1
|
Abdelhamid AM, Zeinelabdeen Y, Manie T, Khallaf E, Assal RA, Youness RA. miR-17-5p/STAT3/H19: A novel regulatory axis tuning ULBP2 expression in young breast cancer patients. Pathol Res Pract 2024; 263:155638. [PMID: 39388743 DOI: 10.1016/j.prp.2024.155638] [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: 08/04/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND AND AIM UL-16 binding protein 2 (ULBP2) is a highly altered ligand for the activating receptor, NKG2D in breast cancer (BC). However, the mechanism behind its de-regulation in BC patients remains to be explored. The sophisticated crosstalk between miR-17-5p, the lncRNA H19, and STAT3 as a possible upstream regulatory loop for ULBP2 in young BC patients and cell lines remains as an unexplored area. Therefore, this study aimed at unravelling the ncRNA circuit regulating ULBP2 in young BC patients and cell lines. PATIENTS AND METHODS A total of 30 BC patients were recruited for this study. The expression levels of miR-17-5p, lncRNA H19, and STAT3 were examined in 30 BC tissues compared to their normal counterparts. In addition, the expression signatures of those transcripts were compared in young (<40 years) and old BC (≥40 years) patients. miR-17-5p oligonucleotides, STAT3 and H19 siRNAs were transfected in MDA-MB-231 cells using HiPerfect® Transfection Reagent. miR-17-5p and the transcripts of the target genes quantified using RT-qPCR. Their relative expression was calculated using the 2-ΔΔCT method. RESULTS Through acting as a ceRNA circuit that antagonizes the function of miR-17-5p, H19 prevented the miR-17-5p-induced downregulation of STAT3; this mechanism further contributes to the pathogenesis of BC. Ectopic expression of miR-17-5p in MDA-MB-231 cells displayed its prominent role as an indirect potential activator of NK cells by significantly repressing the expression levels of the oncogenic mediator STAT3 and the oncogenic lncRNA H19 and inducing ULBP2 expression level by 3 folds in TNBC cell lines compared to mock cells. Furthermore, knocking down of STAT3 repressed the lncRNA H19 and increased ULBP2 expression levels, whereas siRNAs against H19 increased the expression levels of ULBP2. CONCLUSION This study highlighted the crosstalk between the novel regulatory network composed of miR-17-5p, H19 and STAT3, and their impact on ULBP2 in BC. Moreover, this study underscored the potential role of miR-17-5p in counteracting the immune evasion tactics, particularly the shedding of ULBP2 in young BC patients, through the modulation of the STAT3/H19/ULBP2 regulatory axis. Thus, targeting this novel regulatory network could potentially enhance our understanding and advance the future application of the innate system-mediated immunotherapy in BC.
Collapse
Affiliation(s)
- A M Abdelhamid
- Biotechnology School, Nile University, Giza 12588, Egypt
| | - Y Zeinelabdeen
- Faculty of Medical Sciences/UMCG, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, the Netherlands
| | - T Manie
- Department of Breast Surgery, National Cancer Institute, Cairo University, Cairo, Egypt
| | - E Khallaf
- Department of General Surgery, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - R A Assal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - R A Youness
- Molecular Genetics Research Team (MGRT), Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University, New Administrative Capital 11835, Egypt.
| |
Collapse
|
2
|
Li JX, Qu YD, Xia CL, Zhang W, Wang SS, Ou SJ, Yang Y, Qi Y, Xu CP. Analysis of PANoptosis-related ceRNA network reveals lncRNA MIR17HG involved in osteogenic differentiation inhibition impaired by tumor necrosis factor-α. Mol Biol Rep 2024; 51:909. [PMID: 39145884 PMCID: PMC11327206 DOI: 10.1007/s11033-024-09810-0] [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: 04/28/2024] [Accepted: 07/19/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Inflammatory cytokines such as Interleukin 1β(IL1β), IL6,Tumor Necrosis Factor-α (TNF-α) can inhibit osteoblast differentiation and induce osteoblast apoptosis. PANoptosis, a newly identified type of programmed cell death (PCD), may be influenced by long noncoding RNA (lncRNAs) which play important roles in regulating inflammation. However, the potential role of lncRNAs in inflammation and PANoptosis during osteogenic differentiation remains unclear. This study aimed to investigate the regulatory functions of lncRNAs in inflammation and apoptosis during osteogenic differentiation. METHODS AND RESULTS High-throughput sequencing was used to identify differentially expressed genes involved in osteoblast differentiation under inflammatory conditions. Two lncRNAs associated with inflammation and PANoptosis during osteogenic differentiation were identified from sequencing data and Gene Expression Omnibus (GEO) databases. Their functionalities were analyzed using diverse bioinformatics methodologies, resulting in the construction of the lncRNA-miRNA-mRNA network. Among these, lncRNA (MIR17HG) showed a high correlation with PANoptosis. Bibliometric methods were employed to collect literature data on PANoptosis, and its components were inferred. PCR and Western Blotting experiments confirmed that lncRNA MIR17HG is related to PANoptosis in osteoblasts during inflammation. CONCLUSIONS Our data suggest that TNF-α-induced inhibition of osteogenic differentiation and PANoptosis in MC3T3-E1 osteoblasts is associated with MIR17HG. These findings highlight the critical role of MIR17HG in the interplay between inflammation, PANoptosis, and osteogenic differentiation, suggesting potential therapeutic targets for conditions involving impaired bone formation and inflammatory responses.
Collapse
Affiliation(s)
- Jia-Xuan Li
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu-Dun Qu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Chang-Liang Xia
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Wei Zhang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Song-Song Wang
- School of Medicine, XiaMen University, Xiamen, Fujian, China
| | - Shuan-Ji Ou
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yang Yang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yong Qi
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| | - Chang-Peng Xu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| |
Collapse
|
3
|
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
|
4
|
Zhang R, Zeng Y, Deng JL. Long non-coding RNA H19: a potential biomarker and therapeutic target in human malignant tumors. Clin Exp Med 2023; 23:1425-1440. [PMID: 36484927 DOI: 10.1007/s10238-022-00947-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/08/2022] [Indexed: 12/13/2022]
Abstract
Long non-coding RNAs play important roles in cellular functions and disease development. H19, as a long non-coding RNA, is pervasively over-expressed in almost all kinds of human malignant tumors. Although many studies have reported that H19 is closely associated with tumor cell proliferation, apoptosis, invasion, metastasis, and chemoresistance, the role and mechanism of H19 in gene regulation and tumor development are largely unclear. In this review, we summarized the recent progress in the study of the major functions and mechanisms of H19 lncRNA in cancer development and progression. H19 possesses both oncogenic and tumor-suppressing activities, presumably through regulating target gene transcription, mRNA stability and splicing, and competitive inhibition of endogenous RNA degradation. Studies indicate that H19 may involve in cell proliferation and apoptosis, tumor initiation, migration, invasion, metastasis and chemoresistance and may serve as a potential biomarker for early diagnosis, prognosis, and novel molecular target for cancer therapy.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Pharmacy, Anhui No.2 Provincial People's Hospital, Hefei, 230041, People's Republic of China
| | - Ying Zeng
- Department of Pharmacy, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410008, People's Republic of China
| | - Jun-Li Deng
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, People's Republic of China.
| |
Collapse
|
5
|
Tang W, Zhang L, Li J, Guan Y. KCNQ1OT1 promotes retinoblastoma progression by targeting miR-339-3p that suppresses KIF23. Int Ophthalmol 2023:10.1007/s10792-023-02641-1. [PMID: 37198502 DOI: 10.1007/s10792-023-02641-1] [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: 08/20/2021] [Accepted: 01/19/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are involved in tumor formation and development. KCNQ1OT1 regulates the malignant proliferation of retinoblastoma (RB), but the specific mechanism remains to be further investigated. METHODS The KCNQ1OT1, miR-339-3p and KIF23 expression levels in RB were detected by qRT-PCR and western blotting. The cell viability, proliferation, migration ability and caspase-3 activity of RB cells were evaluated by CCK-8, BrdU, transwell and caspase-3 activity analysis. Western blot was used to detect the Bax and Bcl-2 protein expression in RB cells. The binding relationship between KCNQ1OT1, miR-339-3p and KIF23 was detected by luciferase, RIP and RNA pull-down assay. RESULTS KCNQ1OT1 and KIF23 were up-regulated frequently in RB, and miR-339-3p was down-regulated. Functional studies showed that downregulation of KCNQ1OT1 or KIF23 inhibited the survival and migration of RB cells, and facilitated apoptosis. Interference with miR-339-3p showed the opposite effect. Mechanisms suggested that KCNQ1OT1 exited its oncogenic activity by positively regulating the expression of KIF23 and sponging miR-339-3p. CONCLUSION KCNQ1OT1/miR-339-3p/KIF23 may be a new biomarker for the diagnosis and treatment of RB.
