1
|
Hamdy NM, Barakat BM, El-Sisi MG, Shaker FH, Sallam AAM, Elazazy O, Darwish SF, Elmakromy GM, Ibrahim IH, Anwar MM. Comprehensive review and in silico analysis of the role of noncoding RNAs in retinoblastoma: A step-toward ncRNA precision. Int J Biol Macromol 2025; 311:144036. [PMID: 40345278 DOI: 10.1016/j.ijbiomac.2025.144036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Noncoding RNAs (ncRNAs) have greatly revolutionized our understanding of gene regulation and its main role in oncogenesis, particularly in retinoblastoma (RB), the most prevalent type of intraocular malignancy in children. Despite recent significant therapeutic advances, the prognosis for RB remains unclear owing to late diagnosis and resistance to conventional treatments. This review comprehensively explores the multiple roles of ncRNAs-microRNAs (miRNAs), long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), and PIWI-interacting RNAs (piRNAs)-in RB pathogenesis. miRNA dysregulation serves as the initial cascade for modulating cell proliferation, apoptosis, and metastasis. Similarly, lncRNAs demonstrate dual behavior, functioning either as oncogenic drivers or tumor suppressors by interacting with several molecular targets and interacting with different signaling pathways, such as the PI3K/Akt and Wnt/β-catenin pathways. Additionally, circRNAs, owing to their persistent stability and unique ability to act as miRNA sponge main binding sites, affect various normal physiological processes, influencing tumor progression and chemoresistance. Emerging data also highlight the intricate crosstalk between piRNAs and other ncRNAs in retinal homeostasis and oncogenesis, with promising future implications for their utility as diagnostic biomarkers in liquid biopsy types. This comprehensive review consolidates the latest knowledge on the molecular mechanisms of noncoding RNAs (ncRNAs) in retinoblastoma (RB), along with in silico analysis of ncRNA-gene interactions, providing a guide for precision medical approaches. However, future research should aim to utilize ncRNAs as a vital clinical tool to improve the early diagnosis, prognosis, and targeted treatment of RB.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| | - Bassant M Barakat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al Baha University, Al Baha 1988, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11651, Egypt
| | - Mona G El-Sisi
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Fatma H Shaker
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Al-Aliaa M Sallam
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ola Elazazy
- Biochemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Samar F Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Gena M Elmakromy
- Internal Medicine Department, Faculty of Medicine, Badr University In Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Iman Hassan Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| |
Collapse
|
2
|
Tie J, Guo J, Huang Y, Huang Z, Yan Z, Yuan J, Shen X, Wang J. A novel lncRNA enhances autophagy to suppress extracellular matrix via modulating TP53INP1 in human trabecular meshwork cells under oxidative stress. Sci Rep 2025; 15:5049. [PMID: 39934637 PMCID: PMC11814328 DOI: 10.1038/s41598-024-81300-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 11/26/2024] [Indexed: 02/13/2025] Open
Abstract
Primary open-angle glaucoma (POAG) can result in irreversible blindness. As an important etiological factor, oxidative stress can elicit extraordinary increase of extracellular matrix (ECM) in trabecular meshwork-schlemm canal, to increase aqueous humor outflow resistance and elevate intraocular pressure. Although autophagy plays an important role in clearing ECM, the functions of long non-coding RNAs (lncRNAs) in autophagy induced by oxidative stress in human trabecular meshwork cells (HTMCs) remain unclear. In our study, oxidative stress induced the expression of ECM and autophagy in TMCs after H2O2 treatment. Meanwhile, a novel lncRNA ENST00000523905 and tumor protein 53-induced nuclear protein 1 (TP53INP1) were elevated in TMCs treated with H2O2. Similar to treatment with 3-MA (an inhibitor of autophagy), knocking-down the expression of TP53INP1 or ENST00000523905 could suppress the autophagy of TMCs induced by H2O2, which increased the level of ECM. Furthermore, the inhibition of ENST00000523905 decreased the expression of TP53INP1. ENST00000523905 could recruit and directly bind with CCAAT/enhancer (C/EBPβ), which can promote the expression of TP53INP1. Taken together, our findings demonstrated that ENST00000523905 may increase autophagy via enhancing TP53INP1 expression through binding with C/EBPβ, resulting in oxidative stress-induced decrease in ECM in HTMCs.
Collapse
Affiliation(s)
- Jinjun Tie
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
- Department of Ophthalmology, The Affiliated Hospital of Gui Zhou Medical University, Guiyang, Guizhou, China
| | - Junhong Guo
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
| | - Yijia Huang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
| | - Zihan Huang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
| | - Zhichao Yan
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China
| | - Jiemei Yuan
- Department of Ophthalmology, The Affiliated Hospital of Gui Zhou Medical University, Guiyang, Guizhou, China
| | - Xiaoli Shen
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China.
| | - Jiantao Wang
- Shenzhen Eye Hospital, Shenzhen Eye Institute, Jinan University, 18 Zetian Road, Futian District, Shenzhen, 518040, Guangdong, China.
| |
Collapse
|
3
|
Zhang X, Zheng C, Zhao J, Xu X, Yao J. LncRNA MEG3 regulates ferroptosis of lens epithelial cells via PTBP1/GPX4 axis to participate in age-related cataract. J Cell Physiol 2024; 239:e31330. [PMID: 38828927 DOI: 10.1002/jcp.31330] [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: 11/01/2023] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
Age-related cataract (ARC) is regarded as the principal cause of vision impairment among the aged. The regulatory role of long noncoding RNAs (LncRNAs) in ARC remains unclear. The lncRNA maternally expressed gene 3 (MEG3) has been reported to promote ARC progression, and the underlying mechanism was further investigated in this study. Lens epithelium samples were collected to verify the expression of MEG3. Lens epithelial cells (LECs) were treated with H2O2 to mimic microenvironment of ARC in vitro. Cell viability, reactive oxygen species, and ferroptosis were evaluated during the in viro experiments. In the present work, lncRNA MEG3 was highly expressed in ARC group, compared with normal group. MEG3 was induced, cell viability and glutathione peroxidase 4 (GPX4) level were inhibited, and ferroptosis was promoted in H2O2 treated LECs. LncRNA MEG3 silence reversed the effects of H2O2 on viability and ferroptosis in LECs. Thereafter, lncRNA MEG3 was found to bind to PTBP1 for GPX4 degradation. Silencing of GPX4 reversed the regulation of lncRNA MEG3 inhibition in H2O2-treated LECs. To sum up, lncRNA MEG3 exhibited high expression in ARC. In H2O2-induced LECs, inhibition of lncRNA MEG3 accelerated cell viability and repressed ferroptosis by interaction with PTBP1 for GPX4 messenger RNA decay. Targeting lncRNA MEG3 may be a novel treatment of ARC.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Clinical Forensic Medicine, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Chuanfei Zheng
- Department of Clinical Forensic Medicine, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Jiuhong Zhao
- Department of Human Anatomy and Histology, School of Fundamental Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xiaoming Xu
- Department of Clinical Forensic Medicine, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Jun Yao
- Department of Forensic Genetics and Biology, School of Forensic Medicine, China Medical University, Shenyang, China
| |
Collapse
|
4
|
Choudhary S, Singh MK, Kashyap S, Seth R, Singh L. Wnt/β-Catenin Signaling Pathway in Pediatric Tumors: Implications for Diagnosis and Treatment. CHILDREN (BASEL, SWITZERLAND) 2024; 11:700. [PMID: 38929279 PMCID: PMC11201634 DOI: 10.3390/children11060700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
The evolutionarily conserved Wnt signaling has a significant and diverse role in maintaining cell homeostasis and tissue maintenance. It is necessary in the regulation of crucial biological functions such as embryonal development, proliferation, differentiation, cell fate, and stem cell pluripotency. The deregulation of Wnt/β-catenin signaling often leads to various diseases, including cancer and non-cancer diseases. The role of Wnt/β-catenin signaling in adult tumors has been extensively studied in literature. Although the Wnt signaling pathway has been well explored and recognized to play a role in the initiation and progression of cancer, there is still a lack of understanding on how it affects pediatric tumors. This review discusses the recent developments of this signaling pathway in pediatric tumors. We also focus on understanding how different types of variations in Wnt signaling pathway contribute to cancer development and provide an insight of tissue specific mutations that lead to clinical progression of these tumors.
Collapse
Affiliation(s)
- Sahar Choudhary
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.C.); (R.S.)
| | | | - Seema Kashyap
- Department of Ocular Pathology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Rachna Seth
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.C.); (R.S.)
| | - Lata Singh
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.C.); (R.S.)
| |
Collapse
|
5
|
Ma X, Li X, Sun Q, Luan F, Feng J. Molecular Biological Research on the Pathogenic Mechanism of Retinoblastoma. Curr Issues Mol Biol 2024; 46:5307-5321. [PMID: 38920989 PMCID: PMC11202574 DOI: 10.3390/cimb46060317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Retinoblastoma (RB) is the most common intraocular malignant tumor in children, primarily attributed to the bi-allelic loss of the RB1 gene in the developing retina. Despite significant progress in understanding the basic pathogenesis of RB, comprehensively unravelling the intricate network of genetics and epigenetics underlying RB tumorigenesis remains a major challenge. Conventional clinical treatment options are limited, and despite the continuous identification of genetic loci associated with cancer pathogenesis, the development of targeted therapies lags behind. This review focuses on the reported genomic and epigenomic alterations in retinoblastoma, summarizing potential therapeutic targets for RB and providing insights for research into targeted therapies.
Collapse
Affiliation(s)
| | | | | | - Fuxiao Luan
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; (X.M.); (X.L.); (Q.S.)
| | - Jing Feng
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; (X.M.); (X.L.); (Q.S.)
| |
Collapse
|
6
|
Zhou L, Tong Y, Ho BM, Li J, Chan HYE, Zhang T, Du L, He JN, Chen LJ, Tham CC, Yam JC, Pang CP, Chu WK. Etiology including epigenetic defects of retinoblastoma. Asia Pac J Ophthalmol (Phila) 2024; 13:100072. [PMID: 38789041 DOI: 10.1016/j.apjo.2024.100072] [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: 02/24/2024] [Revised: 04/09/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Retinoblastoma (RB), originating from the developing retina, is an aggressive intraocular malignant neoplasm in childhood. Biallelic loss of RB1 is conventionally considered a prerequisite for initiating RB development in most RB cases. Additional genetic mutations arising from genome instability following RB1 mutations are proposed to be required to promote RB development. Recent advancements in high throughput sequencing technologies allow a deeper and more comprehensive understanding of the etiology of RB that additional genetic alterations following RB1 biallelic loss are rare, yet epigenetic changes driven by RB1 loss emerge as a critical contributor promoting RB tumorigenesis. Multiple epigenetic regulators have been found to be dysregulated and to contribute to RB development, including noncoding RNAs, DNA methylations, RNA modifications, chromatin conformations, and histone modifications. A full understanding of the roles of genetic and epigenetic alterations in RB formation is crucial in facilitating the translation of these findings into effective treatment strategies for RB. In this review, we summarize current knowledge concerning genetic defects and epigenetic dysregulations in RB, aiming to help understand their links and roles in RB tumorigenesis.
