51
|
The DNMT1/miR-34a Axis Is Involved in the Stemness of Human Osteosarcoma Cells and Derived Stem-Like Cells. Stem Cells Int 2019; 2019:7028901. [PMID: 31781245 PMCID: PMC6875320 DOI: 10.1155/2019/7028901] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/25/2019] [Indexed: 12/23/2022] Open
Abstract
The DNA methyltransferase 1 (DNMT1)/miR-34a axis promoted carcinogenesis of various types of cancers. However, no literature reported its contribution to the stemness of osteosarcoma cancer stem-like cells (OSLCs). We sought to determine whether the DNMT1/miR-34a axis facilitates the stemness of OSLCs. We here revealed the higher DNMT1 activity and expression, lower miR-34a expression with high methylation of its promoter, and stronger stemness of OSLCs, as manifested by elevated sphere and colony formation capacities; CD133, CD44, ABCG2, Bmi1, Sox2, and Oct4 protein amounts in vitro; and carcinogenicity in a nude mouse xenograft model, when compared to the parental U2OS cells. 5-Azacytidine (Aza-dC) repressed DNMT1 activation and upregulated miR-34a expression by promoter demethylation and suppressed the stemness of OSLCs in a dose-dependent manner. DNMT1 knockdown increased miR-34a and reduced the stemness of OSLCs. Transfection with a miR-34a mimic repressed the stemness of OSLCs but did not alter DNMT1 activity and expression. Conversely, DNMT1 overexpression declined miR-34a levels, promoting the stemness of U2OS cells. Transfection with a miR-34a inhibitor enhanced the stemness of U2OS cells, without affecting the DNMT1 activity and expression. Importantly, reexpression of miR-34a could rescue the effects of DNMT1 overexpression on miR-34a inhibition as well as the stemness promotion without affecting the activity and expression of DNMT1. Our results revealed that aberrant activation of DNMT1 caused promoter methylation of miR-34a, leading to miR-34a underexpression, and the role of the DNMT1/miR-34a axis in promoting and sustaining the stemness of OSLCs.
Collapse
|
52
|
Galamb O, Barták BK, Kalmár A, Nagy ZB, Szigeti KA, Tulassay Z, Igaz P, Molnár B. Diagnostic and prognostic potential of tissue and circulating long non-coding RNAs in colorectal tumors. World J Gastroenterol 2019; 25:5026-5048. [PMID: 31558855 PMCID: PMC6747286 DOI: 10.3748/wjg.v25.i34.5026] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/26/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are members of the non-protein coding RNA family longer than 200 nucleotides. They participate in the regulation of gene and protein expression influencing apoptosis, cell proliferation and immune responses, thereby playing a critical role in the development and progression of various cancers, including colorectal cancer (CRC). As CRC is one of the most frequently diagnosed malignancies worldwide with high mortality, its screening and early detection are crucial, so the identification of disease-specific biomarkers is necessary. LncRNAs are promising candidates as they are involved in carcinogenesis, and certain lncRNAs (e.g., CCAT1, CRNDE, CRCAL1-4) show altered expression in adenomas, making them potential early diagnostic markers. In addition to being useful as tissue-specific markers, analysis of circulating lncRNAs (e.g., CCAT1, CCAT2, BLACAT1, CRNDE, NEAT1, UCA1) in peripheral blood offers the possibility to establish minimally invasive, liquid biopsy-based diagnostic tests. This review article aims to describe the origin, structure, and functions of lncRNAs and to discuss their contribution to CRC development. Moreover, our purpose is to summarise lncRNAs showing altered expression levels during tumor formation in both colon tissue and plasma/serum samples and to demonstrate their clinical implications as diagnostic or prognostic biomarkers for CRC.
Collapse
Affiliation(s)
- Orsolya Galamb
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
| | - Barbara K Barták
- 2nd Department of Internal Medicine, Semmelweis University, Budapest H-1088, Hungary
| | - Alexandra Kalmár
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
| | - Zsófia B Nagy
- 2nd Department of Internal Medicine, Semmelweis University, Budapest H-1088, Hungary
| | - Krisztina A Szigeti
- 2nd Department of Internal Medicine, Semmelweis University, Budapest H-1088, Hungary
| | - Zsolt Tulassay
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
| | - Peter Igaz
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
- 2nd Department of Internal Medicine, Semmelweis University, Budapest H-1088, Hungary
| | - Béla Molnár
- Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest H-1088, Hungary
| |
Collapse
|
53
|
Amirkhah R, Naderi-Meshkin H, Shah JS, Dunne PD, Schmitz U. The Intricate Interplay between Epigenetic Events, Alternative Splicing and Noncoding RNA Deregulation in Colorectal Cancer. Cells 2019; 8:cells8080929. [PMID: 31430887 PMCID: PMC6721676 DOI: 10.3390/cells8080929] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) results from a transformation of colonic epithelial cells into adenocarcinoma cells due to genetic and epigenetic instabilities, alongside remodelling of the surrounding stromal tumour microenvironment. Epithelial-specific epigenetic variations escorting this process include chromatin remodelling, histone modifications and aberrant DNA methylation, which influence gene expression, alternative splicing and function of non-coding RNA. In this review, we first highlight epigenetic modulators, modifiers and mediators in CRC, then we elaborate on causes and consequences of epigenetic alterations in CRC pathogenesis alongside an appraisal of the complex feedback mechanisms realized through alternative splicing and non-coding RNA regulation. An emphasis in our review is put on how this intricate network of epigenetic and post-transcriptional gene regulation evolves during the initiation, progression and metastasis formation in CRC.