Collapse
Affiliation(s)
- Wenting Tang
- Department of Ophthalmology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Li Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Jing Li
- Department of Ophthalmology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Yu Guan
- Department of Ophthalmology, The 2nd Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, No. 4, North 4th Erhuan Street, Chengdu, 610051, Sichuan, China.
| |
Collapse
|
6
|
Xu C, Jiang ZB, Shao L, Zhao ZM, Fan XX, Sui X, Yu LL, Wang XR, Zhang RN, Wang WJ, Xie YJ, Zhang YZ, Nie XW, Xie C, Huang JM, Wang J, Wang J, Leung ELH, Wu QB. β-Elemene enhances erlotinib sensitivity through induction of ferroptosis by upregulating lncRNA H19 in EGFR-mutant non-small cell lung cancer. Pharmacol Res 2023; 191:106739. [PMID: 36948327 DOI: 10.1016/j.phrs.2023.106739] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/04/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
Nearly half of all Asian non-small cell lung cancer (NSCLC) patients harbour epidermal growth factor receptor (EGFR) mutations, and first-generation EGFR tyrosine kinase inhibitors (TKIs) are one of the first-line treatments that have improved the outcomes of these patients. Unfortunately, 20% of these patients can not benefit from the treatment. The basis of this primary resistance is poorly understood. Therefore, overcoming EGFR-TKI primary resistance and maintaining the efficacy of TKIs has become a key issue. β-Elemene, a sesquiterpene compound extracted from Curcuma aromatica Salisb. (wenyujing), has shown potent antitumor effects. In this research, we found that β-elemene combined with erlotinib enhanced the cytotoxicity of erlotinib to primary EGFR-TKI-resistant NSCLC cells with EGFR mutations and that ferroptosis was involved in the antitumor effect of the combination treatment. We found that lncRNA H19 was significantly downregulated in primary EGFR-TKI-resistant NSCLC cell lines and was upregulated by the combination treatment. Overexpression or knockdown of H19 conferred sensitivity or resistance to erlotinib, respectively, in both in vitro and in vivo studies. The high level of H19 enhanced the cytotoxicity of erlotinib by inducing ferroptosis. In conclusion, our data showed that β-elemene combined with erlotinib could enhance sensitivity to EGFR-TKIs through induction of ferroptosis via H19 in primary EGFR-TKI-resistant lung cancer, providing a promising strategy to overcome EGFR-TKI resistance in NSCLC patients.
Collapse
Affiliation(s)
- Cong Xu
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Ze-Bo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Le Shao
- The First Hospital, Hunan University of Chinese Medicine, Changsha 410007, Hunan Province, China
| | - Zi-Ming Zhao
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Xinbing Sui
- College of Pharmacy, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China; Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
| | - Li-Li Yu
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Xuan-Run Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Ruo-Nan Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Wen-Jun Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Ya-Jia Xie
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Yi-Zhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Xiao-Wen Nie
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Chun Xie
- Cancer Center, Faculty of Health Science, University of Macau, Macau (SAR) 999078, China; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR) 999078, China
| | - Ju-Min Huang
- Cancer Center, Faculty of Health Science, University of Macau, Macau (SAR) 999078, China; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR) 999078, China
| | - Jing Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China
| | - Jue Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science, University of Macau, Macau (SAR) 999078, China; MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR) 999078, China.
| | - Qi-Biao Wu
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, China; Zhuhai MUST Science and Technology Research Institute, Zhuhai 51900, Guangdong, China; Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
7
|
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
|
8
|
Marangon D, Castro e Silva JH, Lecca D. Neuronal and Glial Communication via Non-Coding RNAs: Messages in Extracellular Vesicles. Int J Mol Sci 2022; 24:ijms24010470. [PMID: 36613914 PMCID: PMC9820657 DOI: 10.3390/ijms24010470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Extracellular vesicles (EVs) have been increasingly recognized as essential players in cell communication in many organs and systems, including the central nervous system (CNS). A proper interaction between neural cells is fundamental in the regulation of neurophysiological processes and its alteration could induce several pathological phenomena, such as neurodegeneration, neuroinflammation, and demyelination. EVs contain and transfer complex molecular cargoes typical of their cells of origin, such as proteins, lipids, carbohydrates, and metabolites to recipient cells. EVs are also enriched in non-coding RNAs (e.g., microRNAs, lncRNAs, and circRNA), which were formerly considered as cell-intrinsic regulators of CNS functions and pathologies, thus representing a new layer of regulation in the cell-to-cell communication. In this review, we summarize the most recent and advanced studies on the role of EV-derived ncRNAs in the CNS. First, we report the potential of neural stem cell-derived ncRNAs as new therapeutic tools for neurorepair. Then, we discuss the role of neuronal ncRNAs in regulating glia activation, and how alteration in glial ncRNAs influences neuronal survival and synaptic functions. We conclude that EV-derived ncRNAs can act as intercellular signals in the CNS to either propagate neuroinflammatory waves or promote reparative functions.
Collapse
|
9
|
Ahangar Davoodi N, Najafi S, Naderi Ghale-Noie Z, Piranviseh A, Mollazadeh S, Ahmadi Asouri S, Asemi Z, Morshedi M, Tamehri Zadeh SS, Hamblin MR, Sheida A, Mirzaei H. Role of non-coding RNAs and exosomal non-coding RNAs in retinoblastoma progression. Front Cell Dev Biol 2022; 10:1065837. [PMID: 36619866 PMCID: PMC9816416 DOI: 10.3389/fcell.2022.1065837] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Retinoblastoma (RB) is a rare aggressive intraocular malignancy of childhood that has the potential to affect vision, and can even be fatal in some children. While the tumor can be controlled efficiently at early stages, metastatic tumors lead to high mortality. Non-coding RNAs (ncRNAs) are implicated in a number of physiological cellular process, including differentiation, proliferation, migration, and invasion, The deregulation of ncRNAs is correlated with several diseases, particularly cancer. ncRNAs are categorized into two main groups based on their length, i.e. short and long ncRNAs. Moreover, ncRNA deregulation has been demonstrated to play a role in the pathogenesis and development of RB. Several ncRNAs, such as miR-491-3p, miR-613,and SUSD2 have been found to act as tumor suppressor genes in RB, but other ncRNAs, such as circ-E2F3, NEAT1, and TUG1 act as tumor promoter genes. Understanding the regulatory mechanisms of ncRNAs can provide new opportunities for RB therapy. In the present review, we discuss the functional roles of the most important ncRNAs in RB, their interaction with the genes responsible for RB initiation and progression, and possible future clinical applications as diagnostic and prognostic tools or as therapeutic targets.
Collapse
Affiliation(s)
- Nasrin Ahangar Davoodi
- Eye Research Center, Rassoul Akram Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zari Naderi Ghale-Noie
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ashkan Piranviseh
- Brain and Spinal Cord Injury Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammadamin Morshedi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran,School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran,School of Medicine, Kashan University of Medical Sciences, Kashan, Iran,*Correspondence: Amirhossein Sheida, ; Hamed Mirzaei, ,
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran,*Correspondence: Amirhossein Sheida, ; Hamed Mirzaei, ,
| |
Collapse
|
10
|
Mandal M, Banerjee I, Mandal M. Nanoparticle-mediated gene therapy as a novel strategy for the treatment of retinoblastoma. Colloids Surf B Biointerfaces 2022; 220:112899. [DOI: 10.1016/j.colsurfb.2022.112899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/05/2022]
|
11
|
Wang J, Cao B, Sun R, Chen Y, Feng J. Exosome-transported lncRNA H19 induces blood-brain barrier disruption in cerebral ischemic stroke via the H19/microRNA-18a/VEGF axis. Neuroscience 2022; 500:41-51. [PMID: 35931357 DOI: 10.1016/j.neuroscience.2022.07.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 10/16/2022]
Abstract
Long non-coding RNA H19 (lncRNA H19) is transcribed from the H19 gene. We previously reported the role of lncRNA H19 in the pathogenesis of cerebral ischemic stroke. The present study aimed to elucidate the relationship between lncRNA H19 and blood-brain barrier breakdown induced by cerebral ischemic stroke. We observed that plasma levels of lncRNA H19 were positively associated with the extent of blood-brain barrier damage. In cellular co-culture models, neurons expressed and transported lncRNA H19 to astrocytes via exosomes and contributed to an increase in endothelium permeability induced by oxygen-glucose deprivation. Inhibition of neuronal exosomal lncRNA H19 regulated astrocytic microRNA (miR)-18a and vascular endothelial growth factor (VEGF) expression. Further, lncRNA H19 induced a decrease in tight junction proteins expression via the lncRNA H19/miR-18a/VEGF axis. This study highlights the transportation of lncRNA H19 by exosomes and the relationship between lncRNA H19 and blood-brain barrier breakdown.
Collapse
Affiliation(s)
- Jue Wang
- Department of Neurology, Shengjing Hospital of China Medical University
| | - Bin Cao
- Department of Neurology, Shengjing Hospital of China Medical University
| | - Ruize Sun
- Department of Neurology, Shengjing Hospital of China Medical University
| | - Yuhua Chen
- Department of Developmental Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University.