Collapse
Affiliation(s)
- Linbin Zhou
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Yan Tong
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Bo Man Ho
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jiahui Li
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Hoi Ying Emily Chan
- Medicine Programme Global Physician-Leadership Stream, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Tian Zhang
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Lin Du
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jing Na He
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Li Jia Chen
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Clement C Tham
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jason C Yam
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Chi Pui Pang
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China.
| | - Wai Kit Chu
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China.
| |
Collapse
|
7
|
Hazazi A, AlShehah AA, Khan FR, Hakami MA, Almarshadi F, Abalkhail A, Nassar SA, Almasoudi HH, Ali AA, Abu-Alghayth MH, Kukreti N, Binshaya AS. From diagnosis to therapy: The transformative role of lncRNAs in eye cancer management. Pathol Res Pract 2024; 254:155081. [PMID: 38211388 DOI: 10.1016/j.prp.2023.155081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/13/2024]
Abstract
The genomic era has brought about a transformative shift in our comprehension of cancer, unveiling the intricate molecular landscape underlying disease development. Eye cancers (ECs), encompassing diverse malignancies affecting ocular tissues, pose distinctive challenges in diagnosis and management. Long non-coding RNAs (lncRNAs), an emerging category of non-coding RNAs, are pivotal actors in the genomic intricacies of eye cancers. LncRNAs have garnered recognition for their multifaceted roles in gene expression regulation and influence on many cellular processes. Many studies support that the lncRNAs have a role in developing various cancers. Recent investigations have pinpointed specific lncRNAs associated with ECs, including retinoblastoma and uveal melanoma. These lncRNAs exert control over critical pathways governing tumor initiation, progression, and metastasis, endowing them with the ability to function as evaluation, predictive, and therapeutic indicators. The article aims to synthesize the existing information concerning the functions of lncRNAs in ECs, elucidating their regulatory mechanisms and clinical significance. By delving into the lncRNAs' expanding relevance in the modulation of oncogenic and tumor-suppressive networks, we gain a deeper understanding of the molecular complexities intrinsic to these diseases. In our exploration of the genomic intricacies of ECs, lncRNAs introduce a fresh perspective, providing an opportunity to function as clinical and therapeutic indicators, and they also have therapeutic benefits that show promise for advancing the treatment of ECs. This comprehensive review bridges the intricate relationship between lncRNAs and ECs within the context of the genomic era.
Collapse
Affiliation(s)
- Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | | | - Farhan R Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Fahad Almarshadi
- Department of Public Health, College of Public Health and Health Informatics, University of Ha'il, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Public Health and Health Informatics, Qassim University, Qassim, Saudi Arabia
| | - Somia A Nassar
- Department of Medical Laboratory Sciences, College of Applied medical sciences, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia; Department of Parasitology & Animal Diseases, National Research Centre, 33 Bohouth St., Dokki, Giza 12622, Egypt
| | - Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Amer Al Ali
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia
| | - Mohammed H Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Abdulkarim S Binshaya
- Department of Medical Laboratory Sciences, College of Applied medical sciences, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia.
| |
Collapse
|
8
|
Byroju VV, Nadukkandy AS, Cordani M, Kumar LD. Retinoblastoma: present scenario and future challenges. Cell Commun Signal 2023; 21:226. [PMID: 37667345 PMCID: PMC10478474 DOI: 10.1186/s12964-023-01223-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/12/2023] [Indexed: 09/06/2023] Open
Abstract
With an average incidence of 1 in every 18,000 live births, retinoblastoma is a rare type of intraocular tumour found to affect patients during their early childhood. It is curable if diagnosed at earlier stages but can become life-threateningly malignant if not treated timely. With no racial or gender predisposition, or even environmental factors known to have been involved in the incidence of the disease, retinoblastoma is often considered a clinical success story in pediatric oncology. The survival rate in highly developed countries is higher than 95% and they have achieved this because of the advancement in the development of diagnostics and treatment techniques. This includes developing the already existing techniques like chemotherapy and embarking on new strategies like enucleation, thermotherapy, cryotherapy, etc. Early diagnosis, studies on the etiopathogenesis and genetics of the disease are the need of the hour for improving the survival rates. According to the Knudson hypothesis, also known as the two hit hypothesis, two hits on the retinoblastoma susceptibility (RB) gene is often considered as the initiating event in the development of the disease. Studies on the molecular basis of the disease have also led to deciphering the downstream events and thus in the discovery of biomarkers and related targeted therapies. Furthermore, improvements in molecular biology techniques enhanced the development of efficient methods for early diagnosis, genetic counseling, and prevention of the disease. In this review, we discuss the genetic and molecular features of retinoblastoma with a special emphasis on the mutation leading to the dysregulation of key signaling pathways involved in cell proliferation, DNA repair, and cellular plasticity. Also, we describe the classification, clinical and epidemiological relevance of the disease, with an emphasis on both the traditional and innovative treatments to tackle retinoblastoma. Video Abstract.
Collapse
Affiliation(s)
- Vishnu Vardhan Byroju
- Department of Biochemistry, American International Medical University, Gros Islet, St. Lucia, USA
| | | | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, and Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Lekha Dinesh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad, India.
| |
Collapse
|
9
|
Yan L, Guo L. Exercise-regulated white adipocyte differentitation: An insight into its role and mechanism. J Cell Physiol 2023; 238:1670-1692. [PMID: 37334782 DOI: 10.1002/jcp.31056] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023]
Abstract
White adipocytes play a key role in the regulation of fat mass amount and energy balance. An appropriate level of white adipocyte differentiation is important for maintaining metabolic homeostasis. Exercise, an important way to improve metabolic health, can regulate white adipocyte differentiation. In this review, the effect of exercise on the differentiation of white adipocytes is summarized. Exercise could regulate adipocyte differentiation in multiple ways, such as exerkines, metabolites, microRNAs, and so on. The potential mechanism underlying the role of exercise in adipocyte differentiation is also reviewed and discussed. In-depth investigation of the role and mechanism of exercise in white adipocyte differentiation would provide new insights into exercise-mediated improvement of metabolism and facilitate the application of exercise-based strategy against obesity.
Collapse
Affiliation(s)
- Linjing Yan
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences (Shanghai University of Sport), Ministry of Education, Shanghai, China
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- Key Laboratory of Exercise and Health Sciences (Shanghai University of Sport), Ministry of Education, Shanghai, China
| |
Collapse
|
10
|
Long non-coding RNAs involved in retinoblastoma. J Cancer Res Clin Oncol 2023; 149:401-421. [PMID: 36305946 DOI: 10.1007/s00432-022-04398-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/05/2022] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Retinoblastoma (RB) is the most common childhood tumor that can occur in the retina and develop in a sporadic or heritable form. Although various traditional treatment options have been used for patients with RB, identifying novel strategies for childhood cancers is necessary. MATERIAL AND METHODS Recently, molecular-based targeted therapies have opened a greater therapeutic window for RB. Long non-coding RNAs (lncRNAs) presented a potential role as a biomarker for the detection of RB in various stages. CONCLUSION LncRNAs by targeting several miRNA/transcription factors play critical roles in the stimulation or suppression of RB. In this review, we summarized recent progress on the functions of tumor suppressors or oncogenes lncRNAs in RB.
Collapse
|
11
|
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
|
12
|
He L, Tang L, Wang R, Liu L, Zhu P, Jiang K, Tu G. Long noncoding RNA KB-1980E6.3 promotes breast cancer progression through the PI3K/AKT signalling pathway. Pathol Res Pract 2022; 234:153891. [DOI: 10.1016/j.prp.2022.153891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/26/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
13
|
Fernandez-Diaz D, Rodriguez-Vidal C, Silva-Rodríguez P, Paniagua L, Blanco-Teijeiro MJ, Pardo M, Piñeiro A, Bande M. Applications of Non-Coding RNAs in Patients With Retinoblastoma. Front Genet 2022; 13:842509. [PMID: 35432447 PMCID: PMC9008704 DOI: 10.3389/fgene.2022.842509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
Retinoblastoma (RB) is the most common primary intraocular malignancy in childhood. In the carcinogenic process of neoplasms such as RB, the role of non-coding RNAs (ncRNAs) has been widely demonstrated recently. In this review, we aim to provide a clinical overview of the current knowledge regarding ncRNAs in relation to RB. Although ncRNAs are now considered as potential diagnostic biomarkers, prognostic factors, and therapeutic targets, further studies will facilitate enhanced understanding of ncRNAs in RB physiopathology and define the roles ncRNAs can play in clinical practice.
Collapse
Affiliation(s)
- Daniel Fernandez-Diaz
- Department of Ophthalmology, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | | | - Paula Silva-Rodríguez
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Clinical University Hospital, Santiago de Compostela, Spain
| | - Laura Paniagua
- Department of Ophthalmology, University Hospital of Coruña, A Coruña, Spain
| | - María José Blanco-Teijeiro
- Department of Ophthalmology, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - María Pardo
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Grupo Obesidómica, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Antonio Piñeiro
- Department of Ophthalmology, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Manuel Bande
- Department of Ophthalmology, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Tumores Intraoculares en el Adulto, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- *Correspondence: Manuel Bande,
| |
Collapse
|
14
|
Zhang J, Liu X, Gao Y. The long noncoding RNA MEG3 regulates Ras-MAPK pathway through RASA1 in trophoblast and is associated with unexplained recurrent spontaneous abortion. Mol Med 2021; 27:70. [PMID: 34238211 PMCID: PMC8265043 DOI: 10.1186/s10020-021-00337-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/30/2021] [Indexed: 12/28/2022] Open
Abstract
Background Maternally Expressed Gene 3 (MEG3) is expressed at low levels in placental villi during preeclampsia; however, its roles in unexplained recurrent spontaneous abortion (URSA) remain unclear. In this study, we aimed to explore the relationship between MEG3 and URSA. Methods The differentially expressed lncRNAs (MEG3) and its downstream genes (RASA1) were identified using bioinformatics analysis of Genomic Spatial Event (GSE) database. The expression levels of MEG3 in embryonic villis (with gestational ages of 49–63 days) and primary trophoblasts were determined using quantitative RT-PCR assay. A mouse model of Embryo implantation, Cell Counting Kit-8 (CCK-8), flow cytometry, and Transwell migration assays were performed to determine the implantation, proliferative, apoptotic, and invasive capacities of trophoblast. The level of phosphorylated core proteins in the RAS-MAPK pathway were analyzed using Western blot assay. The mechanisms of MEG3 in the regulation of RASA1 were studied by RNA pulldown, RNA immunoprecipitation (RIP), DNA pulldown, and chromatin immunoprecipitation (ChIP) assays. Results MEG3 had a low expression level in embryonic villis of 102 URSA patients compared with those of 102 normal pregnant women. MEG3 could promote proliferation and invasion, inhibit the apoptosis of primary trophoblast of URSA patients (PT-U cells), as well as promote embryo implantation of mouse. Besides, MEG3 also promoted the phosphorylation of rapidly accelerated fibrosarcoma (Raf), mitogen-activated protein kinase kinase (MEK), and extracellular-signal-regulated kinase (ERK) proteins. The results of RNA pull down and RIP assays showed that MEG3 bound with the enhancer of zeste homolog 2 (EZH2). The DNA pulldown assay revealed that MEG3 could bind to the promoter sequence of the RAS P21 Protein Activator 1 (RASA1) gene. Further, the ChIP assay showed that MEG3 promoted the binding of EZH2 to the promoter region of the RASA1 gene. Conclusions The inactivation of MEG3 in embryonic villi association with URSA; MEG3 inhibited the expression of RASA1 by mediating the histone methylation of the promoter of RASA1 gene by EZH2, thereby activating the RAS-MAPK pathway and enhancing the proliferative and invasive capacities of trophoblasts. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00337-9.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Obstetrics and Gynecology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, 518020, People's Republic of China
| | - Xinqiong Liu
- Department of Obstetrics and Gynecology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, 518020, People's Republic of China
| | - Yali Gao
- Department of Ophthalmology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, 518020, People's Republic of China.