Collapse
Affiliation(s)
- Raheleh Amirkhah
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
- Nastaran Center for Cancer Prevention (NCCP), Mashhad 9185765476, Iran
| | - Hojjat Naderi-Meshkin
- Nastaran Center for Cancer Prevention (NCCP), Mashhad 9185765476, Iran
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad 9177949367, Iran
| | - Jaynish S Shah
- Gene & Stem Cell Therapy Program Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Philip D Dunne
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Ulf Schmitz
- Gene & Stem Cell Therapy Program Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia.
- Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia.
- Computational BioMedicine Laboratory Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia.
| |
Collapse
|
54
|
Cancer stem cell fate determination: a nuclear phenomenon. THE NUCLEUS 2019. [DOI: 10.1007/s13237-019-00281-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
55
|
Lu H, Hao L, Yang H, Chen J, Liu J. miRNA-34a suppresses colon carcinoma proliferation and induces cell apoptosis by targeting SYT1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2887-2897. [PMID: 31934125 PMCID: PMC6949727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 04/23/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND MicroRNAs are emerging as the important regulators in cancer-related processes. This research were performed to find the function and mechanism of miR-34a effect on colon cancer. METHODS In this study, we examined the expression of miR-34a in colon cancer tissues and cell lines by qRT-PCR. In vitro cell functional assays studies were built to define miR-34a and SYT1 function involved in cell growth, migration, and invasion and apoptosis. EGFP reporter assay was used to determine the relationship of SYT1 and miR-181a. To confirmed the relationship between SYT1 and miR-34a, the SYT1 restoration rescued miR-34a mediated growth and inhibited cell apoptosis were detect. RESULT Our studies show that microRNA-34a (miR-34a) is downregulated in human colon cancer relative to normal colon mucosal epithelial cells, and downexpression of miR-34a promotes cell proliferation, migration, and invasion, nevertheless overexpression of miR-34 facilitates cell apoptosis in vitro. Furthermore, SYT1 3'-UTR is found to be down-regulated directly by miR-34a, demonstrating that SYT1 is a important target of miR-34a in colon cancer. The knockdown of SYT1 markedly inhibits colon cancer cell proliferation, migration, and invasion, and induces cell apoptosis, indicating that SYT1 may function as an oncogene in colon cancer. The restoration of SYT1 expression can counteract the effect of miR-34a on cell proliferation, and induces cell apoptosis, of colon cancer cells. CONCLUSION Together, these results indicate that miR-34a is a new regulator of SYT1, and both miR-34a and SYT1 play the important roles in the pathogenesis of colon cancer.
Collapse
Affiliation(s)
- Haichao Lu
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| | - Liang Hao
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| | - Hongfan Yang
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jianshe Chen
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jinxin Liu
- Department of Gastrointestinal Surgery, The First People's Hospital of Nanning Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
56
|
Zhang L, Niu H, Ma J, Yuan BY, Chen YH, Zhuang Y, Chen GW, Zeng ZC, Xiang ZL. The molecular mechanism of LncRNA34a-mediated regulation of bone metastasis in hepatocellular carcinoma. Mol Cancer 2019; 18:120. [PMID: 31349837 PMCID: PMC6659280 DOI: 10.1186/s12943-019-1044-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 06/26/2019] [Indexed: 12/22/2022] Open
Abstract
Background Bone metastasis (BM) has long been recognized as a major threat to the quality of life of hepatocellular cancer (HCC) patients. While LncRNA34a (Lnc34a) has been shown to regulate colon cancer stem cell asymmetric division, its effect on HCC BM remains unknown. Methods In situ hybridization and quantitative real-time polymerase chain reaction (qRT-PCR) were used to detect the expression of Lnc34a in HCC tissues and cell lines. Ventricle injection model was constructed to explore the effect of Lnc34a on BM in vivo. The methylation of miR-34a promoter and histones deacetylation were examined by using bisulfate-sequencing PCR and chromatin immunoprecipitation assays. RNA pull down and RNA immunoprecipitation were performed to investigated the interaction between Lnc34a and epigenetic regulators. Dual-luciferase reporter assay was conducted to find miR-34a target. The involvement of TGF-β pathway in the BM from HCC was determined by qRT-PCR, western, and elisa assays. Results We found that Lnc34a was significantly overexpressed in HCC tissues and associated with BM. Both in vitro and in vivo experiments indicate that the restoration or knockdown of Lnc34a expression in HCC cells had a marked effect on cellular migration, invasion, and metastasis. Mechanistic analyses suggested that Lnc34a epigenetically suppresses miR-34a expression through recruiting DNMT3a via PHB2 to methylate miR-34a promoter and HDAC1 to promote histones deacetylation. On the other hand, miR-34a targets Smad4 via the TGF-β pathway, followed by altering the transcription of the downstream genes (i.e., CTGF and IL-11) that are associated with BM. Conclusions Our study is the first to document the pro-bone metastatic role of Lnc34a in BM of HCC and reveal a novel mechanism for the activation of the TGF-β signaling pathway in HCC BM, providing evidence of a potential therapeutic strategy in HCC BM. Electronic supplementary material The online version of this article (10.1186/s12943-019-1044-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Hao Niu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Jie Ma
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Bao-Ying Yuan
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Yu-Han Chen
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Yuan Zhuang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Gen-Wen Chen
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.
