the cell receptor-mediated signaling and cell adhesion pathways. A five-lncRNA signature predicts the prognosis of patients with lung adenocarcinoma. These prognostic lncRNAs may be potential diagnostic markers. The present results may help elucidate the pathogenesis of lung adenocarcinoma.
Collapse
|
39
|
Mycobacterium smegmatis But Not Mycobacterium avium subsp. hominissuis Causes Increased Expression of the Long Non-Coding RNA MEG3 in THP-1-Derived Human Macrophages and Associated Decrease of TGF-β. Microorganisms 2019; 7:microorganisms7030063. [PMID: 30818784 PMCID: PMC6463094 DOI: 10.3390/microorganisms7030063] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/13/2019] [Accepted: 02/26/2019] [Indexed: 11/16/2022] Open
Abstract
Pathogenic mycobacteria are able to persist intracellularly in macrophages, whereas non-pathogenic mycobacteria are effectively combated and eliminated after their phagocytosis. It is known that TGF-β plays an important role in this context. Infection with pathogenic mycobacteria such as Mycobacterium tuberculosis or M. avium leads to production of active TGF-β, which blocks the ability of IFN-γ and TNF-α to inhibit intracellular replication. On the other hand, it is known that the long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) is involved in the regulation of TGF-β. In this study, we show how the infection of THP-1-derived human macrophages with the saprophytic M. smegmatis but not with the facultatively pathogenic M. avium subsp. hominissuis leads to increased MEG3 expression. This is associated with the downregulation of DNA methyltransferases (DNMT) 1 and 3b, which are known to regulate MEG3 expression via promoter hypermethylation. Consequently, we observe a significant downregulation of TGF-β in M. smegmatis-infected macrophages but not in M. avium subsp. hominissuis pointing to lncRNAs as novel mediators of host cell response during mycobacterial infections.
Collapse
|
40
|
Abstract
Long noncoding RNAs (lncRNAs) have recently considered as central regulators in diverse biological processes and emerged as vital players controlling tumorigenesis. Several lncRNAs can be classified into oncogenes and tumor suppressor genes depending on their function in cancer. A maternally expressed gene 3 (MEG3) gene transcripts a 1.6 kb lncRNA whose act as an antitumor component in different cancer cells, such as breast, liver, glioma, colorectal, cervical, gastric, lung, ovarian and osteosarcoma cancer cells. The present review highlights biological function of MEG3 to repress tumor through regulating the major tumor suppressor genes p53 and Rb, inhibiting angiogenesis-related factor, or controlling miRNAs. On the other hand, previous studies have also suggested that MEG3 mediates epithelial-mesenchymal transition (EMT). However, deregulation of MEG3 is associated with the development and progression of cancer, suggesting that MEG3 may function as a potential biomarker and therapeutic target for human cancers.
Collapse
|
41
|
Bai Y, Zhang Q, Su Y, Pu Z, Li K. Modulation of the Proliferation/Apoptosis Balance of Vascular Smooth Muscle Cells in Atherosclerosis by lncRNA-MEG3 via Regulation of miR-26a/Smad1 Axis. Int Heart J 2019; 60:444-450. [PMID: 30745534 DOI: 10.1536/ihj.18-195] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The balance between proliferation and apoptosis of vascular smooth muscle cells (VSMCs) plays a critical role in the initiation of atherosclerosis. LncRNA-MEG3 is involved in the pathophysiology of atherosclerosis through regulation of endothelial cell proliferation and migration. Its effect on the dysfunction of VSMCs and the corresponding mechanisms are actively researched. In this study, we observed that downregulated lncRNA-MEG3 expression was inversely correlated with the microRNA-26a level in coronary artery disease tissues. The overexpression of lncRNA-MEG3 could inhibit VSMCs proliferation while facilitating apoptosis. Moreover, alteration in the miR-26a/Smad1 axis could antagonize this effect. Bioinformatic analysis indicated that lncRNA-MEG3 could interact with miR-26a via complementary binding sites. The enforced expression of lncRNA-MEG3 could reduce the level of miR-26a in VSMCs, while the expression of Smad1 increases. Further, the direct binding between lncRNA-MEG3 and miR-26a was confirmed via dual-luciferase reporter assay, which indicated that lnc-MEG3 could sponge miR-26a as a competing endogenous RNA. In summary, we propose that lncRNA-MEG3 modulates the proliferation/apoptosis balance of VSMCs in atherosclerosis by regulating the miR-26a/Smad1 axis.