| |
Collapse
|
15
|
Liu J, Qu X. The roles of long non-coding RNAs in ocular diseases. Exp Eye Res 2021; 207:108561. [PMID: 33812869 DOI: 10.1016/j.exer.2021.108561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 03/11/2021] [Accepted: 03/25/2021] [Indexed: 11/19/2022]
Abstract
In recent years, lncRNAs have been shown to regulate gene expression at the epigenetic, transcriptional and translational level, thus exerting various functions in biological and pathological processes involving cell proliferation, apoptosis, cell cycle and immune response. An increasing number of researches have unveiled that lncRNAs are dysregulated in pathogenesis and the development of different ocular diseases, such as glaucoma, cataract, retinal disease and ocular tumors. Also, it has been reported that lncRNAs may exert significant roles in various ocular diseases. Here, we summarized the functions of lncRNAs on relevant ocular diseases and further clarified their mechanisms. Here, several previous studies with detailed information of lncRNAs which have been proved to be the diagnostic or prognostic biomarkers and potential therapeutic targets were included. Also, it is our hope to provide a thorough knowledge of the functions of lncRNAs in eye diseases and the methods by which lncRNAs can influence ocular diseases.
Collapse
Affiliation(s)
- Jinlu Liu
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, No.4, Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Xiaohan Qu
- Department of Thoracic Surgery, The First Hospital of China Medical University, No.155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
16
|
Gao Y, Luo X, Zhang J. Sp1-mediated up-regulation of lnc00152 promotes invasion and metastasis of retinoblastoma cells via the miR-30d/SOX9/ZEB2 pathway. Cell Oncol (Dordr) 2021; 44:61-76. [PMID: 32488851 DOI: 10.1007/s13402-020-00522-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2020] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Previously, we found that long non-coding RNA (lncRNA) MEG3 may act as a tumour suppressor in retinoblastoma. Overall, however, little is known about the role of lncRNAs in retinoblastoma. Here, we aimed to determine the expression and clinical significance of lnc00152 in retinoblastoma. METHODS Lnc00152 and its downstream targets were selected using GEO datasets. The level of lnc00152 in primary patient samples was determined using RT-qPCR. Odds ratios of invasion and metastasis were calculated using logistic regression analysis. Recurrence-free survival was assessed using Cox regression analysis. Scratch wound healing, transwell and tumorigenesis assays were used to determine migration and invasion abilities of retinoblastoma cells in vitro and in vivo. Levels of EMT-related proteins were measured using Western blotting. Binding sites between lnc00152 and its targets were validated using dual-luciferase reporter and RNA pull-down assays. Lnc00152 activating transcription factors were determined using ChIP assays. RESULTS We found that Lnc00152 was significantly up-regulated in retinoblastoma tumour tissues, and was a risk factor for tumour invasion, metastasis and recurrence. Lnc00152 overexpressing retinoblastoma cells exhibited a tendency to transform into mesenchymal cells, with significantly increased migration and invasion capacities, significantly decreased E-cadherin expression levels, and significantly increased N-cadherin, SOX9 and ZEB2 expression levels. In addition, we found that lnc00152, which was activated by Sp1, could inhibit miR-30d as an endogenous miRNA 'sponge', thereby regulating the expression of SOX9 and ZEB2. CONCLUSIONS Our data indicate that Lnc00152 may be associated with retinoblastoma invasion, metastasis and prognosis. In addition, we conclude that Lnc00152, which can be activated by Sp1, can induce EMT via the miR-30d/SOX9/ZEB2 pathway and, by doing so, promote the invasion and metastasis of retinoblastoma cells.
Collapse
Affiliation(s)
- Yali Gao
- Department of Ophthalmology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, 518020, Shenzhen, People's Republic of China
| | - Xiaoling Luo
- Department of Ophthalmology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, 518020, Shenzhen, People's Republic of China
| | - Jun Zhang
- Department of Obstetrics and Gynaecology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, 518020, Shenzhen, People's Republic of China.
| |
Collapse
|
17
|
Wang J, Wang S, Chen L, Tan J. SCARA5 suppresses the proliferation and migration, and promotes the apoptosis of human retinoblastoma cells by inhibiting the PI3K/AKT pathway. Mol Med Rep 2021; 23:202. [PMID: 33495818 PMCID: PMC7821225 DOI: 10.3892/mmr.2021.11841] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
Retinoblastoma (RB) is the most common ocular malignancy that occurs during childhood. Scavenger receptor class A member 5 (SCARA5) is considered to function as an anti-oncogene in several types of malignant tumor. The present study investigated the functional role and underlying mechanism of SCARA5 in human RB cells. Reverse transcription-quantitative PCR and western blotting were used to detect the relative expression levels of SCARA5 in four human RB cell lines. In addition, transfection was performed to either knockdown or induce overexpression of SCARA5 in human RB Y79 cells. The proliferation, migration and apoptosis of RB cells was then measured by Cell Counting Kit 8 assay, 5-ethynyl-2′-deoxyuridine assay, clone formation assay, Transwell assay, Hoechst staining and TUNEL staining, respectively. Western blotting was performed to detect changes in the expression levels of key proteins involved in the PI3K/AKT and apoptotic pathways. The present study revealed that SCARA5 was expressed at lower levels in four tumorigenic human RB cell lines compared with in a human retinal pigment non-tumorigenic cell line. Functional analysis demonstrated that overexpression of SCARA5 decreased the proliferation and migration, and promoted the apoptosis of human RB cells in vitro, whereas in vivo experiments revealed a decrease in RB progression following SCARA5 overexpression. In addition, overexpression of SCARA5 inhibited phosphorylated (p)-PI3K and p-AKT expression, and knockdown of SCARA5 increased p-PI3K and p-AKT expression; however, no changes in total PI3K and AKT expression were observed. Bcl-2 exhibited similar changes in expression to those displayed by p-PI3K and p-AKT, whereas Bax and cleaved caspase-3 exhibited trends in expression that were the opposite to those shown by p-PI3K and p-AKT. In conclusion, the present results demonstrated that SCARA5 could inhibit the proliferation and promote the apoptosis of RB cell lines by suppressing the PI3K/AKT signaling pathway, thus suggesting a novel strategy for treating RB.
Collapse
Affiliation(s)
- Jinwei Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Sha Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lu Chen
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jia Tan
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
18
|
Xu L, Zhu S, Tang A, Liu W. LncRNA MBLN1-AS1 inhibits the progression of retinoblastoma through targeting miR-338-5p-Wnt/β-catenin signaling pathway. Inflamm Res 2021; 70:217-227. [PMID: 33454803 DOI: 10.1007/s00011-020-01432-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/07/2020] [Accepted: 12/13/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE AND DESIGN Retinoblastoma is the most common primary intraocular malignancy of childhood, which brings a heavy burden to the countries across the world, especially the developing countries. It has been shown that lncRNA muscleblind-like 1 antisense RNA 1 (MBNL1-AS1) exerts anti-tumor effects in various cancers, including bladder cancer, papillary thyroid cancer, and retinoblastoma. In the present study, we hypothesized that MBNL1-AS1 might play a protective role against retinoblastoma. METHODS The expression of MBNL1-AS1 and its potential target miR-338-5p were evaluated in retinoblastoma cell line by real-time quantitative PCR and western blot. The involvement of MBNL1-AS1-miR-338-5p in the cell proliferation was evaluated by cell counting kit-8 (CCK8), and colony formation assay. The cell migration was evaluated by Transwell assay in Y79 cells, a retinoblastoma cell line. The involvement of MBNL1-AS1-miR-338-5p in tumor formation was also evaluated in mice. RESULTS It was found that MBNL1-AS1 overexpression inhibited proliferation and migration in Y79 cells. In addition, the inhibitory effects of MBNL1-AS1 on Y79 cells were significantly reversed in the presence of miR-338-5p mimics, and MBNL1-AS1 overexpression significantly decreased miR-338-5p level in Y79 cells. Furthermore, MBNL1-AS1 overexpression significantly inhibited Wnt/β-catenin signaling pathway, and this inhibitory effect was almost lost in the presence of miR-338-5p mimics. Finally, our in vivo study showed that MBNL1-AS1 overexpression significantly inhibited Y79-induced retinoblastoma in mice, and this inhibitory effect was lost in the presence of miR-338-5p mimics. CONCLUSION Our study shows that MBNL1-AS1 exerts its anti-tumor effect by targeting miR-338-5p, thereby inactivating wnt/β-catenin signaling pathway in retinoblastoma.
Collapse
Affiliation(s)
- Lei Xu
- Department of Ophthalmology, The First Affiliated Hospital of Gannan Medical University, No. 23 Qingnian Road, Zhanggong District, Ganzhou, 341000, Jiangxi Province, China
| | - Shenyu Zhu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi Province, China
| | - Aidong Tang
- Department of Ophthalmology, The First Affiliated Hospital of Gannan Medical University, No. 23 Qingnian Road, Zhanggong District, Ganzhou, 341000, Jiangxi Province, China
| | - Wanrong Liu
- Department of Ophthalmology, The First Affiliated Hospital of Gannan Medical University, No. 23 Qingnian Road, Zhanggong District, Ganzhou, 341000, Jiangxi Province, China.
| |
Collapse
|
19
|
Lyv X, Wu F, Zhang H, Lu J, Wang L, Ma Y. Long Noncoding RNA ZFPM2-AS1 Knockdown Restrains the Development of Retinoblastoma by Modulating the MicroRNA-515/HOXA1/Wnt/β-Catenin Axis. Invest Ophthalmol Vis Sci 2021; 61:41. [PMID: 32561925 PMCID: PMC7415309 DOI: 10.1167/iovs.61.6.41] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose The tumor-initiating function of long non-coding RNA (lncRNA), zinc finger protein multitype 2 antisense RNA 1 (ZFPM2-AS1) was reported in lung cancer, yet the relevance of ZFPM2-AS1 in retinoblastoma (RB), a malignancy representing 2.5% to 4% incidence of cancers among children, has not been elucidated. Thus, we attempted to assess the effect of ZFPM2-AS1 and underlying mechanism in RB progression. Methods First, comparing the differentially expressed lncRNAs in normal retinal tissues as well as RB tissues, the target lncRNA ZFPM2-AS1 was screened out. We then assayed the ZFPM2-AS1 expression in three RB cell lines, and carried out methylthiazol tetrazolium (MTT), transwell assays, and flow cytometric analyses to examine the role of si-ZFPM2-AS1 on cell behaviors. Following online database predication, the correlations between ZFPM2-AS1 and microR-515 (miR-515) or homeobox A1 (HOXA1) were corroborated by dual-luciferase reporter gene assays. Quantitative real-time PCR along with Western blot assays was fulfilled to ascertain the expression of relevant genes. Results ZFPM2-AS1 was significantly overexpressed in RB tissues and cell lines, and ZFPM2-AS1 silencing curtailed the growth and metastasis of RB cells both in vitro and in vivo. Bioinformatic websites and dual-luciferase reporter gene assays disclosed that ZFPM2-AS1 might perform as a competing endogenous RNA for miR-515 and positively correlate with HOXA1 to activate the Wnt/β-catenin signaling pathway. Conclusions Altogether, these data demonstrated that ZFPM2-AS1 interacted with HOXA1 to promote RB development via mediating miR-515, establishing a promising therapeutic biomarker for RB and prognosis.