| | - Zuo-Lin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
57
|
Huang S, Zhan Z, Li L, Guo H, Yao Y, Feng M, Deng J, Xiong J. LINC00958-MYC positive feedback loop modulates resistance of head and neck squamous cell carcinoma cells to chemo- and radiotherapy in vitro. Onco Targets Ther 2019; 12:5989-6000. [PMID: 31413594 PMCID: PMC6661987 DOI: 10.2147/ott.s208318] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/06/2019] [Indexed: 12/13/2022] Open
Abstract
Background Aberrant long non-coding RNA (lncRNA) expression contributes cancer development and resistance to therapy. This study first assessed expression of lncRNA LINC00958 in a variety of human cancers using GEPIA database data and then associated it with prognosis of head and neck squamous cell carcinoma (HNSCC) and investigated LINC00958 interaction with c-Myc and the c-Myc-related gene interplay in HNSCC cells. Materials and methods A cohort of 48 HNSCC vs normal tissues was collected for qRT-PCR analysis of LINC00958 and c-Myc expression and statistical analyses. HNSCC cell lines were subjected to transfection with LINC00958 and c-Myc siRNAs or cDNA and their negative control siRNA or empty vector for qRT-PCR, Western blot, cell viability, colony formation, luciferase reporter, chromatin immunoprecipitation, and RNA immunoprecipitation assays. Results The data showed that LINC00958 expression was upregulated in HNSCC tissues and cell lines, upregulation of which was associated with poor tumor differentiation, advanced tumor stage, and shorter overall survival of patients. In vitro, LINC00958 expression induced HNSCC cell viability and colony formation, whereas knockdown of LINC00958 expression enhanced HNSCC cell sensitivity to ionizing radiation and cisplatin treatment. Mechanistically, LINC00958 is a direct target of c-Myc and can enhance the transcriptional activity of c-Myc, thus to form a positive feedback gene network in HNSCC cells, and in turn to modulate HNSCC cell resistance to chemo- and radiotherapy. Conclusion This study demonstrated the LINC00958 interplay with c-Myc as a feedback loop facilitated HNSCC development and resistance to chemo- and radiotherapy. Targeting of such a network could be further evaluated as a novel therapeutic strategy for HNSCC patients.
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Zhengyu Zhan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Li Li
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Hui Guo
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Miao Feng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| |
Collapse
|
58
|
Tan W, Tang H, Jiang X, Ye F, Huang L, Shi D, Li L, Huang X, Li L, Xie X, Xie X. Metformin mediates induction of miR-708 to inhibit self-renewal and chemoresistance of breast cancer stem cells through targeting CD47. J Cell Mol Med 2019; 23:5994-6004. [PMID: 31273952 PMCID: PMC6714171 DOI: 10.1111/jcmm.14462] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/26/2019] [Accepted: 05/20/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer stem cells (BCSCs) have been considered responsible for cancer progression, recurrence, metastasis and drug resistance. However, the mechanisms by which cells acquire self-renewal and chemoresistance properties are remaining largely unclear. Herein, we evaluated the role of miR-708 and metformin in BCSCs, and found that the expression of miR-708 is significantly down-regulated in BCSCs and tumour tissues, and correlates with chemotherapy response and prognosis. Moreover, miR-708 markedly inhibits sphere formation, CD44+ /CD24- ratio, and tumour initiation and increases chemosensitivity of BCSCs. Mechanistically, miR-708 directly binds to cluster of differentiation 47 (CD47), and regulates tumour-associated macrophage-mediated phagocytosis. On the other hand, CD47 is essential for self-renewal, tumour initiation and chemoresistance of BCSCs, and correlates with the prognosis of breast cancer patients. In addition, the anti-type II diabetes drug metformin are found to be involved in the miR-708/CD47 signalling pathway. Therefore, our study demonstrated that miR-708 plays an important tumour suppressor role in BCSCs self-renewal and chemoresistance, and the miR-708/CD47 regulatory axis may represent a novel therapeutic mechanism of metformin in BCSCs.