Collapse
Affiliation(s)
- Yang Bai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Qiangnu Zhang
- School of Basic Medical Sciences, Lanzhou University
| | - Yijiang Su
- Department of Cardiothoracic Surgery, Shanghai General Hospital, The First People's Hospital Affiliated with Shanghai Jiaotong University
| | - Zhenye Pu
- Department of Cardiothoracic Surgery, Zhongda Hospital Affiliated to Southeast University
| | - Kunsheng Li
- Department of Cardiothoracic Surgery, Nanjing First Hospital, Nanjing Heart Institute, Nanjing Medical University
| |
Collapse
|
42
|
Long noncoding RNAs in cancer cells. Cancer Lett 2019; 419:152-166. [PMID: 29414303 DOI: 10.1016/j.canlet.2018.01.053] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 12/11/2022]
Abstract
Long noncoding RNA (lncRNA) has recently been investigated as key modulators that regulate many biological processes in human cancers via diverse mechanisms. LncRNAs can interact with macromolecules such as DNA, RNA, or protein to exert cellular effects and to act as either tumor promoters or tumor suppressors in various malignancies. Moreover, the aberrant expression of lncRNAs may be detected in multiple cancer phenotypes by employing the rapidly developing modern gene chip technology and bioinformatics analysis. Herein, we highlight the mechanisms of action of lncRNAs, their functional cellular roles and their involvement in cancer progression. Finally, we provide an overview of recent progress in the lncRNA field and future potential for lncRNAs as cancer diagnostic markers and therapeutics.
Collapse
|
43
|
Lan Y, Li YJ, Li DJ, Li P, Wang JY, Diao YP, Ye GD, Li YF. Long noncoding RNA MEG3 prevents vascular endothelial cell senescence by impairing miR-128-dependent Girdin downregulation. Am J Physiol Cell Physiol 2018; 316:C830-C843. [PMID: 30576236 DOI: 10.1152/ajpcell.00262.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) are commonly associated with various biological functions, in which the function of lncRNA maternally expressed gene 3 (MEG3) has been identified in various cancers. Strikingly, an association between MEG3 with microRNAs (miRNAs), mRNAs, and proteins has been reported. This study investigates the role of MEG3 in vascular endothelial cell (VEC) senescence. Expression of Girdin and miR-128 was monitored in the blood vessel samples of young and old mice/healthy volunteers, along with the measurement of human umbilical vein endothelial cells (HUVECs). The relationship between MEG3/Girdin and miR-128 was determined and verified. Loss- and gain-of-function approaches were applied to analyze the regulatory effects of MEG3 on platelet phagocytosis and lipoprotein oxidation of HUVEC membrane. In addition, the effect of MEG3 on HUVEC senescence was evaluated by detection of the reactive oxygen species, telomerase activity, and telomere length. To further analyze the MEG3-mediated regulatory mechanism, miR-128 upregulation and inhibition were introduced into the HUVECs. Downregulated Girdin and upregulated miR-128 were found in the blood vessels of old individuals and old mice, as well as in senescent HUVECs. MEG3 downregulation was found to be capable of inhibiting Girdin but enhancing miR-128 expression. It was also indicated to inhibit platelet phagocytosis and reduce telomerase activity and telomere length, while enhancing lipoprotein oxidation and reactive oxygen species production, which ultimately contributed in preventing and protecting HUEVCs from senescence. These findings provide evidence supporting that MEG3 leads to miR-128 downregulation and Girdin upregulation, which promotes platelet phagocytosis, thus protecting VECs from senescence.