Collapse
|
20
|
Cáceres-Durán MÁ, Ribeiro-dos-Santos Â, Vidal AF. Roles and Mechanisms of the Long Noncoding RNAs in Cervical Cancer. Int J Mol Sci 2020; 21:ijms21249742. [PMID: 33371204 PMCID: PMC7766288 DOI: 10.3390/ijms21249742] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/03/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Cervical cancer (CC) continues to be one of the leading causes of death for women across the world. Although it has been determined that papillomavirus infection is one of the main causes of the etiology of the disease, genetic and epigenetic factors are also required for its progression. Among the epigenetic factors are included the long noncoding RNAs (lncRNAs), transcripts of more than 200 nucleotides (nt) that generally do not code for proteins and have been associated with diverse functions such as the regulation of transcription, translation, RNA metabolism, as well as stem cell maintenance and differentiation, cell autophagy and apoptosis. Recently, studies have begun to characterize the aberrant regulation of lncRNAs in CC cells and tissues, including Homeobox transcript antisense RNA (HOTAIR), H19, Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), Cervical Carcinoma High-Expressed 1 (CCHE1), Antisense noncoding RNA in the inhibitors of cyclin-dependent kinase 4 (ANRIL), Growth arrest special 5 (GAS5) and Plasmacytoma variant translocation 1 (PVT1). They have been associated with several disease-related processes such as cell growth, cell proliferation, cell survival, metastasis and invasion as well as therapeutic resistance, and are novel potential biomarkers for diagnosis and prognosis in CC. In this review, we summarize the current literature regarding the knowledge we have about the roles and mechanisms of the lncRNAs in cervical neoplasia.
Collapse
Affiliation(s)
- Miguel Ángel Cáceres-Durán
- Laboratory of Human and Medical Genetics, Institute of Biological Sciences, Graduate Program of Genetics and Molecular Biology, Federal University of Pará, Belém 66075-110, Brazil; (M.Á.C.-D.); (Â.R.-d.-S.)
| | - Ândrea Ribeiro-dos-Santos
- Laboratory of Human and Medical Genetics, Institute of Biological Sciences, Graduate Program of Genetics and Molecular Biology, Federal University of Pará, Belém 66075-110, Brazil; (M.Á.C.-D.); (Â.R.-d.-S.)
- Graduate Program in Oncology and Medical Sciences, Center of Oncology Researches, Federal University of Pará, Belém 66073-005, Brazil
| | - Amanda Ferreira Vidal
- Laboratory of Human and Medical Genetics, Institute of Biological Sciences, Graduate Program of Genetics and Molecular Biology, Federal University of Pará, Belém 66075-110, Brazil; (M.Á.C.-D.); (Â.R.-d.-S.)
- Correspondence: ; Tel.: +55-91-3201-7843
| |
Collapse
|
21
|
Gao Y, Luo X, Zhang J. Activation of LncRNA FOXD2-AS1 by H3K27 acetylation regulates VEGF-A expression by sponging miR-205-5p in recurrent pterygium. J Cell Mol Med 2020; 24:14139-14151. [PMID: 33098266 PMCID: PMC7754060 DOI: 10.1111/jcmm.16024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 12/25/2022] Open
Abstract
LncRNA FOXD2‐AS1 is abnormally expressed in many diseases. However, the molecular mechanisms whereby FOXD2‐AS1 is involved in recurrent pterygium remain unknown. Here, qRT‐PCR was performed to quantify FOXD2‐AS1 expression, while CCK‐8, flow cytometer and neoplasm xenograft assays were used to investigate its function. Dual‐luciferase reporter, RIP and RNA pull‐down assays were conducted to address the relationship between FOXD2‐AS1, miR‐205‐5p and VEGF‐A, while ChIP assays were used to detect H3K27 acetylation at the FOXD2‐AS1 promoter. FOXD2‐AS1 expression was up‐regulated in recurrent pterygium tissues. Moreover, a high FOXD2‐AS1 expression was associated with advanced stages, increased microvessel density and shorter recurrent‐free survival. In addition, ROC analysis showed that FOXD2‐AS1 is a valid predictor of recurrent pterygium. Furthermore, we show that FOXD2‐AS1 induced proliferation and inhibited apoptosis in a cell line derived from recurrent pterygia (HPF‐R) at least partially through the regulation of the miR‐205‐VEGF pathway. In addition, the up‐regulation of FOXD2‐AS1 was attributed to the H3K27 acetylation at the promoter region. In conclusion, FOXD2‐AS1 is activated via its H3K27 acetylation and regulates VEGF‐A expression by sponging miR‐205‐5p in recurrent pterygium. Our results may provide a basis for the development of new therapeutic targets and biomarkers for recurrent pterygium.
Collapse
Affiliation(s)
- Yali Gao
- Department of Ophthalmology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Xiaoling Luo
- Department of Ophthalmology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Jun Zhang
- Department of Obstetrics and Gynecology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, China
| |
Collapse
|
22
|
Wang Y, Wang J, Hao H, Luo X. lncRNA KCNQ1OT1 promotes the proliferation, migration and invasion of retinoblastoma cells by upregulating HIF-1α via sponging miR-153-3p. J Investig Med 2020; 68:1349-1356. [DOI: 10.1136/jim-2020-001431] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2020] [Indexed: 12/19/2022]
Abstract
It is reported that lncRNA KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) is oncogenic in many cancers. This work aimed at probing into its expression and biological functions in retinoblastoma (RB) as well as its regulatory effects on miR-153-3p and hypoxia-inducible factor-1α (HIF-1α). In our study, RB samples in pair were collected, and quantitative real-time PCR (qRT-PCR) was employed for examining the expression levels of KCNQ1OT1, miR-153-3p and HIF-1α. KCNQ1OT1 short hairpin RNAs were transfected into SO-Rb50 and HXO-RB44 cell to inhibit the expression of KCNQ1OT1. The proliferative activity, colony formation ability and apoptosis were examined through cell counting kit-8 assay, colony formation assays, Transwell assay and flow cytometry, respectively. qRT-PCR and western blot analysis were used for analyzing the changes of miR-153-3p and HIF-1α induced by KCNQ1OT1. The regulatory relationships between miR-153-3p and KCNQ1OT1, miR-153-3p and HIF-1α were examined by dual luciferase reporter gene assay and RNA-binding protein immunoprecipitation assay. The results of our study showed that KCNQ1OT1 expression was markedly enhanced in RB tissue samples, and KCNQ1OT1 knockdown had an inhibitory effect on the proliferation, migration, invasion and viability of RB cells. There were two validated binding sties between KCNQ1OT1 and miR-153-3p, and KCNQ1OT1 negatively regulated the expression of miR-153-3p in RB cells. HIF-1α was a target gene of miR-153-3p, and could be positively regulated by KCNQ1OT1. In conclusion, our study indicates that KCNQ1OT1 can increase the malignancy of RB cells via regulating miR-153-3p/HIF-1α axis.
Collapse
|
23
|
高 亚, 罗 小, 孟 婷, 朱 敏, 田 渼, 陆 晓. [DNMT1 protein promotes retinoblastoma proliferation by silencing MEG3 gene]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1239-1245. [PMID: 32990237 PMCID: PMC7544569 DOI: 10.12122/j.issn.1673-4254.2020.09.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To investigate whether DNMT1 protein induces retinoblastoma proliferation by silencing MEG3 gene. METHODS Two retinoblastoma cell lines (HXO-RB44 and SO-RB50) and a normal human retinal pigment epithelial (RPE) cell line were transfected with the plasmid pcDNA-DNMT1 or si-DNMT1 for up-regulating or interference of DNMT1 expression, and with pcDNA-MEG3 or si-MEG3 for up-regulating or interference of MEG3 expression. Western blotting was used to detect the changes in the expression of DNMT1 protein in the transfected cells, and CCK-8 and EdU assays were used to detect the changes in cell proliferation. Real-time quantitative PCR (qRT-PCR) was performed to detect MEG3 expression in SO-RB50 and HXO-RB44 cells after transfection, and the methylation level of MEG3 gene promoter after interference of DNMT1 expression was detected using methylation-specific PCR. RESULTS SO-RB50 and HXO-RB44 cells showed significantly increased expression of DNMT1 protein as compared with normal RPE cells (P < 0.05). In HXO-RB44 cells, transfection with pcDNADNMT1 resulted in significantly increased expression of DNMT1 protein, enhanced cell proliferation ability, and significantly reduced expression of MEG3 (P < 0.05). In SO-RB50 cells, transfection with si-DNMT1 significantly reduced the expression of DNMT1 protein, suppressed the cell proliferation, and increased MEG3 expression (P < 0.05). Interference of DNMT1 significantly reduced the methylation level of MEG3 gene promoter. After reversing the regulatory effect of DNMT1 on MEG3 gene, DNMT1 protein showed significantly weakened ability to regulate retinoblastoma cell proliferation (P < 0.05). CONCLUSIONS In retinoblastoma cells, the up-regulation of DNMT1 protein induces promoter methylation and inactivation of MEG3 gene and eventually leads to abnormal cell proliferation.
Collapse
Affiliation(s)
- 亚莉 高
- 深圳市人民医院//暨南大学第二临床医学院眼科,广东 深圳 518020Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - 小玲 罗
- 深圳市人民医院//暨南大学第二临床医学院眼科,广东 深圳 518020Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - 婷 孟
- 深圳市人民医院//暨南大学第二临床医学院眼科,广东 深圳 518020Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - 敏娟 朱
- 深圳市人民医院//暨南大学第二临床医学院眼科,广东 深圳 518020Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - 渼雯 田
- 深圳市人民医院//暨南大学第二临床医学院眼科,广东 深圳 518020Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - 晓和 陆
- 南方医科大学珠江医院眼科,广东 广州 510280Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
24
|
Tu J, Huang W, Zhang W, Mei J, Zhu C. The emerging role of lncRNAs in chondrocytes from osteoarthritis patients. Biomed Pharmacother 2020; 131:110642. [PMID: 32927251 DOI: 10.1016/j.biopha.2020.110642] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/07/2020] [Accepted: 08/16/2020] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play important roles in many physiological and pathological processes, including osteoarthritis (OA). Recent studies have demonstrated that lncRNAs are involved in the pathogenesis of OA by affecting various essential cellular features of chondrocytes, such as proliferation, apoptosis, inflammation, and degradation of the extracellular matrix (ECM). However, there are only a limited number of studies in this area, indicating that the role of lncRNAs in OA may have been overlooked. The aim of this literature review is to summarize the versatile roles and molecular mechanisms of lncRNAs in chondrocytes involved in OA. At the end of this article, the function of the lncRNA HOX transcript antisense RNA (HOTAIR) in chondrocytes in OA is highlighted. Because lncRNAs affect proliferation, apoptosis, inflammatory responses, and ECM degradation by chondrocytes in OA, they may serve as potential biomarkers or therapeutic targets for the diagnosis or treatment of OA. The specific role and related mechanisms of lncRNAs in OA warrants further investigation.