Collapse
Affiliation(s)
- Weige Tan
- Department of Breast Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hailin Tang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinhua Jiang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Feng Ye
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lili Huang
- Guangzhou School of Medicine, Guangzhou Medical University, Guangzhou, China
| | - Dingbo Shi
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Laisheng Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojia Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Li Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaoming Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xinhua Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
59
|
Wang WT, Han C, Sun YM, Chen TQ, Chen YQ. Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol 2019; 12:55. [PMID: 31174564 PMCID: PMC6556047 DOI: 10.1186/s13045-019-0748-z] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/31/2019] [Indexed: 02/08/2023] Open
Abstract
Noncoding RNAs (ncRNAs) represent a large segment of the human transcriptome and have been shown to play important roles in cellular physiology and disease pathogenesis. Increasing evidence on the functional roles of ncRNAs in cancer progression emphasizes the potential of ncRNAs for cancer treatment. Here, we summarize the roles of ncRNAs in disease relapse and resistance to current standard chemotherapy and radiotherapy; the current research progress on ncRNAs for clinical and/or potential translational applications, including the identification of ncRNAs as therapeutic targets; therapeutic approaches for ncRNA targeting; and ncRNA delivery strategies in potential clinical translation. Several ongoing clinical trials of novel RNA-based therapeutics were also emphasized. Finally, we discussed the perspectives and obstacles to different target combinations, delivery strategies, and system designs for ncRNA application. The next approved nucleic acid drug to treat cancer patients may realistically be on the horizon.
Collapse
Affiliation(s)
- Wen-Tao Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China
| | - Cai Han
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yu-Meng Sun
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tian-Qi Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yue-Qin Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China. .,School of Life Science, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China.
| |
Collapse
|
60
|
Wu Y, Yang X, Chen Z, Tian L, Jiang G, Chen F, Li J, An P, Lu L, Luo N, Du J, Shan H, Liu H, Wang H. m 6A-induced lncRNA RP11 triggers the dissemination of colorectal cancer cells via upregulation of Zeb1. Mol Cancer 2019; 18:87. [PMID: 30979372 PMCID: PMC6461827 DOI: 10.1186/s12943-019-1014-2] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/26/2019] [Indexed: 02/08/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs) have emerged as critical players in cancer progression, but their functions in colorectal cancer (CRC) metastasis have not been systematically clarified. Methods lncRNA expression profiles in matched normal and CRC tissue were checked using microarray analysis. The biological roles of a novel lncRNA, namely RP11-138 J23.1 (RP11), in development of CRC were checked both in vitro and in vivo. Its association with clinical progression of CRC was further analyzed. Results RP11 was highly expressed in CRC tissues, and its expression increased with CRC stage in patients. RP11 positively regulated the migration, invasion and epithelial mesenchymal transition (EMT) of CRC cells in vitro and enhanced liver metastasis in vivo. Post-translational upregulation of Zeb1, an EMT-related transcription factor, was essential for RP11-induced cell dissemination. Mechanistically, the RP11/hnRNPA2B1/mRNA complex accelerated the mRNA degradation of two E3 ligases, Siah1 and Fbxo45, and subsequently prevented the proteasomal degradation of Zeb1. m6A methylation was involved in the upregulation of RP11 by increasing its nuclear accumulation. Clinical analysis showed that m6A can regulate the expression of RP11, further, RP11 regulated Siah1-Fbxo45/Zeb1 was involved in the development of CRC. Conclusions m6A-induced lncRNA RP11 can trigger the dissemination of CRC cells via post-translational upregulation of Zeb1. Considering the high and specific levels of RP11 in CRC tissues, our present study paves the way for further investigations of RP11 as a predictive biomarker or therapeutic target for CRC. Electronic supplementary material The online version of this article (10.1186/s12943-019-1014-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yingmin Wu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Xiangling Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Zhuojia Chen
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, Guangdong, China
| | - Lin Tian
- Department of Pharmacy, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Feng Chen
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Jiexin Li
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Panpan An
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Linlin Lu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Nan Luo
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Jun Du
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Hong Shan