| |
Collapse
|
12
|
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: 2.5] [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
|
13
|
Yan J, Deng Y, Cai Y, Cong W. LncRNA MIR17HG
promotes the proliferation, migration, and invasion of retinoblastoma cells by up‐regulating
HIF
‐1α expression via sponging
miR
‐155‐5p. Kaohsiung J Med Sci 2022; 38:554-564. [PMID: 35253975 DOI: 10.1002/kjm2.12523] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 12/28/2022] Open
Affiliation(s)
- Jian Yan
- Department of Ophthalmology Longgang District Central Hospital Shenzhen China
| | - Yi‐Xuan Deng
- Department of Ophthalmology Longgang District Central Hospital Shenzhen China
| | - Yu‐Lian Cai
- Department of Ophthalmology Longgang District Central Hospital Shenzhen China
| | - Wen‐Dong Cong
- Department of Neurology Longgang District Central Hospital Shenzhen China
| |
Collapse
|
14
|
Ren H, Guo X, Li F, Xia Q, Chen Z, Xing Y. Four Autophagy-Related Long Noncoding RNAs Provide Coexpression and ceRNA Mechanisms in Retinoblastoma through Bioinformatics and Experimental Evidence. ACS OMEGA 2021; 6:33976-33984. [PMID: 34926945 PMCID: PMC8674985 DOI: 10.1021/acsomega.1c05259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/15/2021] [Indexed: 06/14/2023]
Abstract
Retinoblastoma (RB) is the most common type of intraocular malignant tumor that lowers the quality of life among children worldwide. Long noncoding RNAs (lncRNAs) are reported to play a dual role in tumorigenesis and development of RB. Autophagy is also reported to be involved in RB occurrence. Although several studies of autophagy-related lncRNAs in RB have been explored before, there are still unknown potential mechanisms in RB. In the present study, we mined dataset GSE110811 from the Gene Expression Omnibus database and downloaded autophagy-related genes from the Human Autophagy Database for further bioinformatic analysis. By implementing the differential expression analysis and Pearson correlation analysis on the lncRNA expression matrix and autophagy-related genes expression matrix, we identified four autophagy-related lncRNAs (namely, N4BP2L2-IT2, SH3BP5-AS1, CDKN2B-AS1, and LINC-PINT) associated with RB. We then performed differential expression analysis on microRNA (miRNA) from dataset GSE39105 for further analyses of lncRNA-miRNA-mRNA regulatory mechanisms. With the miRNA-lncRNA module on the StarBase 3.0 website, we predicted the differentially expressed miRNAs that could target the autophagy-related lncRNAs and constructed a potential lncRNA-miRNA-mRNA regulatory network. Furthermore, the functional annotations of these target genes in regulatory networks were presented using the Cytoscape and the Metascape annotation tool. Finally, the expression pattern of the four autophagy-related lncRNAs was evaluated via qRT-PCR. In conclusion, our findings suggest that the four autophagy-related lncRNAs could be critical molecules associated with the development of RB and affect the occurrence and development of RB through the lncRNA-miRNA-mRNA regulatory network. Genes (GRP13B, IFT88, EPHA3, GABARAPL1, and EIF4EBP1) may serve as potential novel therapeutic targets and biomarkers in RB.
Collapse
|
15
|
Li H, Fan D, Wang W, Zhang X, Song L, Huang Y. MiR-142-5p serves as a tumor suppressor in retinoblastoma cells by regulating MYCN. Biochem Biophys Res Commun 2021; 574:20-26. [PMID: 34425282 DOI: 10.1016/j.bbrc.2021.07.099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 10/20/2022]
Abstract
Retinoblastoma is an intraocular malignant tumor and generally occurred in childhood. Here, we intended to appraise the functional influence of microRNA-142-5p (miR-142-5p) in retinoblastoma. MiR-142-5p was declined, and MYCN was upregulated in retinoblastoma tissues and cells. Moreover, miR-142-5p restricted cell proliferation, migration, invasion, and enhanced cell apoptosis in retinoblastoma cells. MYCN was adversely controlled by miR-142-5p. Besides, the inhibition of miR-142-5p-mediated effects on retinoblastoma progression were blocked by MYCN overexpression in retinoblastoma cells. This research illustrated that miR-142-5p restricted retinoblastoma progression via interacting with MYCN.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Ophthlmology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China.
| | - Dongsheng Fan
- Department of Ophthlmology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Wanli Wang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Xinli Zhang
- Department of Ophthlmology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Lili Song
- Department of Ophthlmology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Yanxia Huang
- Department of Ophthlmology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| |
Collapse
|
16
|
Xia M, Duan LJ, Lu BN, Pang YZ, Pang ZR. LncRNA AFAP1-AS1/miR-27b-3p/VEGF-C axis modulates stemness characteristics in cervical cancer cells. Chin Med J (Engl) 2021; 134:2091-2101. [PMID: 34334630 PMCID: PMC8440026 DOI: 10.1097/cm9.0000000000001665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: Long non-coding RNA (lncRNA) actin filament-associated protein 1 antisense RNA 1 (AFAP1-AS1) functions as a competing endogenous RNA to regulate target genes expression by sponging microRNAs (miRs) to play cancer-promoting roles in cancer stem cells. However, the regulatory mechanism of AFAP1-AS1 in cervical cancer (CC) stem cells is unknown. The present study aimed to provide a new therapeutic target for the clinical treatment of CC. Methods: Hyaluronic acid receptor cluster of differentiation 44 variant exon 6 (CD44v6)(+) CC cells were isolated by flow cytometry (FCM). Small interfering RNAs of AFAP1-AS1 (siAFAP1-AS1) were transfected into the (CD44v6)(+) cells. The levels of AFAP1-AS1 were measured by quantitative real-time PCR (qRT-PCR). Sphere formation assay, cell cycle analysis, and Western blotting were used to detect the effect of siAFAP1-AS1. RNA pull-down and luciferase reporter assay were used to verify the relationship between miR-27b-3p and AFAP1-AS1 or vascular endothelial growth factor (VEGF)-C. Results: CD44v6(+) CC cells had remarkable stemness and a high level of AFAP1-AS1. However, AFAP1-AS1 knockdown with siAFAP1-AS1 suppressed the cell cycle transition of G(1)/S phase and inhibited self-renewal of CD44v6(+) CC cells, the levels of the stemness markers octamer-binding transcription factor 4 (OCT4), osteopontin (OPN), and cluster of differentiation 133 (CD133), and the epithelial-mesenchymal transition (EMT)-related proteins Twist1, matrix metalloprotease (MMP)-9, and VEGF-C. In the mechanism study, miR-27b-3p/VEGF-C signaling was demonstrated to be a key downstream of AFAP1-AS1 in the CD44v6(+) CC cells. Conclusions: LncRNA AFAP1-AS1 knockdown inhibits the CC cell stemness by upregulating miR-27b-3p to suppress VEGF-C.
Collapse
Affiliation(s)
- Meng Xia
- School of Pharmacy, Minzu University of China, Beijing 100081, China Department of Orthopedics, Bayannaoer City Hospital, Bayannaoer, Inner Mongolia 015000, China Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing 100081, China
| | | | | | | | | |
Collapse
|
17
|
Jo DH, Lee S, Bak E, Cho CS, Han YT, Kim K, Suh YG, Kim JH. Antitumor Activity of Novel Signal Transducer and Activator of Transcription 3 Inhibitors on Retinoblastoma. Mol Pharmacol 2021; 100:63-72. [PMID: 34016717 DOI: 10.1124/molpharm.120.000231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/13/2021] [Indexed: 11/22/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a plausible therapeutic target in the treatment of retinoblastoma, the most common intraocular malignant tumor in children. STAT3, a transcription factor of several genes related to tumorigenesis, is activated in retinoblastoma tumors as well as other cancers. In this study, we investigated the structure-activity relationship of a library of STAT3 inhibitors, including a novel series of derivatives of the previously reported compound with a Michael acceptor (compound 1). We chose two novel STAT3 inhibitors, compounds 11 and 15, from the library based on their inhibitory effects on the phosphorylation and transcription activity of STAT3. These STAT3 inhibitors effectively suppressed the phosphorylation of STAT3 and inhibited the expression of STAT3-related genes CCND1, CDKN1A, BCL2, BCL2L1, BIRC5, MYC, MMP1, MMP9, and VEGFA Intraocularly administered STAT3 inhibitors decreased the degree of tumor formation in the vitreous cavity of BALB/c nude mice of an orthotopic transplantation model. It is noteworthy that compounds 11 and 15 did not induce in vitro and in vivo toxicity on retinal constituent cells and retinal tissues, respectively, despite their potent antitumor effects. We suggest that these novel STAT3 inhibitors be used in the treatment of retinoblastoma. SIGNIFICANCE STATEMENT: The current study suggests the novel STAT3 inhibitors with Michael acceptors possess antitumor activity on retinoblastoma, the most common intraocular cancer in children. Based on detailed structure-activity relationship studies, we found a 4-fluoro and 3-trifluoro analog (compound 11) and a monochloro analog (compound 15) of the parental compound (compound 1) inhibited STAT3 phosphorylation, leading to suppressed retinoblastoma in vitro and in vivo.
Collapse
Affiliation(s)
- Dong Hyun Jo
- Department of Anatomy and Cell Biology (D.H.J.) Department of Ophthalmology (E.B., J.H.K.), and Department of Biomedical Sciences (J.H.K.), Seoul National University College of Medicine, Seoul, Republic of Korea; College of Pharmacy, CHA University, Pocheon-si, Republic of Korea (S.L., K.K., Y.-G.S.); Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (C.S.C.); and College of Pharmacy, Dankook University, Cheonan-si, Republic of Korea (Y.T.H.)
| | - Seungbeom Lee
- Department of Anatomy and Cell Biology (D.H.J.) Department of Ophthalmology (E.B., J.H.K.), and Department of Biomedical Sciences (J.H.K.), Seoul National University College of Medicine, Seoul, Republic of Korea; College of Pharmacy, CHA University, Pocheon-si, Republic of Korea (S.L., K.K., Y.-G.S.); Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (C.S.C.); and College of Pharmacy, Dankook University, Cheonan-si, Republic of Korea (Y.T.H.)
| | - Eunoo Bak
- Department of Anatomy and Cell Biology (D.H.J.) Department of Ophthalmology (E.B., J.H.K.), and Department of Biomedical Sciences (J.H.K.), Seoul National University College of Medicine, Seoul, Republic of Korea; College of Pharmacy, CHA University, Pocheon-si, Republic of Korea (S.L., K.K., Y.-G.S.); Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (C.S.C.); and College of Pharmacy, Dankook University, Cheonan-si, Republic of Korea (Y.T.H.)
| | - Chang Sik Cho
- Department of Anatomy and Cell Biology (D.H.J.) Department of Ophthalmology (E.B., J.H.K.), and Department of Biomedical Sciences (J.H.K.), Seoul National University College of Medicine, Seoul, Republic of Korea; College of Pharmacy, CHA University, Pocheon-si, Republic of Korea (S.L., K.K., Y.-G.S.); Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (C.S.C.); and College of Pharmacy, Dankook University, Cheonan-si, Republic of Korea (Y.T.H.)