Collapse
Affiliation(s)
- Yong Lan
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yong-Jun Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Da-Jun Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Peng Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Ji-Yang Wang
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yong-Peng Diao
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Guo-Dong Ye
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yang-Fang Li
- Beijing Neurosurgical Institute, Capital Medical University , Beijing , People's Republic of China
| |
Collapse
|
44
|
Zhang Y, Wu J, Jing H, Huang G, Sun Z, Xu S. Long noncoding RNA
MEG3
inhibits breast cancer growth via upregulating endoplasmic reticulum stress and activating NF‐κB and p53. J Cell Biochem 2018; 120:6789-6797. [PMID: 30556250 DOI: 10.1002/jcb.27982] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Yan Zhang
- Department of Pathology Huaihe Hospital, Henan University Kaifeng China
| | - Jiang Wu
- Department of Pathology Huaihe Hospital, Henan University Kaifeng China
| | - Hong Jing
- Department of Pathology Huaihe Hospital, Henan University Kaifeng China
| | - Gui Huang
- Department of Pathology Huaihe Hospital, Henan University Kaifeng China
| | - Zhulei Sun
- Department of Pathology Huaihe Hospital, Henan University Kaifeng China
| | - Shouming Xu
- Henan Key Laboratory of Plant Stress Biology, School of Life Science, Henan University Kaifeng China
| |
Collapse
|
45
|
Liu G, Zhao X, Zhou J, Cheng X, Ye Z, Ji Z. Long non-coding RNA MEG3 suppresses the development of bladder urothelial carcinoma by regulating miR-96 and TPM1. Cancer Biol Ther 2018; 19:1039-1056. [PMID: 30461333 DOI: 10.1080/15384047.2018.1480279] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We aimed at investigating effects of long non-coding RNA maternally expressed 3 (MEG3) on the proliferation, cell cycle and apoptosis of bladder urothelial carcinoma cells and regulatory relationships among lncRNA MEG3, miR-96 and α-tropomyosin 1 (TPM1). Human clinical data from The Cancer Genome Atlas (TCGA) which contains bladder urothelial carcinoma tissues and adjacent tissues were used for analysis. The expression profiles of MEG3, miR-96, TPM1, cell cycle-related genes and apoptosis-related genes were examined by real-time quantitative polymerase chain reaction (RT-qPCR) and western blot. Regulating relationship among MEG3, miR-96 and TPM1 was confirmed by dual luciferase reporter assay. MTT assay and flow cytometry were performed to observe cell proliferation, cell cycle and apoptosis. The effects of lncRNA MEG3 on bladder urothelial carcinoma were confirmed both in vivo and in vitro. The mRNA expression and protein expression of MEG3, TPM1 were down-regulated in carcinoma tissues, whereas miR-96 expression was up-regulated. MEG3 overexpression resulted in miR-96 downregulation along with TPM1 upregulation, which inhibited cell proliferation and cell cycle but promoted cell apoptosis of bladder urothelial carcinoma cells in vitro, and at the same time inhibited tumor growth in vivo. In this process, expressions of apoptosis-related protein BCL2 associated X (Bax), cleaved-caspase 3 was up-regulated, whereas apoptosis regulator protein (Bcl-2) expression was suppressed when MEG3 was overexpressed, and cell cycle-related protein Cyclin D1 was down-regulated. LncRNA MEG3 low-expression promotes the proliferation and inhibits apoptosis of bladder urothelial carcinoma cells by regulating miR-96 along with TPM1.
Collapse
Affiliation(s)
- Guanghua Liu
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Xin Zhao
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Jingmin Zhou
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Xiangming Cheng
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Zixing Ye
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Zhigang Ji
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| |
Collapse
|
46
|
Guo T, Li J, Zhang L, Hou W, Wang R, Zhang J, Gao P. Multidimensional communication of microRNAs and long non-coding RNAs in lung cancer. J Cancer Res Clin Oncol 2018; 145:31-48. [PMID: 30417217 DOI: 10.1007/s00432-018-2767-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/06/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE Non-coding RNAs (ncRNAs) have been a hot topic for many years in the field of cancer research, especially miRNAs and lncRNAs. Because they play critical roles in regulating various cellular processes and are more often involved in tumorigenesis than protein-coding genes. But the cross talk between miRNAs and lncRNAs in cancer has been scarcely studied. This article aims to provide a retrospective review of the latest research on the link between miRNAs and lncRNAs in lung cancer and discusses their potential role as diagnostic biomarkers and therapeutic targets for lung cancer in clinical practice. METHODS We reviewed literatures about ncRNAs and lung cancer from PUBMED databases in this article. RESULTS As shown in our review, miRNAs and lncRNAs could represent underlying targets for diagnosis, therapy, prognosis, and drug resistence of lung cancer. By acting as ceRNAs, lncRNAs can competitively inhibit the expression levels of miRNAs, and the lncRNA/miRNA axis can contribute to tumorigenesis, metastasis, and mutidrug resistance in lung cancer via various classic signaling pathways or related proteins. CONCLUSION Based on present knowledge, ncRNAs may provide a novel perspective to understand the pathogenesis of lung cancer and could be candidates in screening of therapeutic targets for lung cancer.