- Key Laboratory of Biomedical Imaging of Guangdong Province, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Huanliang Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China. .,Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
| | - Hongsheng Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, and Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
61
|
Shin VY, Chen J, Cheuk IWY, Siu MT, Ho CW, Wang X, Jin H, Kwong A. Long non-coding RNA NEAT1 confers oncogenic role in triple-negative breast cancer through modulating chemoresistance and cancer stemness. Cell Death Dis 2019; 10:270. [PMID: 30894512 PMCID: PMC6426882 DOI: 10.1038/s41419-019-1513-5] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/20/2022]
Abstract
Triple-negative breast cancer (TNBC) is a malignant subtype of breast cancer with the absence of targeted therapy, resulting in poor prognosis in patients. Chemotherapy remains the mainstay of treatment for TNBC; however, development of drug resistance is the main obstacle for successful treatments. In recent years, long non-coding RNA (lncRNA) has been implicated in multiple biological functions in various diseases, particularly cancers. Accumulating evidence suggested that lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) expression is dysregulated in many human cancers and thus is a useful prognostic marker for cancer patients. Nevertheless, the mechanism of how NEAT1 confers drug resistance in TNBC is still largely unknown. We performed lncRNA profiling by the LncRNA Profiler qPCR Array Kit in normal control (NC) and breast cancers (BC) blood samples and further validated in a larger cohort of samples by qRT-PCR. Gene expression level and localization were investigated by qRT-PCR, western blotting, and immunofluorescence staining. Flow cytometric analysis was carried out to detect cancer stem cells. Functional studies were performed both in vitro and in vivo xenograft model. Among 90 lncRNAs, NEAT1 was highly expressed in the blood samples of breast cancer patients than in NC. In particular, the expression of NEAT1 was higher in TNBC tissues than other subgroups. Functional studies revealed that NEAT1 conferred oncogenic role by regulating apoptosis and cell cycle progression in TNBC cells. We identified that knockdown of NEAT1 sensitized cells to chemotherapy, indicating the involvement in chemoresistance. Importantly, shNEAT1 reduced stem cell populations such as CD44+/CD24−, ALDH+, and SOX2+, implicating that NEAT1 was closely related to cancer stemness in TNBC. Our data highlighted the roles of NEAT1 chemoresistance and cancer stemness, suggesting that it could be used as a new clinical therapeutic target for treating TNBC patients especially those with drug resistance.
Collapse
Affiliation(s)
| | - Jiawei Chen
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | | | - Man-Ting Siu
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chi-Wang Ho
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Hongchuan Jin
- Department of Medical Oncology, Key Laboratory of Biotherapy in Zhejiang, Sir Runrun Shaw Hospital, Medical School of Zhejiang University, Zhejiang, China
| | - Ava Kwong
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong. .,Department of Surgery, The Hong Kong Sanatorium and Hospital, Happy Valley, Hong Kong. .,The Hong Kong Hereditary Breast Cancer Family Registry, Shau Kei Wan, Hong Kong.
| |
Collapse
|
62
|
Liao M, Liao W, Xu N, Li B, Liu F, Zhang S, Wang Y, Wang S, Zhu Y, Chen D, Xie W, Jiang Y, Cao L, Yang BB, Zhang Y. LncRNA EPB41L4A-AS1 regulates glycolysis and glutaminolysis by mediating nucleolar translocation of HDAC2. EBioMedicine 2019; 41:200-213. [PMID: 30796006 PMCID: PMC6444057 DOI: 10.1016/j.ebiom.2019.01.035] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/02/2019] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND LncRNAs have been found to be involved in various aspects of biological processes. In this study, we aimed to uncover the molecular mechanisms of lncRNA EPB41L4A-AS1 in regulating glycolysis and glutaminolysis in cancer cells. METHODS The expression of EPB41L4A-AS1 in cancer patients was analyzed in TCGA and GEO datasets. The level of cellular metabolism was determined by extracellular flux analyzer. The relationship between p53 and EPB41L4A-AS1 was explored by qRT-PCR, luciferase assay and ChIP assay. The interactions between EPB41L4A-AS1 and HDAC2 or NPM1 were determined by RNA immunoprecipitation, RNA pull-down assay and RNA-FISH- immunofluorescence. FINDINGS EPB41L4A-AS1 was a p53-regulated gene. Low expression and deletion of lncRNA EPB41L4A-AS1 were found in a variety of human cancers and associated with poor prognosis of cancer patients. Knock down EPB41L4A-AS1 expression triggered Warburg effect, demonstrated as increased aerobic glycolysis and glutaminolysis. EPB41L4A-AS1 interacted and colocalized with HDAC2 and NPM1 in nucleolus. Silencing EPB41L4A-AS1 reduced the interaction between HDAC2 and NPM1, released HDAC2 from nucleolus and increased its distribution in nucleoplasm, enhanced HDAC2 occupation on VHL and VDAC1 promoter regions, and finally accelerated glycolysis and glutaminolysis. Depletion of EPB41L4A-AS1 increased the sensitivity of tumor to glutaminase inhibitor in tumor therapy. INTERPRETATION EPB41L4A-AS1 functions as a repressor of the Warburg effect and plays important roles in metabolic reprogramming of cancer.