Collapse
Affiliation(s)
- Jiajie Tu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China.
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Weiwei Zhang
- Departments of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jiawei Mei
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Chen Zhu
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
25
|
Tu Y, Xie L, Chen L, Yuan Y, Qin B, Wang K, Zhu Q, Ji N, Zhu M, Guan H. Long non-coding RNA MEG3 promotes cataractogenesis by upregulating TP53INP1 expression in age-related cataract. Exp Eye Res 2020; 199:108185. [PMID: 32841649 DOI: 10.1016/j.exer.2020.108185] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/04/2020] [Accepted: 08/01/2020] [Indexed: 01/02/2023]
Abstract
Age-related cataract (ARC) is the leading cause of visual impairment or even blindness among the aged population globally. Long non-coding RNA (LncRNA) has been proven to be the potential regulator of ARC. The latest study reveals that maternally expressed gene 3 (MEG3) promotes the apoptosis and inhibits the proliferation of multiple cancer cells. However, the expression and role of MEG3 in ARC are unclear. In this study, we investigated the effects of MEG3 in ARC and explored the regulatory mechanisms underlying these effects. We observed that MEG3 expression was up-regulated in the age-related cortical cataract (ARCC) lens capsules and positively correlated with the histological degree of ARCC. The pro-apoptosis protein, active caspase-3 and Bax increased in the anterior lens capsules of ARCC tissue, while the anti-apoptotic protein Bcl-2 decreased compared to normal lens. Knockdown of MEG3 increased the viability and inhibited the apoptosis of LECs upon the oxidative stress induced by H2O2. MEG3 was localized in both nucleus and cytoplasm in LECs. MEG3 facilitated TP53INP1 expression via acting as miR-223 sponge and promoting P53 expression. Additionally, TP53INP1 knockdown alleviated H2O2-induced lens turbidity. In summary, MEG3 promoted ARC progression by up-regulating TP53INP1 expression through suppressing miR-223 and promoting P53 expression, which would provide a novel insight into the pathogenesis of ARC.
Collapse
Affiliation(s)
- Yuanyuan Tu
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China; Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Laiqing Xie
- Department of Ophthalmology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lili Chen
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - You Yuan
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bai Qin
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Kun Wang
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiujian Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Na Ji
- Department of Ophthalmology, The Affiliated Eye Hospital of Suzhou Vocational Health College, Suzhou, Jiangsu, China
| | - Manhui Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Huaijin Guan
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
26
|
Abstract
Genomic imprinting is a parent-of-origin dependent phenomenon that restricts transcription to predominantly one parental allele. Since the discovery of the first long noncoding RNA (lncRNA), which notably was an imprinted lncRNA, a body of knowledge has demonstrated pivotal roles for imprinted lncRNAs in regulating parental-specific expression of neighboring imprinted genes. In this Review, we will discuss the multiple functionalities attributed to lncRNAs and how they regulate imprinted gene expression. We also raise unresolved questions about imprinted lncRNA function, which may lead to new avenues of investigation. This Review is dedicated to the memory of Denise Barlow, a giant in the field of genomic imprinting and functional lncRNAs. With her passion for understanding the inner workings of science, her indominable spirit and her consummate curiosity, Denise blazed a path of scientific investigation that made many seminal contributions to genomic imprinting and the wider field of epigenetic regulation, in addition to inspiring future generations of scientists.
Collapse
Affiliation(s)
- William A. MacDonald
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mellissa R. W. Mann
- Department of Obstetrics, Gynaecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
27
|
LincRNA-ROR is activated by H3K27 acetylation and induces EMT in retinoblastoma by acting as a sponge of miR-32 to activate the Notch signaling pathway. Cancer Gene Ther 2020; 28:42-54. [PMID: 32439866 DOI: 10.1038/s41417-020-0181-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/19/2022]
Abstract
Recent studies have suggested that lincRNA-ROR is involved in the tumorigenesis of different types of cancers. However, the role of lincRNA-ROR in retinoblastoma has not been determined. We investigated lincRNA-ROR levels in 58 retinoblastoma and adjacent non-tumor tissues by quantitative reverse transcription PCR. Recurrence-free survival was analyzed using Cox regression analyses. Cell migration and invasion abilities were detected by wound-healing, Transwell invasion, and bioluminescence imaging assays. Western blotting was performed to detect epithelial-mesenchymal transition markers. Interactions between lincRNA-ROR, miR-32-5p, and Notch1 were confirmed by Luciferase, RNA pull-down, and RIP assays. Histone acetylation was detected by chromatin immunoprecipitation assays. We showed that lincRNA-ROR was significantly upregulated in retinoblastoma tissues, and overexpression of lincRNA-ROR was significantly correlated with optic nerve invasion, nodal or distant metastasis, and recurrence. We also showed that lincRNA-ROR is a critical promoter of retinoblastoma cell metastasis, both in vivo and in vitro. Further, we demonstrated that lincRNA-ROR activates the Notch signaling pathway by acting as a sponge of miR-32-5p. Upregulation of lincRNA-ROR was attributed to the CBP-mediated H3K27 acetylation at the promoter region. Our results reveal a potential competing endogenous RNA regulatory pathway, in which lincRNA-ROR modulates the epithelial-mesenchymal transition program by competitively binding to endogenous miR-32-5p and regulating Notch signaling pathway activity in retinoblastoma cells, which may provide new insights into novel molecular therapeutic targets for retinoblastoma.
Collapse
|
28
|
Sun X, Shen H, Liu S, Gao J, Zhang S. Long noncoding RNA SNHG14 promotes the aggressiveness of retinoblastoma by sponging microRNA‑124 and thereby upregulating STAT3. Int J Mol Med 2020; 45:1685-1696. [PMID: 32236565 PMCID: PMC7169960 DOI: 10.3892/ijmm.2020.4547] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/30/2020] [Indexed: 12/11/2022] Open
Abstract
A long noncoding RNA called small nucleolar RNA host gene 14 (SNHG14) has been validated as a key regulator of cellular processes in multiple types of human cancer. However, to the best of our knowledge, the expression status and specific roles of SNHG14 in retinoblastoma (RB) have not been studied. The aims of the present study were to determine the expression status of SNHG14 in RB, assess the effects of SNHG14 on malignant characteristics of RB cells and investigate the mechanisms of action of SNHG14 in RB. SNHG14 expression levels in RB tissue samples and cell lines were measured by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Cell proliferation, apoptosis, migration and invasion in vitro, and tumor growth in vivo were quantitated by the Cell Counting Kit‑8 assay, flow cytometry, migration and invasion assays, and mouse tumor xenograft experiments, respectively. The target microRNA (miRNA) of SNHG14 was predicted by bioinformatics analysis and was subsequently validated by a luciferase reporter assay, RNA immunoprecipitation (RIP) assay, RT‑qPCR, and western blot analysis. SNHG14 was identified to be significantly overexpressed in RB tissues and cell lines. SNHG14 overexpression was markedly associated with the intraocular international retinoblastoma classification stage, optic nerve invasion, and differentiation grade among patients with RB. The patients in the SNHG14 high‑expression group exhibited shorter overall survival compared with the SNHG14 low‑expression group. Functional analysis revealed that SNHG14 silencing inhibited cell proliferation, migration and invasion, and increased apoptosis in vitro, and decreased tumor growth in vivo. SNHG14 directly interacted with, and functioned as a competing endogenous RNA (ceRNA) of, miR‑124, consequently upregulating signal transducer and activator of transcription 3 (STAT3). miR‑124 inhibition and STAT3 expression recovery attenuated the effects of the SNHG14 silencing on RB cells. In conclusion, SNHG14 served as a ceRNA to upregulate STAT3 by sponging miR‑124. Therefore, targeting the SNHG14/miR‑124/STAT3 pathway may be an effective therapeutic strategy against RB.
Collapse
Affiliation(s)
- Xiaowen Sun
- Department of Ophthalmology, People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Hui Shen
- Department of Ophthalmology, People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Shubin Liu
- Department of Oncology, Binzhou People's Hospital, Binzhou, Shandong 256610, P.R. China
| | - Jing Gao
- Department of Ophthalmology, Weifang Ophthalmic Hospital, Weifang, Shandong 261041, P.R. China
| | - Shuyan Zhang
- Department of Ophthalmology, People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| |
Collapse
|
29
|
Wang Y, Sun D, Sheng Y, Guo H, Meng F, Song T. XIST promotes cell proliferation and invasion by regulating miR-140-5p and SOX4 in retinoblastoma. World J Surg Oncol 2020; 18:49. [PMID: 32127028 PMCID: PMC7055023 DOI: 10.1186/s12957-020-01825-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 02/19/2020] [Indexed: 12/13/2022] Open
Abstract
Background Retinoblastoma (RB) is the most common intraocular malignancy in children. Long non-coding RNA X-inactive specific transcript (lncRNA XIST) has been reported to be associated with RB, but research on the mechanism of XIST is not well studied. Methods Expressions of XIST, microRNA-140-5p (miR-140-5p), and sex-determining region Y-related high-mobility group box 4 (SOX4) were analyzed by qRT-PCR or Western blot. Relationships of XIST, SOX4, and miR-140-5p were evaluated by dual-luciferase reporter assay and Spearman’s analysis. Cell Counting Kit-8 (CCK-8) and Transwell assay were performed to assess the function of XIST on RB cell proliferation and invasion. Results In RB tissues, XIST and SOX4 expressions were obviously increased, but the miR-140-5p expression was markedly reduced. XIST expression was positively related to SOX4 expression while negatively correlated with miR-140-5p expression, and negative correlation was exhibited between miR-140-5p and SOX4 expression in RB tissues. XIST was confirmed to directly bind to miR-140-5p, and SOX4 was one target of miR-140-5p. XIST knockdown could impede RB cell proliferation and invasion, while miR-140-5p inhibition reversed the effects. In addition, XIST overexpression or miR-140-5p inhibition could abrogate the inhibition of SOX4 silencing on cell proliferation and invasion of RB cells. Conclusions XIST was obviously increased in RB tissues and cells, and XIST inhibition repressed the proliferation and invasion of RB cells by miR-140-5p/SOX4 axis, which may provide new understandings of the XIST molecular mechanism in RB.