| | - Young Taek Han
- Department of Anatomy and Cell Biology (D.H.J.) Department of Ophthalmology (E.B., J.H.K.), and Department of Biomedical Sciences (J.H.K.), Seoul National University College of Medicine, Seoul, Republic of Korea; College of Pharmacy, CHA University, Pocheon-si, Republic of Korea (S.L., K.K., Y.-G.S.); Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (C.S.C.); and College of Pharmacy, Dankook University, Cheonan-si, Republic of Korea (Y.T.H.)
| | - Kyeojin Kim
- Department of Anatomy and Cell Biology (D.H.J.) Department of Ophthalmology (E.B., J.H.K.), and Department of Biomedical Sciences (J.H.K.), Seoul National University College of Medicine, Seoul, Republic of Korea; College of Pharmacy, CHA University, Pocheon-si, Republic of Korea (S.L., K.K., Y.-G.S.); Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (C.S.C.); and College of Pharmacy, Dankook University, Cheonan-si, Republic of Korea (Y.T.H.)
| | - Young-Ger Suh
- Department of Anatomy and Cell Biology (D.H.J.) Department of Ophthalmology (E.B., J.H.K.), and Department of Biomedical Sciences (J.H.K.), Seoul National University College of Medicine, Seoul, Republic of Korea; College of Pharmacy, CHA University, Pocheon-si, Republic of Korea (S.L., K.K., Y.-G.S.); Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (C.S.C.); and College of Pharmacy, Dankook University, Cheonan-si, Republic of Korea (Y.T.H.)
| | - Jeong Hun Kim
- Department of Anatomy and Cell Biology (D.H.J.) Department of Ophthalmology (E.B., J.H.K.), and Department of Biomedical Sciences (J.H.K.), Seoul National University College of Medicine, Seoul, Republic of Korea; College of Pharmacy, CHA University, Pocheon-si, Republic of Korea (S.L., K.K., Y.-G.S.); Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (C.S.C.); and College of Pharmacy, Dankook University, Cheonan-si, Republic of Korea (Y.T.H.)
| |
Collapse
|
18
|
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: 1.3] [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
|
19
|
Luan L, Hu Q, Wang Y, Lu L, Ling J. Knockdown of lncRNA NEAT1 expression inhibits cell migration, invasion and EMT by regulating the miR-24-3p/LRG1 axis in retinoblastoma cells. Exp Ther Med 2021; 21:367. [PMID: 33732340 PMCID: PMC7903428 DOI: 10.3892/etm.2021.9798] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Retinoblastoma (RB) is the most common primary intraocular cancer type that occurs during retinal development in childhood. Previous studies have reported that long non-coding RNAs (lncRNAs) are involved in the development of RB. Therefore, the aim of the present study was to investigate the effects and underlying regulatory mechanisms of nuclear paraspeckle assembly transcript 1 (NEAT1) in RB. The expression levels of NEAT1, microRNA (miR)-24-3p and leucine-rich-α-2-glycoprotein (LRG1) were detected using reverse transcription-quantitative PCR (RT-qPCR). Moreover, the protein expression levels of LRG1, matrix metalloproteinase 9, N-cadherin and E-cadherin were detected via western blotting. Furthermore, cell migration and invasion abilities were evaluated via Transwell assays. The targeting relationships between miR-24-3p and NEAT1 or LRG1 were predicted using online software and confirmed via dual-luciferase reporter assay. In the present study, NEAT1 and LRG1 were upregulated, and miR-24-3p was downregulated in RB tissues and cells compared with the corresponding healthy tissues and cells. Moreover, miR-24-3p was identified as a target of NEAT and LRG1 was demonstrated to be a direct target gene of miR-24-3p. Knockdown of NEAT1 or LRG1 significantly suppressed RB cell migration and invasion ability, while the effects were reversed by an miR-24-3p inhibitor. In addition, the downregulation of LRG1 caused by miR-24-3p was restored following the overexpression of NEAT1 in RB cells. It was also demonstrated that NEAT1 knockdown inhibited the epithelial-to-mesenchymal transition (EMT) pathway by inhibiting the expression of LRG via targeting miR-24-3p. In conclusion, the present results suggest that silencing of NEAT1 suppresses cell migration, invasion and the EMT process by downregulating LRG1 expression via sponging miR-24-3p in RB, thus indicating that NEAT1 may be a potential candidate for RB treatment.
Collapse
Affiliation(s)
- Lan Luan
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Qiang Hu
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yan Wang
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Lu Lu
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jiaojiao Ling
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
20
|
Quiñones-Díaz BI, Reyes-González JM, Sánchez-Guzmán V, Conde-Del Moral I, Valiyeva F, Santiago-Sánchez GS, Vivas-Mejía PE. MicroRNA-18a-5p Suppresses Tumor Growth via Targeting Matrix Metalloproteinase-3 in Cisplatin-Resistant Ovarian Cancer. Front Oncol 2020; 10:602670. [PMID: 33392094 PMCID: PMC7774672 DOI: 10.3389/fonc.2020.602670] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/17/2020] [Indexed: 12/22/2022] Open
Abstract
Cumulating evidence indicates that dysregulation of microRNAs (miRNAs) plays a central role in the initiation, progression, and drug resistance of cancer cells. However, the specific miRNAs contributing to drug resistance in ovarian cancer cells have not been fully elucidated. Aimed to identify potential miRNAs involved in platinum resistance, we performed a miRNA expression profile in cisplatin-sensitive and cisplatin-resistant ovarian cancer cells, and we found several differentially abundant miRNAs in the pair of cell lines. Notably, miR-18a-5p (miR-18a), a member of the oncogenic associated miR-17-92 cluster, was decreased in cisplatin-resistant as compared with cisplatin-sensitive cells. Real-time PCR analysis confirmed these findings. We then studied the biological, molecular, and therapeutic consequences of increasing the miR-18a levels with oligonucleotide microRNA mimics (OMM). Compared with a negative control OMM, transient transfection of a miR-18a-OMM reduced cell growth, cell proliferation, and cell invasion. Intraperitoneal injections of miR-18a-OMM-loaded folate-conjugated liposomes significantly reduced the tumor weight and the number of nodules in ovarian cancer-bearing mice when compared with a control-OMM group. Survival analysis using the Kaplan-Meier plotter database showed that ovarian cancer patients with high miR-18a levels live longer in comparison to patients with lower miR-18a levels. Bioinformatic analyses, real-time-PCR, Western blots, and luciferase reporter assays revealed that Matrix Metalloproteinase-3 (MMP-3) is a direct target of miR-18a. Small-interfering RNA (siRNA)-mediated silencing of MMP-3 reduced cell viability, cell growth, and the invasiveness potential of cisplatin-resistant ovarian cancer cells. Our study suggests that targeting miR-18a is a plausible therapeutic strategy for cisplatin-resistant ovarian cancer.
Collapse
Affiliation(s)
| | | | | | | | - Fatma Valiyeva
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
| | | | - Pablo E Vivas-Mejía
- Department of Biochemistry, University of Puerto Rico, San Juan, Puerto Rico.,Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
21
|
Liu Z, Huang Y, Zhang F, Tang H, Wang Y. Autophagy-related gene 7 deficiency caused by miR-154-5p overexpression suppresses the cell viability and tumorigenesis of retinoblastoma by increasing cell apoptosis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1451. [PMID: 33313196 PMCID: PMC7723560 DOI: 10.21037/atm-20-6009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background Retinoblastoma is a rare cancer of the retina that accounts for 3% of all childhood cancers. The aim of this study was to illuminate the oncogenic role and potential molecular mechanisms of the microRNA miR-154-5p and autophagy-related gene 7 (ATG7) in retinoblastoma, and to establish a nude mouse model in order to explore new therapeutic horizons for the disease. Methods Quantitative reverse transcription-polymerase chain reaction and western blot were performed to detect the expression levels of miR-154-5p and ATG7. The targeting relationship between miR-154-5p and ATG7 was analyzed by employing the luciferase reporter assay. MiR-154-5p mimic and pcDNA-ATG7 were transfected, either alone or in combination, into Y79 cells. The subsequent in vitro experiments involved four groups: the control group, miR-154-5p group, ATG7 group, and miR-154-5p + ATG7 group. Orthotopic xenograft models were established by injecting BALB/c athymic nude mice with treated and untreated Y79 cells. Results Y79 cells were transfected with miR-NC or miR-154-5p. Compared to those in the control group, the mRNA expression levels of miR-154-5p were increased in the miR-154-5p mimic group; in contrast, decreases were observed in the mRNA and protein expression levels of ATG7. Y79 cells were transfected with PcDNA or pcDNA-ATG7. The mRNA expression level of ATG7 was increased in pcDNA-ATG7 group. MiR-154-5p was found to have an element complementary to the three prime untranslated region of ATG7. Overexpression of miR-154-5p inhibited Y79 cells proliferation and migration, and promoted Y79 cells apoptosis via targeting of ATG7. In the in vivo experiment, the tumors of the miR-154-5p group of mice were significantly reduced in weight. Tumor growth and the protein levels of Survivin were both suppressed when miR-154-5p was overexpressed in vivo; however, cell apoptosis and the protein levels of p21 were promoted. In the miR-154-5p group, the expression levels of miR-154-5p were upregulated compared to those in the control group, but the ATG7 expression level was downregulated. Conclusions MiR-154-5p overexpression downregulated ATG7, which inhibited cell proliferation and apoptosis in vitro, as well as tumor formation in vivo.