Collapse
Affiliation(s)
- Meijian Liao
- School of Life Sciences, Tsinghua University, Beijing 100084, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | - Weijie Liao
- School of Life Sciences, Tsinghua University, Beijing 100084, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | - Naihan Xu
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, PR China
| | - Bing Li
- School of Life Sciences, Tsinghua University, Beijing 100084, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | - Fuhai Liu
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, PR China
| | - Shikuan Zhang
- School of Life Sciences, Tsinghua University, Beijing 100084, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | - Yanzhi Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | - Songmao Wang
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | - Yuanchang Zhu
- School of Life Sciences, Tsinghua University, Beijing 100084, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | - Deheng Chen
- Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518055, PR China
| | - Weidong Xie
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, PR China
| | - Yuyang Jiang
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, China Medical University, Shenyang 110013, PR China.
| | - Burton B Yang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.
| | - Yaou Zhang
- State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, PR China.
| |
Collapse
|
63
|
Wu XB, Feng X, Chang QM, Zhang CW, Wang ZF, Liu J, Hu ZQ, Liu JZ, Wu WD, Zhang ZP, Liu XQ. Cross-talk among AFAP1-AS1, ACVR1 and microRNA-384 regulates the stemness of pancreatic cancer cells and tumorigenicity in nude mice. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:107. [PMID: 30819221 PMCID: PMC6396461 DOI: 10.1186/s13046-019-1051-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
Abstract
Background Pancreatic cancer (PC) represents one of the most aggressive forms of cancer. The role of long non-coding RNAs (lncRNAs) has been highlighted in various malignancies including PC. The aim of the present study was to investigate the effects associated with actin filament-associated protein 1 antisense RNA 1 (AFAP1-AS1) on the progression of PC and the underlying mechanism. Methods Microarray-based gene expression profiling of PC was performed to identify PC-related lncRNAs, after which the expression of AFAP1-AS1 and cancer stem cell (CSC) markers in PC tissues and cells were determined accordingly. The potential microRNA-384 (miR-384) capable of binding to AFAP1-AS1, in addition to its ability to regulate activin receptor A type I (ACVR1) were analyzed. In order to investigate the effect of the AFAP1-AS1/miR-384/ACVR1 axis on self-renewal ability, tumorigenicity, invasion, migration and stemness of PC cells, shRNA-AFAP1-AS1, miR-384 mimic and inhibitor were cloned into cells. Results High expression of AFAP1-AS1 and ACVR1 with low expression of miR-384 were detected in PC tissues. ACVR1 was determined to be down-regulated when miR-384 was overexpressed, while the inhibition of AFAP1-AS1 decreased its ability to binding competitively to miR-384, resulting in the down-regulation of ACVR1 and enhancing miR-384 expression, ultimately inhibiting the progression of PC. The knockdown of AFAP1-AS1 or overexpression of miR-384 was confirmed to impair PC cell self-renewal ability, tumorigenicity, invasion, migration and stemness. Conclusions Taken together, AFAP1-AS1 functions as an endogenous RNA by competitively binding to miR-384 to regulate ACVR1, thus conferring inhibitory effects on PC cell stemness and tumorigenicity. Electronic supplementary material The online version of this article (10.1186/s13046-019-1051-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xu-Bo Wu
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, No. 170, Xinsong Road, Xinzhuang Town, Minhang District, Shanghai, 201199, People's Republic of China
| | - Xia Feng
- Department of Hepatopancreatobiliary Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158, Shangtang Road, Zhaohui District, Hangzhou, 310014, Zhejiang Province, People's Republic of China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, People's Republic of China.,Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, People's Republic of China
| | - Qi-Meng Chang
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, No. 170, Xinsong Road, Xinzhuang Town, Minhang District, Shanghai, 201199, People's Republic of China
| | - Cheng-Wu Zhang
- Department of Hepatopancreatobiliary Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158, Shangtang Road, Zhaohui District, Hangzhou, 310014, Zhejiang Province, People's Republic of China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, People's Republic of China.,Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, People's Republic of China
| | - Zhi-Fei Wang
- Department of Hepatopancreatobiliary Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158, Shangtang Road, Zhaohui District, Hangzhou, 310014, Zhejiang Province, People's Republic of China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, People's Republic of China.,Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, People's Republic of China
| | - Jie Liu
- Department of Hepatopancreatobiliary Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158, Shangtang Road, Zhaohui District, Hangzhou, 310014, Zhejiang Province, People's Republic of China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, People's Republic of China.,Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, People's Republic of China
| | - Zhi-Qiu Hu
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, No. 170, Xinsong Road, Xinzhuang Town, Minhang District, Shanghai, 201199, People's Republic of China
| | - Jia-Zhe Liu
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, No. 170, Xinsong Road, Xinzhuang Town, Minhang District, Shanghai, 201199, People's Republic of China
| | - Wei-Ding Wu
- Department of Hepatopancreatobiliary Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158, Shangtang Road, Zhaohui District, Hangzhou, 310014, Zhejiang Province, People's Republic of China. .,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, People's Republic of China. .,Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, People's Republic of China.
| | - Zi-Ping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, No. 170, Xinsong Road, Xinzhuang Town, Minhang District, Shanghai, 201199, People's Republic of China.