Collapse
Affiliation(s)
- Yuhui Wang
- Department of Clinical Laboratory, Affiliated Hospital of Beihua University, Jilin, 132011, China
| | - Dahong Sun
- Department of Pediatrics, The Third People's Hospital of Qingdao, Qingdao, 266041, China
| | - Ying Sheng
- Department of Pediatrics, The Third People's Hospital of Qingdao, Qingdao, 266041, China
| | - Hong Guo
- Health Management Center, The People's Hospital of Zhangqiu Area, Jinan, 250200, China
| | - Fanchun Meng
- Delivery Room, The People's Hospital of Zhangqiu Area, Jinan, 250200, China
| | - Tingting Song
- No. 2 Department of Oncology, Qingdao Central Hospital, Qingdao Tumor Hospital, 127 Siliu South Road, Shibei District, Qingdao, 266042, China.
| |
Collapse
|
30
|
Tracing tumorigenesis in a solid tumor model at single-cell resolution. Nat Commun 2020; 11:991. [PMID: 32080185 PMCID: PMC7033116 DOI: 10.1038/s41467-020-14777-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/29/2020] [Indexed: 12/28/2022] Open
Abstract
Characterizing the complex composition of solid tumors is fundamental for understanding tumor initiation, progression and metastasis. While patient-derived samples provide valuable insight, they are heterogeneous on multiple molecular levels, and often originate from advanced tumor stages. Here, we use single-cell transcriptome and epitope profiling together with pathway and lineage analyses to study tumorigenesis from a developmental perspective in a mouse model of salivary gland squamous cell carcinoma. We provide a comprehensive cell atlas and characterize tumor-specific cells. We find that these cells are connected along a reproducible developmental trajectory: initiated in basal cells exhibiting an epithelial-to-mesenchymal transition signature, tumorigenesis proceeds through Wnt-differential cancer stem cell-like subpopulations before differentiating into luminal-like cells. Our work provides unbiased insights into tumor-specific cellular identities in a whole tissue environment, and emphasizes the power of using defined genetic model systems. Understanding tumour development at a granular level is a challenge in solid tumours. Here, the authors provide a cell atlas across tumour development in a genetic model of salivary gland squamous cell carcinoma using single-cell transcriptome and epitope profiling.
Collapse
|
31
|
Sun J, Xi HY, Shao Q, Liu QH. Biomarkers in retinoblastoma. Int J Ophthalmol 2020; 13:325-341. [PMID: 32090044 DOI: 10.18240/ijo.2020.02.18] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 10/22/2019] [Indexed: 02/06/2023] Open
Abstract
Retinoblastoma (RB) is the most common intraocular malignancy of childhood caused by inactivation of the Rb genes. The prognosis of RB is better with an earlier diagnosis. Many diagnostic approaches and appropriate clinical treatments have been developed to improve clinical outcomes. However, limitations exist when utilizing current methods. Recently, many studies have identified identify new RB biomarkers which can be used in diagnosis, as prognostic indicators and may contribute to understanding the pathogenesis of RB and help determine specific treatment strategies. This review focuses on recent advances in the discovery of RB biomarkers and discusses their clinical utility and challenges from areas such as epigenetics, proteomics and radiogenomics.
Collapse
Affiliation(s)
- Jie Sun
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Hui-Yu Xi
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China.,Department of Ophthalmology, Xuzhou First People's Hospital of Xuzhou Medical University, Xuzhou Eye Research Institute, Xuzhou 221002, Jiangsu Province, China
| | - Qing Shao
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Qing-Huai Liu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
32
|
Matveishina E, Antonov I, Medvedeva YA. Practical Guidance in Genome-Wide RNA:DNA Triple Helix Prediction. Int J Mol Sci 2020; 21:E830. [PMID: 32012884 PMCID: PMC7037363 DOI: 10.3390/ijms21030830] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 12/15/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) play a key role in many cellular processes including chromatin regulation. To modify chromatin, lncRNAs often interact with DNA in a sequence-specific manner forming RNA:DNA triple helices. Computational tools for triple helix search do not always provide genome-wide predictions of sufficient quality. Here, we used four human lncRNAs (MEG3, DACOR1, TERC and HOTAIR) and their experimentally determined binding regions for evaluating triplex parameters that provide the highest prediction accuracy. Additionally, we combined triplex prediction with the lncRNA secondary structure and demonstrated that considering only single-stranded fragments of lncRNA can further improve DNA-RNA triplexes prediction.
Collapse
Affiliation(s)
- Elena Matveishina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Science, 117312 Moscow, Russia
| | - Ivan Antonov
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Science, 117312 Moscow, Russia
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Yulia A Medvedeva
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Science, 117312 Moscow, Russia
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Department of Computational Biology, Vavilov Institute of General Genetics, Russian Academy of Science, 117971 Moscow, Russia
| |
Collapse
|
33
|
Huang X, Fu C, Liu W, Liang Y, Li P, Liu Z, Sheng Q, Liu P. Chemerin-induced angiogenesis and adipogenesis in 3 T3-L1 preadipocytes is mediated by lncRNA Meg3 through regulating Dickkopf-3 by sponging miR-217. Toxicol Appl Pharmacol 2019; 385:114815. [DOI: 10.1016/j.taap.2019.114815] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/29/2019] [Accepted: 11/08/2019] [Indexed: 01/06/2023]
|
34
|
Wang X, Zhang X, Han Y, Wang Q, Ren Y, Wang B, Hu J. Silence of lncRNA ANRIL represses cell growth and promotes apoptosis in retinoblastoma cells through regulating miR-99a and c-Myc. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2265-2273. [PMID: 31184221 DOI: 10.1080/21691401.2019.1623229] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Retinoblastoma is a rare cancer of the immature retina. This study designed to see the function of the lncRNA ANRIL in retinoblastoma Y79 cells. ANRIL, miR-99a and c-Myc expression in Y79 cells was altered by transfection and then trypan blue, transwell assay and flow cytometry were carried out to evaluate the changes of cell phenotype. The connection between ANRIL, miR-99a and c-Myc was measured by luciferase reporter assay and RNA immunoprecipitation analysis. As a result, ANRIL expression was highly expressed in human retinoblastoma tissue as relative to the adjacent noncancerous tissues. ANRIL suppression inhibited Y79 cells viability, migration, invasion, while promoted apoptosis. ANRIL negatively regulated miR-99a by binding to miR-99a. Silence of miR-99a reversed the ANRIL-knockdown effects on Y79 cells. miR-99a overexpression suppressed Y79 cell viability, migration, invasion, and enhanced apoptosis through downregulating c-Myc. Meanwhile, we found that miR-99a inhibited JAK/STAT and PI3K/AKT pathways. To conclude, it seems that ANRIL suppression inhibits cell growth and metastasis in retinoblastoma Y79 cells by regulating miR-99a and c-Myc.
Collapse
Affiliation(s)
- Xiaomin Wang
- a Department of Ophthalmology, The First Affiliated Hospital of Xinxiang Medical University , Weihui , China
| | - Xinxia Zhang
- a Department of Ophthalmology, The First Affiliated Hospital of Xinxiang Medical University , Weihui , China
| | - Yutong Han
- b Department of Ophthalmology, The Third Affiliated Hospital of Xinxiang Medical University , Xinxiang , China
| | - Qiuli Wang
- b Department of Ophthalmology, The Third Affiliated Hospital of Xinxiang Medical University , Xinxiang , China
| | - Yanfan Ren
- a Department of Ophthalmology, The First Affiliated Hospital of Xinxiang Medical University , Weihui , China
| | - Baojun Wang
- a Department of Ophthalmology, The First Affiliated Hospital of Xinxiang Medical University , Weihui , China
| | - Junxi Hu
- a Department of Ophthalmology, The First Affiliated Hospital of Xinxiang Medical University , Weihui , China
| |
Collapse
|
35
|
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
|
36
|
Sherpa C, Rausch JW, Le Grice SF. Structural characterization of maternally expressed gene 3 RNA reveals conserved motifs and potential sites of interaction with polycomb repressive complex 2. Nucleic Acids Res 2019; 46:10432-10447. [PMID: 30102382 PMCID: PMC6212721 DOI: 10.1093/nar/gky722] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/30/2018] [Indexed: 12/15/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as key players in gene regulation. However, our incomplete understanding of the structure of lncRNAs has hindered molecular characterization of their function. Maternally expressed gene 3 (Meg3) lncRNA is a tumor suppressor that is downregulated in various types of cancer. Mechanistic studies have reported a role for Meg3 in epigenetic regulation by interacting with chromatin-modifying complexes such as the polycomb repressive complex 2 (PRC2), guiding them to genomic sites via DNA-RNA triplex formation. Resolving the structure of Meg3 RNA and characterizing its interactions with cellular binding partners will deepen our understanding of tumorigenesis and provide a framework for RNA-based anti-cancer therapies. Herein, we characterize the architectural landscape of Meg3 RNA and its interactions with PRC2 from a functional standpoint.
Collapse
Affiliation(s)
- Chringma Sherpa
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Frederick, MD 21702, USA
| | - Jason W Rausch
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Frederick, MD 21702, USA
| | - Stuart Fj Le Grice
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Frederick, MD 21702, USA
| |
Collapse
|
37
|
Yu F, Pang G, Zhao G. RETRACTED: ANRIL acts as onco-lncRNA by regulation of microRNA-24/c-Myc, MEK/ERK and Wnt/β-catenin pathway in retinoblastoma. Int J Biol Macromol 2019; 128:583-592. [PMID: 30703428 DOI: 10.1016/j.ijbiomac.2019.01.157] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/19/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Authors. Current research findings in the authors' laboratory are in conflict with previous experimental results published in this paper. It was found that the data reported in figures 4 and 5 were not able to be replicated and so therefore the authors have decided that it is best for the scientific record to retract this paper. All the authors agree with the retraction. The authors apologise for any confusion this paper might have caused readers.
Collapse
Affiliation(s)
- Feifei Yu
- Department of Ophthalmology, The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao 266033, Shandong, China
| | - Guolong Pang
- Department of Ophthalmology, Taian City Central Hospital, Taian 271000, Shandong, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China.
| |
Collapse
|
38
|
Zhang L, Dong Y, Wang Y, Gao J, Lv J, Sun J, Li M, Wang M, Zhao Z, Wang J, Xu W. Long non-coding RNAs in ocular diseases: new and potential therapeutic targets. FEBS J 2019; 286:2261-2272. [PMID: 30927500 DOI: 10.1111/febs.14827] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/21/2019] [Accepted: 03/28/2019] [Indexed: 12/21/2022]
Abstract
Long non-coding RNAs (lncRNAs) are non-protein coding transcripts containing more than 200 nucleotides. In the past, lncRNAs were considered as 'transcript noise' or 'pseudogenes' and were thus ignored. However, in recent years, lncRNAs have been proven to regulate gene expression at the epigenetic, transcriptional and translational level, and thereby influence cell proliferation, apoptosis, viability, immune response and oxidative stress. Furthermore, increasing evidence points to their involvement in different diseases, including cancer and heart diseases. Recently, lncRNAs were shown to be differentially expressed in ocular tissues and play a significant role in the pathogenesis of ophthalmological disorders such as glaucoma, corneal diseases, cataract, diabetic retinopathy, proliferative vitreoretinopathy and ocular tumors. In this review, we summarize the classification and mechanisms of known lncRNAs, while detailing their biological functions and roles in ocular diseases. Moreover, we provide a concise review of the clinical relevance of lncRNAs as novel, potential therapeutic targets in the treatment of eye diseases.