Collapse
Affiliation(s)
- Tingting Guo
- Department of Respiratory Medicine, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, P.R. China
| | - Junyao Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, P.R. China
| | - Lin Zhang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, P.R. China
| | - Wei Hou
- Department of Respiratory Medicine, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, P.R. China
| | - Rongrong Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, P.R. China
| | - Jie Zhang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, P.R. China.
| | - Peng Gao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, 218 Ziqiang Street, Nanguan District, Changchun, 130041, Jilin, P.R. China.
| |
Collapse
|
47
|
García-López S, Albo-Castellanos C, Urdinguio RG, Cañón S, Sánchez-Cabo F, Martínez-Serrano A, Fraga MF, Bernad A. Deregulation of the imprinted DLK1-DIO3 locus ncRNAs is associated with replicative senescence of human adipose-derived stem cells. PLoS One 2018; 13:e0206534. [PMID: 30395586 PMCID: PMC6218046 DOI: 10.1371/journal.pone.0206534] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/15/2018] [Indexed: 12/24/2022] Open
Abstract
Background Human adult adipose-derived stem cells (hADSCs) have become the most promising cell source for regenerative medicine. However the prolonged ex vivo expansion periods required to obtain the necessary therapeutic dose promotes progressive senescence, with the concomitant reduction of their therapeutic potential. Aim and scope A better understanding of the determinants of hADSC senescence is needed to improve biosafety while preserving therapeutic efficiency. Here, we investigated the association between deregulation of the imprinted DLK1-DIO3 region and replicative senescence in hADSC cultures. Methods We compared hADSC cultures at short (PS) and prolonged (PL) passages, both in standard and low [O2] (21 and 3%, respectively), in relation to replicative senescence. hADSCs were evaluated for expression alterations in the DLK1-DIO3 region on chromosome 14q32, and particularly in its main miRNA cluster. Results Comparison of hADSCs cultured at PL or PS surprisingly showed a quite significant fraction (69%) of upregulated miRNAs in PL cultures mapping to the imprinted 14q32 locus, the largest miRNA cluster described in the genome. In agreement, expression of the lncRNA MEG3 (Maternally Expressed 3; Meg3/Gtl2), cultured at 21 and 3% [O2], was also significantly higher in PL than in PS passages. During hADSC replicative senescence the AcK16H4 activating mark was found to be significantly associated with the deregulation of the entire DLK1-DIO3 locus, with a secondary regulatory role for the methylation of DMR regions. Conclusion A direct relationship between DLK1-DIO3 deregulation and replicative senescence of hADSCs is reported, involving upregulation of a very significant fraction of its largest miRNA cluster (14q32.31), paralleled by the progressive overexpression of the lncRNA MEG3, which plays a central role in the regulation of Dlk1/Dio3 activation status in mice.