| | - Xi-Qiang Liu
- Department of Hepatopancreatobiliary Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158, Shangtang Road, Zhaohui District, Hangzhou, 310014, Zhejiang Province, People's Republic of China. .,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, People's Republic of China. .,Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, People's Republic of China.
| |
Collapse
|
64
|
Wang L, Wang E, Wang Y, Mines R, Xiang K, Sun Z, Zhou G, Chen KY, Rakhilin N, Chao S, Ye G, Wu Z, Yan H, Shen H, Everitt J, Bu P, Shen X. miR-34a is a microRNA safeguard for Citrobacter-induced inflammatory colon oncogenesis. eLife 2018; 7:e39479. [PMID: 30543324 PMCID: PMC6314783 DOI: 10.7554/elife.39479] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 12/06/2018] [Indexed: 12/22/2022] Open
Abstract
Inflammation often induces regeneration to repair the tissue damage. However, chronic inflammation can transform temporary hyperplasia into a fertile ground for tumorigenesis. Here, we demonstrate that the microRNA miR-34a acts as a central safeguard to protect the inflammatory stem cell niche and reparative regeneration. Although playing little role in regular homeostasis, miR-34a deficiency leads to colon tumorigenesis after Citrobacter rodentium infection. miR-34a targets both immune and epithelial cells to restrain inflammation-induced stem cell proliferation. miR-34a targets Interleukin six receptor (IL-6R) and Interleukin 23 receptor (IL-23R) to suppress T helper 17 (Th17) cell differentiation and expansion, targets chemokine CCL22 to hinder Th17 cell recruitment to the colon epithelium, and targets an orphan receptor Interleukin 17 receptor D (IL-17RD) to inhibit IL-17-induced stem cell proliferation. Our study highlights the importance of microRNAs in protecting the stem cell niche during inflammation despite their lack of function in regular tissue homeostasis.
Collapse
Affiliation(s)
- Lihua Wang
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
| | - Ergang Wang
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
| | - Yi Wang
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
- Affiliated Hospital of Nanjing University of TCMNanjingChina
| | - Robert Mines
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
| | - Kun Xiang
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
| | - Zhiguo Sun
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
| | - Gaiting Zhou
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
| | - Kai-Yuan Chen
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
| | - Nikolai Rakhilin
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- School of Electrical and Computer EngineeringCornell UniversityNew yorkUnited States
| | - Shanshan Chao
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Gaoqi Ye
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zhenzhen Wu
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Huiwen Yan
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Hong Shen
- Affiliated Hospital of Nanjing University of TCMNanjingChina
| | - Jeffrey Everitt
- Department of Pathology, Animal Pathology CoreDuke UniversityDurhamUnited States
| | - Pengcheng Bu
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiling Shen
- Center for Genomics and Computational BiologyDuke UniversityDurhamUnited States
- Department of Biomedical EngineeringDuke UniversityDurhamUnited States
- School of Electrical and Computer EngineeringCornell UniversityNew yorkUnited States
| |
Collapse
|
65
|
The emerging role of lncRNAs in the regulation of cancer stem cells. Cell Oncol (Dordr) 2018; 41:585-603. [PMID: 30218296 DOI: 10.1007/s13402-018-0406-4] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tumors contain a functional subpopulation of cells that exhibit stem cell properties. These cells, named cancer stem cells (CSCs), play significant roles in the initiation and progression of cancer. Long non-coding RNAs (lncRNAs) can act at the transcriptional, posttranscriptional and translational level. As such, they may be involved in various biological processes such as DNA damage repair, inflammation, metabolism, cell survival, cell signaling, cell growth and differentiation. Accumulating evidence indicates that lncRNAs are key regulators of the CSC subpopulation, thereby contributing to cancer progression. The aim of this review is to overview current knowledge about the functional role and the mechanisms of action of lncRNAs in the initiation, maintenance and regulation of CSCs derived from different neoplasms. These lncRNAs include CTCF7, ROR, DILC, HOTAIR, H19, HOTTIP, ATB, HIF2PUT, SOX2OT, MALAT-1, CUDR, Lnc34a, Linc00617, DYNC2H1-4, PVT1, SOX4 and ARSR Uc.283-plus. Furthermore, we will illustrate how lncRNAs may regulate asymmetric CSC division and contribute to self-renewal, drug resistance and EMT, thus affecting the metastasis and recurrence of different cancers. In addition, we will highlight the implications of targeting lncRNAs to improve the efficacy of conventional drug therapies and to hamper CSC survival and proliferation. CONCLUSIONS lncRNAs are valuable tools in the search for new targets to selectively eliminate CSCs and improve clinical outcomes. LncRNAs may serve as excellent therapeutic targets because they are stable, easily detectable and expressed in tissue-specific contexts.