Collapse
Affiliation(s)
- Lixia Zhang
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Yanhan Dong
- Institute for Translational Medicine, Qingdao University, China
| | - Yujie Wang
- The Clinical Laboratory of Qingdao Municipal Hospital, China
| | - Jinning Gao
- Institute for Translational Medicine, Qingdao University, China
| | - Jiayi Lv
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Jingguo Sun
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Mengjie Li
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Meng Wang
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Zhihong Zhao
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Jianxun Wang
- Institute for Translational Medicine, Qingdao University, China
| | - Wenhua Xu
- Department of Inspection, The Medical Faculty of Qingdao University, China
| |
Collapse
|
39
|
Wu XZ, Cui HP, Lv HJ, Feng L. Knockdown of lncRNA PVT1 inhibits retinoblastoma progression by sponging miR-488-3p. Biomed Pharmacother 2019; 112:108627. [PMID: 30797143 DOI: 10.1016/j.biopha.2019.108627] [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: 07/01/2018] [Revised: 01/12/2019] [Accepted: 01/24/2019] [Indexed: 12/31/2022] Open
Abstract
Emerging evidence suggests that long non-coding RNAs (lncRNAs) play a regulatory role in the pathogenesis and progression of retinoblastoma (RB). lncRNA plasmacytoma variant translocation 1 (PVT1) is highly expressed in a plenty of tumors, and is believed to serve as an oncogene. However, the expression, roles, and action mechanisms of PVT1 in the carcinogenesis and progression of RB are still largely unknown. In this study, we found that PVT1 was upregulated in RB tissues and cell lines. PVT1 levels correlated with optic nerve invasion, and intraocular international retinoblastoma classify (IIRC) stage. In addition, the results demonstrated that patients with RB who showed higher expression of PVT1 had worse overall survivals. In WERI-Rb1 and Y79 cells, PVT1 silencing significantly inhibited cell proliferation, migration, invasion, and cell cycle progression and induced cell apoptosis in vitro. Moreover, in vivo xenograft assay indicated that PVT1 knockdown suppressed the tumor volume and tumor weight. The analysis of the mechanisms of action revealed that the reduction of PVT1 inhibited the expression of notch2 by upregulating miR-488-3p. In general, our results demonstrated that PVT1 may be a novel biomarker for prognosis and a new target for the treatment of RB.
Collapse
Affiliation(s)
- Xue-Zhi Wu
- Department of Ophthalmology, Zhumadian Central Hospital, Henan Province, 463000, China.
| | - Hong-Pei Cui
- Department of Ophthalmology, Henan Eye Hospital & Henan Eye Institute, Henan Provincial People's Hospital, Henan Province, 450003, China
| | - Hai-Jiang Lv
- Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, 450046, China
| | - Lei Feng
- Department of Ophthalmology, The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450008, China
| |
Collapse
|
40
|
Hu XY, Hou PF, Li TT, Quan HY, Li ML, Lin T, Liu JJ, Bai J, Zheng JN. The roles of Wnt/β-catenin signaling pathway related lncRNAs in cancer. Int J Biol Sci 2018; 14:2003-2011. [PMID: 30585264 PMCID: PMC6299370 DOI: 10.7150/ijbs.27977] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/07/2018] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs), with length of more than 200 nucleotides, are not translated into proteins but involved in multiple diverse diseases, especially tumorigenesis. The dysregulation of lncRNAs greatly contributes to the progression of various tumors through specific signaling pathways, including Wnt/β-catenin signaling pathway, which is associated with malignant features of tumors. The interactions between lncRNAs, which have specific expression characteristics in diverse cancer tissues, and Wnt/β-catenin signaling pathway, exhibit potential as novel biomarkers and therapeutic targets. In this review, we aim to present research findings on the roles of Wnt pathway-related lncRNAs and their effects on Wnt/β-catenin signaling to regulate tumorigenesis in different cancer types. Results may be used as basis to develop or improve strategies for treatment of different carcinomas.
Collapse
Affiliation(s)
- Xiao-Yi Hu
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Ping-Fu Hou
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Teng-Teng Li
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Hao-Yu Quan
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Min-Le Li
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Tian Lin
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Jin-Jin Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Jun-Nian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| |
Collapse
|
41
|
Wang S, Liu J, Yang Y, Hao F, Zhang L. PlncRNA-1 is overexpressed in retinoblastoma and regulates retinoblastoma cell proliferation and motility through modulating CBR3. IUBMB Life 2018; 70:969-975. [PMID: 30096220 DOI: 10.1002/iub.1886] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 01/01/2023]
Abstract
PlncRNA-1 has been suggested to function as an oncogenic role in prostate cancer, colorectal cancer, hepatocellular carcinoma, esophageal squamous cell carcinoma, and gastric cancer. The expression pattern of PlncRNA-1 in retinoblastoma remained unknown. Therefore, the aim of this study was to explore the clinical significance of PlncRNA-1 in retinoblastoma patient and the biological function and molecular mechanism of PlncRNA-1 in regulating retinoblastoma cell proliferation, migration, and invasion. The results showed the level of PlncRNA-1 expression was obviously increased in retinoblastoma tissues and cell lines compared with compared with normal retina tissues and retina cell lines, respectively. Meanwhile, patients with advanced stage retinoblastoma had higher levels of PlncRNA-1 expression than patients with early stage retinoblastoma. There was an inverse correlation between PlncRNA-1 expression and CBR3 expression in retinoblastoma tissues, and PlncRNA-1 negatively regulated mRNA and protein expressions of CBR3. The in vitro experiments showed that down-regulation of PlncRNA-1 expression suppressed retinoblastoma cell proliferation, migration and invasion through up-regulating CBR3. In conclusion, PlncRNA-1 serves as an oncogenic lncRNA in regulating retinoblastoma cell proliferation, migration, and invasion through proliferation, migration, and invasion through up-regulating CBR3. © 2018 IUBMB Life, 70(10):969-975, 2018.
Collapse
Affiliation(s)
- Shuna Wang
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Jianwei Liu
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yang Yang
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Fengqin Hao
- Clinical Medical College, Weifang Medical University, Weifang, Shandong, China
| | - Laixia Zhang
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
42
|
Chen X, Qu J. Long non-coding RNA MEG3 suppresses survival, migration, and invasion of cervical cancer. Onco Targets Ther 2018; 11:4999-5007. [PMID: 30174437 PMCID: PMC6109651 DOI: 10.2147/ott.s167053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Long non-coding RNAs nowadays emerge as important biomarkers or potential therapeutic targets discussed in human cancers. Among them, maternally expressed gene 3 (MEG3) is known to be decreased in a variety of malignancies, and this affects tumor cellular proliferation, migration, and invasion. Materials and methods Quantitative real-time PCR was performed to detect the expression of MEG3 in normal cervical epithelium, cervical intraepithelial neoplasia, and cervical squamous cell carcinoma tissues. Gain-of-function and loss-of-function studies were carried out to determine the effect of MEG3 on cell survival, migration, and invasion, which was evaluated by CCK-8 assay, wound healing assay, and transwell assays. mRNA and protein expression of Rac1 were finally determined by quantitative real-time PCR and immunoblotting, respectively. In addition, rescue experiments were performed by overexpression of Rac1. Results The expression of MEG3 was downregulated in cervical intraepithelial neoplasia and squamous cell carcinoma tissues. Forced expression of MEG3 led to reduced abilities of cell survival. Overexpression of MEG3 also inhibited cell migration and invasion in vitro. Cell proliferation marker and EMT markers were changed consistently with the phenotype. In addition, Rac1 was inhibited by MEG3 overexpression at both transcriptional and translational levels. Also, Rac1 could rescue the phenotype caused by long non-coding RNA MEG3. And, it negatively correlated with MEG3 expression in cervical cancer (CC) tissues and cell lines. Conclusion Our findings revealed that MEG3 could negatively regulate CC cell survival, migration, and invasion. It might serve as an important target for CC treatment.
Collapse
Affiliation(s)
- Xiuhui Chen
- Department of Obstetrics and Gynecology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China,
| | - Junying Qu
- Department of Obstetrics and Gynecology, 1st Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
43
|
Binabaj MM, Bahrami A, Bahreyni A, Shafiee M, Rahmani F, Khazaei M, Soleimanpour S, Ghorbani E, Fiuji H, Ferns GA, Ryzhikov M, Avan A, Hassanian SM. The prognostic value of long noncoding RNA MEG3 expression in the survival of patients with cancer: A meta‐analysis. J Cell Biochem 2018; 119:9583-9590. [DOI: 10.1002/jcb.27276] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/22/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Maryam Moradi Binabaj
- Department of Medical Biochemistry, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Afsane Bahrami
- Student Research Committee, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Cellular and Molecular Research Center Birjand University of Medical Sciences Birjnad Iran
| | - Amirhossein Bahreyni
- Department of Clinical Biochemistry and Immunogenetic Research Center, Faculty of Medicine Mazandaran University of Medical Sciences Sari Mazandaran Iran
| | - Mojtaba Shafiee
- Student Research Committee, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Department of Nutrition, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Farzad Rahmani
- Department of Medical Biochemistry, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Saman Soleimanpour
- Department of Microbiology and Virology, School of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Elnaz Ghorbani
- Department of Microbiology Al‐Zahra University Tehran Iran
| | - Hamid Fiuji
- Department of Biochemistry Payame‐Noor University Mashhad Iran
| | - Gordon A. Ferns
- Division of Medical Education Brighton & Sussex Medical School Brighton Sussex UK
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine Washington University, School of Medicine Saint Louis MO
| | - Amir Avan
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Modern Sciences and Technologies, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
44
|
Li S, Wen D, Che S, Cui Z, Sun Y, Ren H, Hao J. Knockdown of long noncoding RNA 00152 (LINC00152) inhibits human retinoblastoma progression. Onco Targets Ther 2018; 11:3215-3223. [PMID: 29922070 PMCID: PMC5995430 DOI: 10.2147/ott.s160428] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background A growing body of evidence supports the involvement of long noncoding RNA 00152 (LINC00152) in the progression and metastasis of multiple cancers. However, the exact roles of LINC00152 in the progression of human retinoblastoma (RB) remain unknown. We explored the expression and biological function of human RB. Materials and methods The expression level of LINC00152 in RB tissues and cells was analyzed using quantitative real-time PCR. The function of LINC00152 was determined using a series of in vitro assays. In vivo, a nude mouse model was established to analyze the function of LINC00152. Gene and protein expressions were detected using quantitative real-time PCR and Western blot assays, respectively. Results The expression of LINC00152 mRNA was upregulated in RB tissues and cell lines. Knockdown of LINC00152 significantly inhibited cell proliferation, colony formation, migration, and invasion and promoted cell apoptosis and caspase-3 and caspase-8 activities in vitro, as well as suppressing tumorigenesis in vivo. We identified several genes related to proliferation, apoptosis, and invasion including Ki-67, Bcl-2, and MMP-9 that were transcriptionally inactivated by LINC00152. Conclusion Taken together, these data implicate LINC00152 as a therapeutic target in RB.