Collapse
Affiliation(s)
- Zhenrong Liu
- Department of Health Management Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yan Huang
- Department of Health Management Center, West China Hospital of Sichuan University, Chengdu, China
| | - Fan Zhang
- Department of Health Management Center, West China Hospital of Sichuan University, Chengdu, China
| | - Huairong Tang
- Department of Health Management Center, West China Hospital of Sichuan University, Chengdu, China
| | - Youjuan Wang
- Department of Health Management Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Shermane Lim YW, Xiang X, Garg M, Le MT, Li-Ann Wong A, Wang L, Goh BC. The double-edged sword of H19 lncRNA: Insights into cancer therapy. Cancer Lett 2020; 500:253-262. [PMID: 33221454 DOI: 10.1016/j.canlet.2020.11.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/25/2020] [Accepted: 11/06/2020] [Indexed: 01/03/2023]
Abstract
H19 long non-coding RNA (lncRNA) has many functions in cancer. Some studies have reported that H19 acts as an oncogene and is involved in cancer progression by activating epithelial-mesenchymal transition (EMT), the cell cycle and angiogenesis via mechanisms like microRNA (miRNA) sponging - the binding to and inhibition of miRNA activity. This makes H19 lncRNA a potential target for cancer therapeutics. However, several conflicting studies have also found that H19 suppresses tumour development. In this review, we shed light on the possible reasons for these conflicting findings. We also summarise the current literature on the applications of H19 lncRNA in cancer therapy in many cancers and explore new avenues for future research. This includes the use of H19 in recombinant vectors, chemoresistance, epigenetic regulation, tumour microenvironment alteration and cancer immunotherapy. The relationship between H19 and the master tumour suppressor gene p53 is also explored. In most studies, H19 knockdown via RNA interference (RNAi) or epigenetic silencing inhibits cancer development. Thus, H19 lncRNA could be a promising target for the development of cancer therapeutics. This warrants further investigations into its translational research to improve cancer therapy outcomes.
Collapse
Affiliation(s)
- Yun Wei Shermane Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Manoj Garg
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida, 201313, India
| | - Minh Tn Le
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore, 119228, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore, 119228, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
23
|
Bai RX, Tang ZY. Long non-coding RNA H19 regulates Bcl-2, Bax and phospholipid hydroperoxide glutathione peroxidase expression in spontaneous abortion. Exp Ther Med 2020; 21:41. [PMID: 33273971 DOI: 10.3892/etm.2020.9473] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 06/05/2020] [Indexed: 12/23/2022] Open
Abstract
Spontaneous abortion (SA) is the most frequently occurring pregnancy disorder and is a serious threat to women's health. Identifying novel risk factors and the molecular mechanisms underlying SA are important. The present study reported that the RNA expression levels of long non-coding RNA H19 were lower in SA group compared with those in the control group, and the expression of Bax was increased and levels of Bcl-2 and phospholipid hydroperoxide glutathione peroxidase (GPX4) were decreased in SA group at both the mRNA and protein levels. H19 expression was positively correlated with Bcl-2 and GPX4 expression and negatively linked with Bax levels. It was demonstrated that silencing H19 downregulated Bcl-2 and GPX4 expression and upregulated Bax expression at both the mRNA and protein levels in HTR-8/SVneo trophoblast cells. In conclusion, the present findings suggested that H19 has important roles in SA by promoting apoptosis and ferroptosis.
Collapse
Affiliation(s)
- Ru-Xia Bai
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Zou-Ying Tang
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
24
|
Differential expression, function and prognostic value of miR-17-92 cluster in ER-positive and triple-negative breast cancer. Cancer Treat Res Commun 2020; 25:100224. [PMID: 33096318 DOI: 10.1016/j.ctarc.2020.100224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/02/2020] [Accepted: 10/11/2020] [Indexed: 12/19/2022]
Abstract
Recent evidence has shown that the miR-17-92 cluster can function either as oncogene or tumor suppressor in human cancers. The function of miR-17-92 in subtypes of breast cancer remains largely unknown. The expression of miR-17-92 is elevated in triple negative breast cancer (TNBC) but reduced in estrogen receptor (ER)-positive breast cancer (ERPBC). We show that increased expression of miRNAs belonging to the miR-17-92 cluster is associated with poor outcome in TNBC, whereas the expression of miR-17-92 miRNAs is with good outcome in ERPBC. We show that ectopic expression of miR-17-92 inhibited cell growth and invasion of ER-positive and HER2-enriched cells. On the contrary, miR-17-92 expression enhanced cell growth and invasion of TNBC cells. Further, we found that miR-17-92 expression sensitized MCF7 cells to chemotherapeutic compounds, whereas it rendered SKBR3 cells resistant to them. We found that expression of ADORA1 was reduced by miR-17-92-expressing breast cancer cells, specifically in ERPBC. We observed an inverse correlation between the expression of ADORA1 and miR-17-92 in human breast cancer. Treatment with DPCPX, a selective ADORA1 antagonist, abolished the difference in the growth of control and miR-17-92 overexpressing MCF7 cells and identified ADORA1 as a key functional target of miR-17-92 in ERPBC. Furthermore, increased expression of ADORA1 in ERPBC is associated with a poor outcome. Our observations underscore the context-dependent role of miR-17-92 in breast cancer subtypes and suggest that miR-17-92 could serve as novel prognostic markers in breast cancer.
Collapse
|
25
|
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: 1.8] [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
|
26
|
Gao Y, Luo X, Meng T, Zhu M, Tian M, Lu X. [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 DOI: 10.12122/j.issn.1673-4254.2020.09.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [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)
- Yali Gao
- Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - Xiaoling Luo
- Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - Ting Meng
- Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - Minjuan Zhu
- Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - Meiwen Tian
- Department of Ophthalmology, Second Clinical Medical College of Ji'nan University/Shenzhen People's Hospital, Shenzhen 518020, China
| | - Xiaohe Lu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
27
|
Ding F, Jiang K, Sheng Y, Li C, Zhu H. RETRACTED: LncRNA MIR7-3HG executes a positive role in retinoblastoma progression via modulating miR-27a-3p/PEG10 axis. Exp Eye Res 2020; 193:107960. [PMID: 32035086 DOI: 10.1016/j.exer.2020.107960] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/13/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022]
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 since upon institutional inspection, the reproducibility of the CCK-8 assay was not sufficient and considered not to be valid and therefore could not support the conclusions of the article.
Collapse
Affiliation(s)
- Fengkui Ding
- Department of Ophthalmology, Jining No.1 People's Hospital, No.6 Jiankang Road, Jining, Shandong Province, 272011, PR China
| | - Kai Jiang
- Department of Ophthalmology, Yuhuangding Hospital, Yantai, Shandong Province, 264000, PR China
| | - Yanjuan Sheng
- Department of Ophthalmology, The Second People's Hospital of Jinan, Jinan, Shandong Province, 250001, PR China
| | - Chuanbao Li
- Department of Ophthalmology, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, 272067, PR China
| | - Huaicheng Zhu
- Department of Ophthalmology, Jining No. 1 People's Hospital, Affiliated Jining No. 1 People's Hospital of Jining Medical University, Jining Medical University, Jining, Shandong Province, 272000, PR China.
| |
Collapse
|
28
|
Zhang Y, Li M, Han X. Icariin affects cell cycle progression and proliferation of human retinal pigment epithelial cells via enhancing expression of H19. PeerJ 2020; 8:e8830. [PMID: 32219038 PMCID: PMC7087489 DOI: 10.7717/peerj.8830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/02/2020] [Indexed: 12/23/2022] Open
Abstract
Background Aberrant proliferation of retinal pigment epithelial (RPE) cells under pathologic condition results in the occurrence of proliferative vitreoretinopathy (PVR). Icariin (ICA)-a flavonol glucoside-has been shown to inhibit proliferation of many cell types, but the effect on RPE cells is unknown. This study aimed to clarify the inhibitory effects of ICA on RPE cells against platelet-derived growth factor (PDGF)-BB-induced cell proliferation, and discuss the regulatory function of H19 in RPE cells. Methods MTS assay was conducted to determine the effects of ICA on cell proliferation. Flow cytometry analysis was performed to detect cell cycle progression. Quantitative real-time PCR and western blot assay were used to measure the expression patterns of genes in RPE cells. Results ICA significantly suppressed PDGF-BB-stimulated RPE cell proliferation in a concentration-dependent manner. Moreover, since administration of ICA induced cell cycle G0/G1 phase arrest, the anti-proliferative activity of ICA may be due to G0/G1 phase arrest in RPE cells. At molecular levels, cell cycle regulators cyclin D1, CDK4, CDK6, p21 and p53 were modulated in response to treatment with ICA. Most importantly, H19 was positively regulated by ICA and H19 depletion could reverse the inhibitory effects of ICA on cell cycle progression and proliferation in PDGF-BB-stimulated RPE cells. Further mechanical explorations showed that H19 knockdown resulted in alternative expressions levels of cyclin D1, CDK4, CDK6, p21 and p53 under ICA treatment. Conclusions Our findings revealed that ICA was an effective inhibitor of PDGF-BB-induced RPE cell proliferation through affecting the expression levels of cell cycle-associated factors, and highlighted the potential application of ICA in PVR therapy. H19 was described as a target regulatory gene of ICA whose disruption may contribute to excessive proliferation of RPE cells, suggesting that modulation of H19 expression may be a novel therapeutic approach to treat PVR.