Collapse
Affiliation(s)
- Silvia García-López
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, Spain
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Carlos III (CNIC), Madrid, Spain
| | - Carmen Albo-Castellanos
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Carlos III (CNIC), Madrid, Spain
| | - Rocio G. Urdinguio
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Hospital Universitaria Central de Asturias (HUCA) and Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo (UO), Asturias, Spain
| | - Susana Cañón
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, Spain
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Carlos III (CNIC), Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Alberto Martínez-Serrano
- Molecular Biology Department (UAM) and Molecular Neuropathology Department, Center of Molecular Biology Severo Ochoa-CSIC, Universidad Autónoma de Madrid, Campus Cantoblanco, Madrid, Spain
| | - Mario F. Fraga
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Hospital Universitaria Central de Asturias (HUCA) and Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Universidad de Oviedo (UO), Asturias, Spain
| | - Antonio Bernad
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, Spain
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Carlos III (CNIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
48
|
Yang Z, Li H, Li J, Lv X, Gao M, Bi Y, Zhang Z, Wang S, Li S, Li N, Cui Z, Zhou B, Yin Z. Association Between Long Noncoding RNAMEG3Polymorphisms and Lung Cancer Susceptibility in Chinese Northeast Population. DNA Cell Biol 2018; 37:812-820. [DOI: 10.1089/dna.2018.4277] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Zitai Yang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Hang Li
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Juan Li
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Xiaoting Lv
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Min Gao
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Yanhong Bi
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Ziwei Zhang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Shengli Wang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Sixuan Li
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Na Li
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Zhigang Cui
- School of Nursing, China Medical University, Shenyang, People's Republic of China
| | - Baosen Zhou
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| | - Zhihua Yin
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Cancer Etiology and Intervention, University of Liaoning Province, Shenyang, People's Republic of China
| |
Collapse
|
49
|
Moradi M, Fallahi H, Rahimi Z. Interaction of long noncoding RNA MEG3 with miRNAs: A reciprocal regulation. J Cell Biochem 2018; 120:3339-3352. [DOI: 10.1002/jcb.27604] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/07/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Mohammad‐Taher Moradi
- Medical Biology Research Center, Kermanshah University of Medical Sciences Kermanshah Iran
| | - Hossein Fallahi
- Medical Biology Research Center, Kermanshah University of Medical Sciences Kermanshah Iran
- Bioinformatics Lab, Department of Biology School of Sciences, Razi University Kermanshah Iran
| | - Zohreh Rahimi
- Medical Biology Research Center, Kermanshah University of Medical Sciences Kermanshah Iran
- Department of Clinical Biochemistry Medical School, Kermanshah University of Medical Sciences Kermanshah Iran
| |
Collapse
|
50
|
Identification of abnormally expressed lncRNAs induced by PM2.5 in human bronchial epithelial cells. Biosci Rep 2018; 38:BSR20171577. [PMID: 29899163 PMCID: PMC6131355 DOI: 10.1042/bsr20171577] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 05/21/2018] [Accepted: 06/11/2018] [Indexed: 01/17/2023] Open
Abstract
To investigate the effect of stimulation of human bronchial epithelial cells (HBECs) by arterial traffic ambient PM2.5 (TAPM2.5) and wood smoke PM2.5 (WSPM2.5) on the expression of long non-coding RNAs (lncRNAs) in order to find new therapeutic targets for treatment of chronic obstructive pulmonary disease (COPD). HBECs were exposed to TAPM2.5 and WSPM2.5 at a series of concentrations. The microarray analysis was used to detect the lncRNA and mRNA expression profiles. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and gene ontology (GO) enrichment were conducted to analyze the differentially expressed lncRNAs and mRNAs. Quantitative real-time PCR (qRT-PCR) was performed to confirm the differential expression of lncRNAs. Western blot was performed to study the expression of autophagy and apoptosis-associated proteins. Flow cytometry was used to detect the apoptotic cells. The results indicated that fine particulate matter (PM2.5)-induced cell damage of HBECs occurred in a dose-dependent manner. The microarray analysis indicated that treatment with TAPM2.5 and WSPM2.5 led to the alteration of lncRNA and mRNA expression profiles. LncRNA maternally expressed gene 3 (MEG3) was significantly up-regulated in HBECs after PM2.5 treatment. The results of Western blot showed that PM2.5 induced cell apoptosis and autophagy by up-regulating apoptosis-associated gene, caspase-3, and down-regulating autophagy-associated markers, Bcl-2 and LC3 expression. In addition, we demonstrated that TAPM2.5 and WSPM2.5 accelerated apoptosis of human bronchial (HBE) cells, silencing of MEG3 suppressed apoptosis and autophagy of HBE cells. These findings suggested that the lncRNA MEG3 mediates PM2.5-induced cell apoptosis and autophagy, and probably through regulating the expression of p53.
Collapse
|