Collapse
Affiliation(s)
- Songhe Li
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Dacheng Wen
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Songtian Che
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhihua Cui
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Yabin Sun
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Hua Ren
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Jilong Hao
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
45
|
Jansen RW, de Jong MC, Kooi IE, Sirin S, Göricke S, Brisse HJ, Maeder P, Galluzzi P, van der Valk P, Cloos J, Eekhout I, Castelijns JA, Moll AC, Dorsman JC, de Graaf P. MR Imaging Features of Retinoblastoma: Association with Gene Expression Profiles. Radiology 2018; 288:506-515. [PMID: 29714679 DOI: 10.1148/radiol.2018172000] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Purpose To identify associations between magnetic resonance (MR) imaging features and gene expression in retinoblastoma. Materials and Methods A retinoblastoma MR imaging atlas was validated by using anonymized MR images from referral centers in Essen, Germany, and Paris, France. Images were from 39 patients with retinoblastoma (16 male and 18 female patients [the sex in five patients was unknown]; age range, 5-90 months; inclusion criterion: pretreatment MR imaging). This atlas was used to compare MR imaging features with genome-wide messenger RNA (mRNA) expression data from 60 consecutive patients obtained from 1995 to 2012 (35 male patients [58%]; age range, 2-69 months; inclusion criteria: pretreatment MR imaging, genome-wide mRNA expression data available). Imaging pathway associations were analyzed by means of gene enrichment. In addition, imaging features were compared with a predefined gene expression signature of photoreceptorness. Statistical analysis was performed with generalized linear modeling of radiology traits on normalized log2-transformed expression values. P values were corrected for multiple hypothesis testing. Results Radiogenomic analysis revealed 1336 differentially expressed genes for qualitative imaging features (threshold P = .05 after multiple hypothesis correction). Loss of photoreceptorness gene expression correlated with advanced stage imaging features, including multiple lesions (P = .03) and greater eye size (P < .001). The number of lesions on MR images was associated with expression of MYCN (P = .04). A newly defined radiophenotype of diffuse-growing, plaque-shaped, multifocal tumors displayed overexpression of SERTAD3 (P = .003, P = .049, and P = .06, respectively), a protein that stimulates cell growth by activating the E2F network. Conclusion Radiogenomic biomarkers can potentially help predict molecular features, such as photoreceptorness loss, that indicate tumor progression. Results imply a possible role for radiogenomics in future staging and treatment decision making in retinoblastoma.
Collapse
Affiliation(s)
- Robin W Jansen
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Marcus C de Jong
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Irsan E Kooi
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Selma Sirin
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Sophia Göricke
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Hervé J Brisse
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Philippe Maeder
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Paolo Galluzzi
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Paul van der Valk
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Jacqueline Cloos
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Iris Eekhout
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Jonas A Castelijns
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Annette C Moll
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Josephine C Dorsman
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Pim de Graaf
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| |
Collapse
|
46
|
Zheng Q, Lin Z, Xu J, Lu Y, Meng Q, Wang C, Yang Y, Xin X, Li X, Pu H, Gui X, Li T, Xiong W, Lu D. Long noncoding RNA MEG3 suppresses liver cancer cells growth through inhibiting β-catenin by activating PKM2 and inactivating PTEN. Cell Death Dis 2018; 9:253. [PMID: 29449541 PMCID: PMC5833746 DOI: 10.1038/s41419-018-0305-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Maternally expressed gene 3 (MEG3) encodes an lncRNA which is suggested to function as a tumor suppressor and has been showed to involve in a variety of cancers. Herein, our findings demonstrate that MEG3 inhibits the malignant progression of liver cancer cells in vitro and in vivo. Mechanistically, MEG3 promotes the expression and maturition of miR122 which targets PKM2. Therefore, MEG3 decreases the expression and nuclear location of PKM2 dependent on miR122. Furthermore, MEG3 also inhibits CyclinD1 and C-Myc via PKM2 in liver cancer cells. On the other hand, MEG3 promotes β-catenin degradation through ubiquitin-proteasome system dependent on PTEN. Strikingly, MEG3 inhibits β-catenin activity through PKM2 reduction and PTEN increase. Significantly, we also found that excessive β-catenin abrogated the effect of MEG3 in liver cancer. In conclusion, our study for the first time demonstrates that MEG3 acts as a tumor suppressor by negatively regulating the activity of the PKM2 and β-catenin signaling pathway in hepatocarcinogenesis and could provide potential therapeutic targets for the treatment of liver cancer.
Collapse
Affiliation(s)
- Qidi Zheng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Zhuojia Lin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Jie Xu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Yanan Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Qiuyu Meng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Chen Wang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Yuxin Yang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xiaoru Xin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xiaonan Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Hu Pu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xin Gui
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Tianming Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Wujun Xiong
- Department of Hepatology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Dongdong Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China.
| |
Collapse
|
47
|
Gao Y, Huang P, Zhang J. Hypermethylation of MEG3 promoter correlates with inactivation of MEG3 and poor prognosis in patients with retinoblastoma. J Transl Med 2017; 15:268. [PMID: 29287592 PMCID: PMC5747938 DOI: 10.1186/s12967-017-1372-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 12/19/2017] [Indexed: 01/01/2023] Open
Abstract
Background In our previous study, we revealed that MEG3 was a tumor suppressor gene in retinoblastoma and inhibited proliferation of retinoblastoma cells by regulating the activity of the Wnt/β-catenin pathway. Here, we further explored the mechanism of MEG3 inactivation in retinoblastoma. Methods MSP and qRT-PCR were performed to detect the methylation status of MEG3 promoter and levels of MEG3 expression, respective. To further explore relationship between MEG3 expression and epigenetic modifications, 5-Aza-CdR was used to interfere with DNA methylation. In addition, we evaluated proliferation, apoptosis and the expression of β-catenin via CCK-8, flow cytometric analysis and western blot analysis, respective. Results Hypermethylation of MEG3 promoter was observed more frequently in retinoblastoma tissues and was highly associated with low MEG3 expression and poor survival of retinoblastoma patients. We also provided evidence demonstrating that hypermethylation of MEG3 promoter depressed MEG3 expression, promoted proliferation, inhibited apoptosis and increased β-catenin expression of retinoblastoma cells in vitro. Conclusions Our present study indicates that promoter silencing by hypermethylation may account for the loss of MEG3 expression and predict poor prognosis.
Collapse
Affiliation(s)
- Yali Gao
- Department of Ophthalmology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, 518020, People's Republic of China
| | - Peng Huang
- Department of Ophthalmology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, 518020, People's Republic of China
| | - Jun Zhang
- Department of Obstetrics and Gynecology, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, 518020, People's Republic of China.
| |
Collapse
|
48
|
Niu ZS, Niu XJ, Wang WH. Long non-coding RNAs in hepatocellular carcinoma: Potential roles and clinical implications. World J Gastroenterol 2017; 23:5860-5874. [PMID: 28932078 PMCID: PMC5583571 DOI: 10.3748/wjg.v23.i32.5860] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 05/10/2017] [Accepted: 07/22/2017] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are a subgroup of non-coding RNA transcripts greater than 200 nucleotides in length with little or no protein-coding potential. Emerging evidence indicates that lncRNAs may play important regulatory roles in the pathogenesis and progression of human cancers, including hepatocellular carcinoma (HCC). Certain lncRNAs may be used as diagnostic or prognostic markers for HCC, a serious malignancy with increasing morbidity and high mortality rates worldwide. Therefore, elucidating the functional roles of lncRNAs in tumors can contribute to a better understanding of the molecular mechanisms of HCC and may help in developing novel therapeutic targets. In this review, we summarize the recent progress regarding the functional roles of lncRNAs in HCC and explore their clinical implications as diagnostic or prognostic biomarkers and molecular therapeutic targets for HCC.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinogenesis/genetics
- Carcinoma, Hepatocellular/diagnosis
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/mortality
- Disease Progression
- Early Detection of Cancer/methods
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/diagnosis
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/mortality
- Molecular Targeted Therapy/methods
- Prognosis
- RNA, Long Noncoding/analysis
- RNA, Long Noncoding/antagonists & inhibitors
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
Collapse
Affiliation(s)
- Zhao-Shan Niu
- Laboratory of Micromorphology, School of Basic Medicine, Medical Department of Qingdao University, Qingdao 266071, Shandong Province, China
| | - Xiao-Jun Niu
- Oncology Specialty, Medical Department of Qingdao University, Qingdao 266071, Shandong Province, China
| | - Wen-Hong Wang
- Department of Pathology, School of Basic Medicine, Medical Department of Qingdao University, Qingdao 266071, Shandong Province, China
| |
Collapse
|
49
|
Wang G, Bu X, Zhang Y, Zhao X, Kong Y, Ma L, Niu S, Wu B, Meng C. LncRNA-UCA1 enhances MMP-13 expression by inhibiting miR-204-5p in human chondrocytes. Oncotarget 2017; 8:91281-91290. [PMID: 29207643 PMCID: PMC5710923 DOI: 10.18632/oncotarget.20108] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/25/2017] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative disease characterized by degeneration of articular cartilage. Increasing studies showed that long noncoding RNAs (lncRNAs) play important roles in the cartilage damage. However, little is known about the role of UCA1 in the osteoarthritis. The expression level of UCA1 was upregulated in the OA cartilage. Overexpression of UCA1 suppressed the miR-204-5p expression in the chondrocytes. The expression of miR-204-5p was downregulated in the OA cartilage. Moreover, the expression of miR-204-5p was negatively correlated with the UCA1 expression in the OA cartilage. Elevated expression of UCA1 promoted the chondrocytes cell proliferation and overexpression of miR-204-5p suppressed chondrocytes cell proliferation. In addition, overexpression of UCA1 decreased the expression of the type II collagen and type IV collagen expression in the chondrocytes. Elevated expression of miR-204-5p promoted the type II collagen and type IV collagen expression in the chondrocytes. We idetified MMP-13 was a direct target gene of miR-204-5p in the chondrocytes. Overexpression of UCA1 enhanced the MMP-13 expression in the chondrocytes. Elevated expression of UCA1 regulated the chondrocytes cell proliferation and collagen expression through inhibiting the miR-204-5p expression.These results suggested that UCA1 played as an important regulator of survival and matrix synthesis of chondrocytes partly through suppressing the miR-204-5p expression.
Collapse
Affiliation(s)
- Guodong Wang
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xianmin Bu
- Department of Pathology, Shandong Jining No.1 People's Hospital, Jining, Shandong, China
| | - Yuanmin Zhang
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xiaowei Zhao
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Ying Kong
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Longfei Ma
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Shuaishuai Niu
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Bin Wu
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Chunyang Meng
- Department of Orthopaedics, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| |
Collapse
|
50
|
He Y, Luo Y, Liang B, Ye L, Lu G, He W. Potential applications of MEG3 in cancer diagnosis and prognosis. Oncotarget 2017; 8:73282-73295. [PMID: 29069869 PMCID: PMC5641212 DOI: 10.18632/oncotarget.19931] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022] Open
Abstract
LncRNAs are emerging as integral functional and regulatory components of normal biological activities and are now considered as critically involved in the development of different diseases including cancer. In this review, we summarized recent findings on maternally expressed gene 3 (MEG3), a noncoding lncRNA, locates in the imprinted DLK1–MEG3 locus on human chromosome 14q32.3 region. MEG3 is expressed in normal tissues but is either lost or decreased in many human tumors and tumor derived cell lines. Studies have demonstrated that MEG3 is associated with cancer initiation, progression, metastasis and chemo-resistance. MEG3 may affect the activities of TP53, MDM2, GDF15, RB1 and some other key cell cycle regulators. In addition, the level of MEG3 showed good correlation with cancer clinicopathological grade. In summary, MEGs is an RNA-based tumor suppressor and is involved in the etiology, progression, and chemosensitivity of cancers. The alteration of MEG3 levels in various cancers suggested the possibility of using MEG3 level for cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Yuqing He
- Institute of Medical Systems Biology, Guangdong Medical University, Dongguan 523808, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Guangdong Medical University, Dongguan 523808, China
| | - Yanhong Luo
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Biyu Liang
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Lei Ye
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Guangxing Lu
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Weiming He
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|