Collapse
Affiliation(s)
- Yibing Zhang
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, China
| | - Min Li
- Department of Pharmacology and Toxicology, Jilin University School of Pharmaceutical Sciences, Changchun, China
| | - Xue Han
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
29
|
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: 3.8] [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
|
30
|
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: 2.3] [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
|
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: 18] [Impact Index Per Article: 4.5] [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
|
Liao B, Wang Z, Zhu Y, Wang M, Liu Y. Long noncoding RNA DRAIC acts as a microRNA-122 sponge to facilitate nasopharyngeal carcinoma cell proliferation, migration and invasion via regulating SATB1. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3585-3597. [PMID: 31497998 DOI: 10.1080/21691401.2019.1656638] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Increasing evidences have revealed that long noncoding RNAs (lncRNAs) are frequently involved in various cancers. However, the expression and function of lncRNA DRAIC in nasopharyngeal carcinoma (NPC) remain unknown. In this study, we found that DRAIC was significantly increased in NPC tissues. Increased expression of DRAIC was positively correlated with advanced clinical stages of NPC patients. Functional assays revealed that ectopic expression of DRAIC enhances NPC cell growth, migration and invasion. DRAIC knockdown represses NPC cell growth, migration and invasion. Mechanistically, we identified two miR-122 binding sites on DRAIC. RNA pull-down, RNA immunoprecipitation, and dual-luciferase reporter assays confirmed the binding of DRAIC to miR-122. Via binding of miR-122, DRAIC upregulated the expression of miR-122 target SATB1, which was abolished by the mutation of miR-122 binding sites on SATB1. Moreover, the oncogenic roles of DRAIC on NPC were reversed by the mutation of miR-122 binding sites on SATB1, simultaneous overexpression of miR-122, or depletion of SATB1. In addition, the expression of SATB1 was also increased and positively associated with that of DRAIC in NPC tissues. In conclusion, these findings revealed the important roles of DRAIC-miR-122-SATB1 axis in NPC and suggested that DRAIC may be a potential therapeutic target for NPC.
Collapse
Affiliation(s)
- Bing Liao
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| | - Zhi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| | - Yaqiong Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| | - Meiqun Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| | - Yuehui Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University , Nanchang , China
| |
Collapse
|
33
|
Ma J, Chen W, Wang K, Tian K, Li Q, Zhao T, Zhang L, Wang L, Wu Z, Zhang J. Identification of the Different Roles and Potential Mechanisms of T Isoforms in the Tumor Recurrence and Cell Cycle of Chordomas. Onco Targets Ther 2019; 12:11777-11791. [PMID: 32099384 PMCID: PMC6997418 DOI: 10.2147/ott.s232526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose The roles of T (brachyury) isoforms in chordomas remain unclear. This study aimed to investigate the different roles and mechanisms of them in chordomas. Patients and methods The expression of T isoforms mRNAs in 57 chordomas was assessed, and a prognosis analysis was conducted. Cell apoptosis, proliferation and cell cycle assays were performed after specific T isoform mRNA knockdown. Whole-transcriptome sequencing, Gene Set Enrichment Analysis, Gene Ontology analysis, Kyoto Encyclopedia of Genes and Genomes analysis and competing endogenous RNA (ceRNA) analysis were conducted. Results As revealed in this study, the T-long isoform was a significant risk factor (hazard ratio [HR], 1.09; P=0.018) and the T-short isoform was a protective factor (HR, 0.24; P=0.012) associated with tumor recurrence. After T-long isoform knockdown, the cell cycle was arrested at G0/G1 phase and cell proliferation was significantly inhibited. A bioinformatic analysis revealed that the upregulation of H19, P21 and GADD45B; downregulation of SKP2 and CDK2; and accompanying changes in the P53 signaling pathway consistently contributed to G0/G1 arrest. After T-short isoform knockdown, the cell cycle was arrested at G2/M phase and cell apoptosis tended to increase slightly (P=0.067). The upregulation of YWHAZ and downregulation of E2F1 and its target genes might contribute to cell cycle arrest in G2/M phase and apoptosis. In addition, the ceRNA network, consisting of long noncoding RNAs, mRNAs and microRNAs, was established. Conclusion The T-long isoform was a risk factor and the T-short isoform was a protective factor for chordoma recurrence. In addition, the cell cycle was the main target of T isoforms knockdown, and the changes in the downstream transcriptome may contribute to the different effects of specific T isoform knockdown on the changes in the cell cycle distributions and apoptosis and proliferation of chordoma cells.
Collapse
Affiliation(s)
- Junpeng Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Kaibing Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Qi Li
- China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China
| | - Tianna Zhao
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.,Beijing Key Laboratory of Brian Tumor, Beijing, People's Republic of China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.,Beijing Key Laboratory of Brian Tumor, Beijing, People's Republic of China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.,Beijing Key Laboratory of Brian Tumor, Beijing, People's Republic of China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.,Beijing Key Laboratory of Brian Tumor, Beijing, People's Republic of China
| |
Collapse
|
34
|
Wang Q, Zhu Y, Zuo G, Chen X, Cheng J, Zhang S. LINC00858 promotes retinoblastoma cell proliferation, migration and invasion by inhibiting miR-3182. Exp Ther Med 2019; 19:999-1005. [PMID: 32010262 PMCID: PMC6966175 DOI: 10.3892/etm.2019.8294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 10/02/2019] [Indexed: 01/13/2023] Open
Abstract
The aim of the present study was to determine the role of long intergenic non-protein coding RNA 858 (LINC00858) in retinoblastoma (RB) and investigate the underlying molecular mechanisms. RB tissues and paracancerous tissues of 27 RB cases were obtained. RB cell lines (SO-RB50, Y79, HXO-RB44 and WERI-Rb1) and a normal retinal epithelial cell line (ARPE-19) were cultured for in vitro experiments. Batches of SO-RB50 and Y79 cells were assigned to groups transfected with small interfering RNA targeting LINC00858 (si-LINC00858 group), microRNA (miR)-3182 mimics or inhibitor, or the respective controls. A Cell Counting Kit-8 and Transwell assays were performed to assess the effect of the transfections on the proliferation, migration and invasion of SO-RB50 and Y79 cells. A luciferase reporter assay was performed using SO-RB50 cells to demonstrate the direct binding of LINC00858 and miR-3182. Reverse transcription-quantitative PCR was employed to detect LINC00858 and miR-3182 expression. Pearson correlation analysis was used to assess the correlation between the expression of LINC00858 and miR-3182. The results indicated that RB tissues and cells exhibited aberrantly elevated LINC00858 expression (P<0.05). Compared with those in the control-transfected group, SO-RB50 and Y79 cells of the si-LINC00858 group had a lower cell proliferation, as well as a lower number of migrated and invaded cells (all P<0.05). miR-3182 was proven to be a target gene of LINC00858, to be abnormally downregulated in RB tissues and cells (P<0.05) and to be negatively correlated with LINC00858 expression. Compared with those in the si-LINC00858 + inhibitor-negative control group, SO-RB50 and Y79 cells of the si-LINC00858 + miR-3182 inhibitor group exhibited a significantly higher relative proliferation, migration and invasion (all P<0.05). In conclusion, LINC00858 promoted RB cell proliferation, migration and invasion, at least partially by inhibiting miR-3182.
Collapse
Affiliation(s)
- Qi Wang
- Department of Ophthalmology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Yanni Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Guojin Zuo
- Department of Ophthalmology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Xiaoming Chen
- Department of Ophthalmology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jinkui Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Shu Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
35
|
Abstract
Retinoblastoma (Rb) is the most common ocular pediatric malignancy that arises from the retina and is caused by a mutation of the two alleles of the tumor suppressor gene, RB1. Although early detection provides the opportunity of controlling the primary tumor with effective therapies, metastatic activity is fatal. Non-coding RNAs (ncRNAs) have emerged as important modifiers of a plethora of biological mechanisms including those involved in cancer. They are classified into short and long ncRNAs according to their length. Deregulation of all these molecules has also been shown to play a critical role in Rb pathogenesis and progression. It is believed that ncRNAs can provide new insights into novel regulatory mechanisms associated with clinical pathological characteristics, facilitating the development of therapeutic alternatives for the treatment of Rb. In this review, we describe a variety of ncRNAs, which capable of regulating the most likely candidate genes involved in the tumorigenesis of Rb, could prove useful in analyzing different aspects of this cancer.
Collapse
Affiliation(s)
- Meropi Plousiou
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Ivan Vannini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
36
|
Ye Y, Shen A, Liu A. Long non-coding RNA H19 and cancer: A competing endogenous RNA. Bull Cancer 2019; 106:1152-1159. [PMID: 31753509 DOI: 10.1016/j.bulcan.2019.08.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/29/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022]
Abstract
Long non-coding RNA (lncRNA) is a class of non-coding RNA with a length of more than 200 nucleotides, which has become a hotspot in the research of tumorigenesis and development in recent years. Accumulating studies have indicated that H19 is abnormally expressed in human malignant tumors, and regulates cell proliferation, migration, invasion, anti-apoptosis and epithelial-mesenchymal transition through various mechanisms, thus playing a carcinogenic or anti-cancer role. H19 has been found to act as a microRNA sponge to indirectly regulate the expression of microRNA downstream target genes thus mediating cancer progression in several cancer types. Even in the same cancer, H19 also sponges various microRNAs to mediate diverse regulatory mechanisms. Tissue-specific expression of H19 suggests that it may be an early diagnostic marker or prognostic indicator of cancers. In this review, we summarize the latest original researches, mainly focusing on the role of H19 sponging microRNAs in cancers. We hope this article can facilitate readers obtain the molecular mechanisms of H19 sponging miRNAs in cancers and provide a broad perspective for further research on cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Yafen Ye
- Nanchang University, The Second Clinical Medical College, 461, Bayi Road, Nanchang, Jiangxi, China; Jiangxi key laboratory of clinical translational cancer research, 1, Minde Road, Nanchang, Jiangxi, China
| | - Ao Shen
- Nanchang University, The Second Clinical Medical College, 461, Bayi Road, Nanchang, Jiangxi, China
| | - Anwen Liu
- The Second Affiliated Hospital of Nanchang University, Department of Oncology, 1, Minde Road, Nanchang, Jiangxi, China; Jiangxi key laboratory of clinical translational cancer research, 1, Minde Road, Nanchang, Jiangxi, China.
| |
Collapse
|
37
|
Tao S, Wang W, Liu P, Wang H, Chen W. Long non-coding RNA T-cell leukemia/lymphoma 6 serves as a sponge for miR-21 modulating the cell proliferation of retinoblastoma through PTEN. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:449-458. [PMID: 31680766 PMCID: PMC6819906 DOI: 10.4196/kjpp.2019.23.6.449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 12/28/2018] [Accepted: 04/15/2019] [Indexed: 12/20/2022]
Abstract
Retinoblastoma (Rb) is one of the most common eye malignancies occur in childhood. The crucial roles of non-coding RNAs, particularly long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), have been widely reported in Rb progression. In the present study, we found the expression of lncRNA T-cell leukemia/lymphoma 6 (TCL6) was significantly downregulated in Rb tissues and cell lines. Knockdown of lncRNA TCL6 promoted cell proliferation while reduced cell apoptosis in Rb cells. Moreover, lncRNA TCL6 serves as a sponge for miR-21, a previously-reported oncogenic miRNA in Rb, by direct targeting to negatively regulated miR-21 expression, therefore modulating Rb proliferation through miR-21. TCL6 overexpression inhibited Rb cell proliferation while miR-21 overexpression exerted an opposing effect; the effect of TCL6 overexpression was partially attenuated by miR-21 overexpression. PTEN/PI3K/AKT signaling pathway was involved in lncRNA TCL6/miR-21 axis modulating Rb cell proliferation. Taken together, lncRNA TCL6 serves as a tumor suppressor by acting as a sponge for miR-21 to counteract miR-21-mediated PTEN repression.
Collapse
Affiliation(s)
- Sisi Tao
- Department of Science and Education, Changsha Hospital for Maternal & Child Health Care, Changsha 410007, Hunan, China
| | - Weidong Wang
- Department of Science and Education, Changsha Hospital for Maternal & Child Health Care, Changsha 410007, Hunan, China
| | - Pengfei Liu
- Department of Orthopedics, Shaoyang County People's Hospital, Shaoyang 422100, Hunan, China
| | - Hua Wang
- Department of Ophthalmology, The First Affiliated Hospital of Human Normal University/Hunan Provincial People's Hospital, Changsha 410002, Hunan, China
| | - Weirong Chen
- Health Management Center, Changsha Hospital for Maternal & Child Health Care, Changsha 410007, Hunan, China
| |
Collapse
|
38
|
Smith CM, Catchpoole D, Hutvagner G. Non-Coding RNAs in Pediatric Solid Tumors. Front Genet 2019; 10:798. [PMID: 31616462 PMCID: PMC6764412 DOI: 10.3389/fgene.2019.00798] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 07/30/2019] [Indexed: 12/15/2022] Open
Abstract
Pediatric solid tumors are a diverse group of extracranial solid tumors representing approximately 40% of childhood cancers. Pediatric solid tumors are believed to arise as a result of disruptions in the developmental process of precursor cells which lead them to accumulate cancerous phenotypes. In contrast to many adult tumors, pediatric tumors typically feature a low number of genetic mutations in protein-coding genes which could explain the emergence of these phenotypes. It is likely that oncogenesis occurs after a failure at many different levels of regulation. Non-coding RNAs (ncRNAs) comprise a group of functional RNA molecules that lack protein coding potential but are essential in the regulation and maintenance of many epigenetic and post-translational mechanisms. Indeed, research has accumulated a large body of evidence implicating many ncRNAs in the regulation of well-established oncogenic networks. In this review we cover a range of extracranial solid tumors which represent some of the rarer and enigmatic childhood cancers known. We focus on two major classes of ncRNAs, microRNAs and long non-coding RNAs, which are likely to play a key role in the development of these cancers and emphasize their functional contributions and molecular interactions during tumor formation.
Collapse
Affiliation(s)
- Christopher M Smith
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | - Daniel Catchpoole
- School of Software, University of Technology Sydney, Sydney, Australia.,The Tumour Bank-CCRU, Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Gyorgy Hutvagner
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
39
|
Yang M, Wei W. Long non-coding RNAs in retinoblastoma. Pathol Res Pract 2019; 215:152435. [PMID: 31202519 DOI: 10.1016/j.prp.2019.152435] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/15/2019] [Accepted: 05/03/2019] [Indexed: 12/31/2022]
Abstract
Retinoblastoma represents 3% of all childhood cancers and is the most common intraocular malignant tumor with a highly aggressive and metastatic phenotype. While recent genetic and epigenetic studies have reported new insights into the mechanism of retinoblastoma development, the involvement of regulatory non-coding RNAs remains unclear. Long non-coding RNAs (lncRNAs) are a group of endogenous non-protein-coding RNAs with the capacity to regulate gene expression at multiple levels. Recent evidence has shown that lncRNAs can regulate many cellular processes, such as cell proliferation, differentiation, migration, and invasion. Several lncRNAs, including BANCR, AFAP1-AS1, NEAT1, XIST, ANRIL, PlncRNA-1, HOTAIR, PANDAR, DANCR, and THOR, promote the progression and metastasis of retinoblastoma. However, some lncRNAs, such as MEG3, MT1JP, and H19, play a tumor suppressive role. Our review summarizes the functional role of lncRNAs in retinoblastoma and their potential clinical applications for diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Ming Yang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wenbin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
40
|
Han N, Zuo L, Chen H, Zhang C, He P, Yan H. Long non-coding RNA homeobox A11 antisense RNA (HOXA11-AS) promotes retinoblastoma progression via sponging miR-506-3p. Onco Targets Ther 2019; 12:3509-3517. [PMID: 31123409 PMCID: PMC6511239 DOI: 10.2147/ott.s195404] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/16/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Long non-coding RNA homeobox A11 antisense RNA (HOXA11-AS) has been reported to be involved in initiation and development of multiple cancers. However, the detailed biological roles and underlying molecular mechanism of HOXA11-AS remain unclear in retinoblastoma (RB). Herein, the goals of this study were to explore the biological function and the potential mechanism of HOXA11-AS in RB. Materials and methods: The expression of HOXA11-AS in RB tissues and cell lines was detected using real-time PCR (qRT-PCR). Cell proliferation, cycle arrest and apoptosis were measured using a cell counting kit 8 and flow cytometry. The target miRNAs of HOXA11-AS was predicted by Starbase2.0 software and was confirmed using a dual-luciferase reporter assay. Result: We found that HOXA11-AS expression was markedly elevated in RB tissues and cell lines compared to normal retina tissues and human retinal epithelial cells, respectively. Functional analysis showed that knockdown of HOXA11-AS in RB cells significantly suppressed cell proliferation, and induced cell cycle arrest at G1/G0 phase and promoted cell apoptosis. We also found that HOXA11-AS could serve as a competing endogenous RNA (ceRNA) that inhibited miR-506-3p expression, which regulated its downstream target NIMA-related kinase 6 (NEK6) in RB. In addition, miR-506-3p inhibitors partially reversed the effect of HOXA11-AS depletion on proliferation, cycle arrest and apoptosis in RB cells. Conclusion: Taken together, these findings demonstrated that HOXA11-AS could promote RB progression by sponging miR-506-3p, suggesting that HOXA-11-AS might be a potential therapeutic target for RB.
Collapse
Affiliation(s)
- Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Ling Zuo
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Han Chen
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Chunxia Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Pei He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| | - Hongtao Yan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, People's Republic of China
| |
Collapse
|
41
|
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: 8.6] [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
|
42
|
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: 6.8] [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
|
43
|
Zhong W, Yang J, Li M, Li L, Li A. Long noncoding RNA NEAT1 promotes the growth of human retinoblastoma cells via regulation of miR-204/CXCR4 axis. J Cell Physiol 2018; 234:11567-11576. [PMID: 30479013 DOI: 10.1002/jcp.27812] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/05/2018] [Indexed: 12/22/2022]
Abstract
Retinoblastoma (RB) is an aggressive eye cancer of infancy and childhood with high mortality. Studies have shown that long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) is closely related to the progression of multiple cancers. However, its role in RB remains unknown. This study aimed to investigate the role and underlying mechanism of NEAT1 in RB. We first detected the expression of NEAT1 in human RB tissues and cell lines. The effects of NEAT1 on the proliferation, migration, and apoptosis of RB cells were analyzed by loss-of-function. The underlying mechanism of NEAT1 in RB was mainly focused on the microRNA 204/C-X-C chemokine receptor type 4 (miR-204/CXCR4) axis. In addition, the role and mechanism of NEAT1 in RB were further evaluated in a mouse xenograft tumor model. We found NEAT1 and CXCR4 expression levels were elevated, whereas miR-204 expression was decreased in RB tissues and cells. Downregulation of NEAT1 significantly decreased the proliferation and migration but promoted the apoptosis of RB cells. NEAT1 functioned as a competing endogenous RNA for miR-204 to regulate CXCR4 expression. Knockdown of NEAT1 suppressed the tumor volume, tumor weight, and CXCR4 expression, whereas increased miR-204 expression in mice. In conclusion, NEAT1 promotes the development of RB via miR-204/CXCR4 axis, which provides a new target for the treatment of RB disease.
Collapse
Affiliation(s)
- Wei Zhong
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jingpu Yang
- Department of Otolaryngology-Head and Neck Surgery, Second Hospital of Jilin University, Changchun, China
| | - Miyang Li
- Department of Clinical Laboratory, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lin Li
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Aipeng Li
- Department of Ophthalmology, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
44
|
Cheng Y, Chang Q, Zheng B, Xu J, Li H, Wang R. LncRNA XIST promotes the epithelial to mesenchymal transition of retinoblastoma via sponging miR-101. Eur J Pharmacol 2018; 843:210-216. [PMID: 30472203 DOI: 10.1016/j.ejphar.2018.11.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/17/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022]
Abstract
Accumulating evidence demonstrated that abnormal expression of long non-coding RNAs (lncRNAs) was closely associated with cancer development including retinoblastoma (RB). LncRNA X inactive specific transcript (XIST) has been found to function as an oncogene or a tumor suppressor in several cancers. However, the role and underlying mechanism of XIST in RB have not been clarified. The expression of XIST, microRNA (miR)- 101, zinc finger E-box binding homeobox (ZEB) 1, and ZEB2 was detected in human RB tissues and cell lines. The effects of XIST on the proliferation, migration, invasion, epithelial to mesenchymal transition (EMT), and apoptosis of RB cells were evaluated after downregulation of XIST. Furthermore, the mechanism of XIST was mainly focused on miR-101/ZEB1 or ZEB2 signaling. We found the expression of XIST, ZEB1 and ZEB2 was increased, whereas miR-101 was reduced in RB tissues and cells. Knockdown of XIST significantly suppressed the proliferation, migration, invasion and EMT, but promoted the apoptosis and caspase-3 activity. Moreover, we found that XIST functioned as a competing endogenous RNA (ceRNA) for miR-101 to regulate the de-repression of its endogenous targets ZEB1 and ZEB2. In conclusion, these findings suggest that XIST may facilitate the progression of RB through acting as a ceRNA for miR-101 to mediate the expression of ZEB1 and ZEB2. This may provide novel therapeutic options for RB.
Collapse
Affiliation(s)
- Yan Cheng
- Ophthalmology of Xi'an First Hospital, Xi'an, Shaanxi 710002, China
| | - Qing Chang
- Ophthalmology of Xi'an First Hospital, Xi'an, Shaanxi 710002, China
| | - Bo Zheng
- Ophthalmology of Xi'an First Hospital, Xi'an, Shaanxi 710002, China.
| | - Jing Xu
- Ophthalmology of Xi'an First Hospital, Xi'an, Shaanxi 710002, China
| | - Hongsong Li
- Ophthalmology of Xi'an First Hospital, Xi'an, Shaanxi 710002, China
| | - Ruoxin Wang
- Ophthalmology of Xi'an First Hospital, Xi'an, Shaanxi 710002, China
| |
Collapse
|
45
|
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: 2.5] [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
|
46
|
Yang G, Fu Y, Lu X, Wang M, Dong H, Li Q. LncRNA HOTAIR/miR-613/c-met axis modulated epithelial-mesenchymal transition of retinoblastoma cells. J Cell Mol Med 2018; 22:5083-5096. [PMID: 30030888 PMCID: PMC6156449 DOI: 10.1111/jcmm.13796] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/16/2018] [Accepted: 05/26/2018] [Indexed: 12/13/2022] Open
Abstract
Since lncRNAs could modulate neoplastic development by modulating downstream miRNAs and genes, this study was carried out to figure out the synthetic contribution of HOTAIR, miR-613 and c-met to viability, apoptosis and proliferation of retinoblastoma cells. Totally 276 retinoblastoma tissues and tumour-adjacent tissues were collected, and human retinoblastoma cell lines (ie, Y79, HXO-Rb44, SO-Rb50 and WERI-RB1) were also gathered. Moreover, transfections of pcDNA3.1-HOTAIR, si-HOTAIR, miR-613 mimic, miR-613 inhibitor, pcDNA3.1/c-met were performed to evaluate the influence of HOTAIR, miR-613 and c-met on viability, apoptosis and epithelial-mesenchymal transition (EMT) of retinoblastoma cells. Dual-luciferase reporter gene assay was also arranged to confirm the targeted relationship between HOTAIR and miR-613, as well as between miR-613 and c-met. Consequently, up-regulated HOTAIR and down-regulated miR-613 expressions displayed associations with poor survival status of retinoblastoma patients (P < 0.05). Besides, inhibited HOTAIR and promoted miR-613 elevated E-cadherin expression, yet decreased Snail and Vimentin expressions (P < 0.05). Simultaneously, cell proliferation and cell viability were also less-motivated (P < 0.05). Nonetheless, c-met prohibited the functioning of miR-613, resulting in promoted cell proliferation and viability, along with inhibited cell apoptosis (P < 0.05). Finally, HOTAIR was verified to directly target miR-613, and c-met was the direct target gene of miR-613 (P < 0.05). In conclusion, the role of lncRNA HOTAIR/miR-613/c-met signalling axis in modulating retinoblastoma cells' viability, apoptosis and expressions of EMT-specific proteins might provide evidences for developing appropriate diagnostic and treatment strategies for retinoblastoma.
Collapse
Affiliation(s)
- Ge Yang
- Department of OphthalmologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityChina
| | - Yang Fu
- Department of General SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityChina
| | - Xiaoyan Lu
- Department of OphthalmologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityChina
| | - Menghua Wang
- Department of OphthalmologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityChina
| | - Hongtao Dong
- Department of OphthalmologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityChina
| | - Qiuming Li
- Department of OphthalmologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou CityChina
| |
Collapse
|
47
|
Hu C, Liu S, Han M, Wang Y, Xu C. Knockdown of lncRNA XIST inhibits retinoblastoma progression by modulating the miR-124/STAT3 axis. Biomed Pharmacother 2018; 107:547-554. [PMID: 30114638 DOI: 10.1016/j.biopha.2018.08.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/21/2018] [Accepted: 08/06/2018] [Indexed: 10/28/2022] Open
Abstract
Long non-coding RNA (lncRNA) X-inactive specific transcript (XIST) was reportedly to be tightly associated with tumorigenesis and progression of multiple cancers. However, the expression, biological function, and action mechanisms of XIST in retinoblastoma (RB) are still unknown. Here, we found that XIST expression was upregulated in RB tissues and cell lines, and that increased XIST expression was positively associated with advanced cTNM stage (III-V) and late differentiation status. We also revealed that knockdown of XIST inhibited RB cell proliferation, promoted cell cycle at G1/G0 phase, and induced cell apoptosis. Mechanistically, XIST directly bound to microRNA (miR)-124 in RB cells. XIST mRNA expression was inversely correlated with miR-124 in RB tissues. Importantly, miR-124 inhibition partially reversed the effect on cell proliferation, cycle arrest and apoptosis by XIST knockdown mediated. In addition, XIST could regulate expression of signal transducer and activator of transcription 3(STAT3), a directly target of miR-124 in RB. These findings implied that XIST promoted RB progression partially by modulating the miR-124/STAT3 axis.
Collapse
Affiliation(s)
- Chunmei Hu
- Department of Tumor and Hematology, The Second Hospital of Jilin University, Nanguan District, Changchun, Jilin, 130041, PR China
| | - Shu Liu
- Department of Ophthalmology, The Second Hospital of Jilin University, Nanguan District, Changchun, Jilin, 130041, PR China
| | - Mei Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Nanguan District, Changchun, Jilin, 130041, PR China
| | - Yingxue Wang
- Department of Electrical Diagnosis, The Second Hospital of Jilin University, Nanguan District, Changchun, Jilin, 130041, PR China
| | - Chunling Xu
- Department of Ophthalmology, The Second Hospital of Jilin University, Nanguan District, Changchun, Jilin, 130041, PR China.
| |
Collapse
|
48
|
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: 2.3] [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
|
49
|
Elchuri SV, Rajasekaran S, Miles WO. RNA-Sequencing of Primary Retinoblastoma Tumors Provides New Insights and Challenges Into Tumor Development. Front Genet 2018; 9:170. [PMID: 29868118 PMCID: PMC5966869 DOI: 10.3389/fgene.2018.00170] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/26/2018] [Indexed: 12/17/2022] Open
Abstract
Retinoblastoma is rare tumor of the retina caused by the homozygous loss of the Retinoblastoma 1 tumor suppressor gene (RB1). Loss of the RB1 protein, pRB, results in de-regulated activity of the E2F transcription factors, chromatin changes and developmental defects leading to tumor development. Extensive microarray profiles of these tumors have enabled the identification of genes sensitive to pRB disruption, however, this technology has a number of limitations in the RNA profiles that they generate. The advent of RNA-sequencing has enabled the global profiling of all of the RNA within the cell including both coding and non-coding features and the detection of aberrant RNA processing events. In this perspective, we focus on discussing how RNA-sequencing of rare Retinoblastoma tumors will build on existing data and open up new area's to improve our understanding of the biology of these tumors. In particular, we discuss how the RB-research field may be to use this data to determine how RB1 loss results in the expression of; non-coding RNAs, causes aberrant RNA processing events and how a deeper analysis of metabolic RNA changes can be utilized to model tumor specific shifts in metabolism. Each section discusses new opportunities and challenges associated with these types of analyses and aims to provide an honest assessment of how understanding these different processes may contribute to the treatment of Retinoblastoma.
Collapse
Affiliation(s)
- Sailaja V. Elchuri
- Department of Nanotechnology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Swetha Rajasekaran
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- Center for RNA Biology, The Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States
| | - Wayne O. Miles
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- Center for RNA Biology, The Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|