Collapse
|
66
|
miR-34a Regulates Multidrug Resistance via Positively Modulating OAZ2 Signaling in Colon Cancer Cells. J Immunol Res 2018; 2018:7498514. [PMID: 30175154 PMCID: PMC6098920 DOI: 10.1155/2018/7498514] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 06/06/2018] [Accepted: 06/12/2018] [Indexed: 02/06/2023] Open
Abstract
Although aberrant expression of miR-34a, an essential tumor suppressor miRNA, has been frequently observed in colon cancer (CCa), whether miR-34a can regulate CCa progression by modulating other facets of this malignancy (such as multidrug resistance, MDR) remains unknown. Here, we report for the first time that miR-34a expression was significantly downregulated in clinical CCa samples from oxaliplatin-resistant patients and in experimentally established multidrug-resistant CCa cells. By using histoculture drug response assay, we further confirmed that clinical CCa samples with lower miR-34a expression appeared to be more resistant to chemotherapy. Functionally, ectopic expression of exogenous miR-34a resensitized multidrug-resistant HCT-8/OR cells to oxaliplatin treatment, whereas miR-34a inhibition augmented the oxaliplatin resistance in chemosensitive HCT-8 cells. Mechanistically, miR-34a positively regulated the mRNA stability of the ornithine decarboxylase antizyme 2 (OAZ2) by directly targeting its three prime untranslated region (3′UTR). Consequently, suppression of the expression of miR-34a/OAZ2 signaling by chemotherapeutic agents significantly enhanced the activation of MDR-associated ATP-binding cassette (ABC) transporters and antiapoptosis pathways, thus leading to MDR development in CCa cells. Collectively, our combined analysis reveals a critical role of miR-34a/OAZ2 cascade in conferring a proper cellular response to CCa chemotherapy.
Collapse
|
67
|
Wang J, Zhou T, Sun Z, Ye T, Zhou S, Li J, Liu Y, Kong L, Tang J, Liu D, Xing H. Zeb1 Regulates the Symmetric Division of Mouse Lewis Lung Carcinoma Stem Cells through Numb mediated by miR-31. Int J Biol Sci 2018; 14:1399-1410. [PMID: 30262992 PMCID: PMC6158737 DOI: 10.7150/ijbs.27446] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/15/2018] [Indexed: 12/13/2022] Open
Abstract
Symmetric cell division (SD) and asymmetric cell division (ASD) were the unique characteristics of stem cells and the mechanisms underlying stem cell renewal. While recent studies have identified the presence of SD and ASD in lung cancer stem cells (CSCs), the mechanisms regulating SD and ASD in cancer state have not been elucidated, mostly due to the lack of stable cellular models of SD and ASD in CSC research. In this study, the interaction between Zeb1, an Epithelial-Mesenchymal Transition (EMT) factor shown to regulate CSCs self-renew, and Numb, which regulates SD and ASD in the normal neural stem cell was investigated using the stable mouse Lewis lung adenocarcinoma SD (LLC-SD) and ASD (LLC-ASD) lines established from our previous study. The most significant finding derived from this line of research is that we have identified and molecularly ordered the axis of Zeb1-miR-31-Numb that regulates the SD, a mechanism of CSC self-renewal that has not been previously described. More specifically, the expression of Zeb1 and Numb were both significantly higher in LLC-SD than LLC-ASD cells. Silencing of Zeb1 or Numb expression lead to decreased ratio of SD and weakened single-cell cloning formation, tumor growth and tumor metastasis, respectively. The rescure experiments have molecularly ordered the regulation of Numb by Zeb1, indirectly mediated by miR-31. Moreover, we also provided preliminary evidence supporting the clinical relevance of our finding. In summary, our study provides a new insight for the self-renew of lung CSCs in which SD is regulated by the axis of Zeb1-miR-31-Numb.
Collapse
Affiliation(s)
- Jianyu Wang
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Tiejun Zhou
- Department of Pathology, The affiliated Hospital of Southwest medical university
| | - Zhiwei Sun
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Ting Ye
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Shixia Zhou
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jingyuan Li
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yongli Liu
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Liangsheng Kong
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Junlin Tang
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Doudou Liu
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - H.Rosie Xing
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing, China
| |
Collapse
|
68
|
Huang X, Xie X, Liu P, Yang L, Chen B, Song C, Tang H, Xie X. Adam12 and lnc015192 act as ceRNAs in breast cancer by regulating miR-34a. Oncogene 2018; 37:6316-6326. [PMID: 30042416 DOI: 10.1038/s41388-018-0410-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/12/2018] [Accepted: 06/26/2018] [Indexed: 12/13/2022]
Abstract
Long non-coding RNAs (lncRNAs) are reported to play vital roles in the progress of multiple cancers. However, the functions of lncRNAs in breast cancer remain to be discovered. We performed microarrays to identify the differentially expressed mRNAs and lncRNAs in breast tissues with or without miR-34a knockout. To explore the functions of the differentially expressed mRNA and lncRNA in breast cancer, we conducted a series of experiments. We found that Adam12 and lnc015192 were significantly upregulated in miR-34a knockout breast tissues. Knockdown of Adam12 and lnc015192 inhibited breast cancer cell migration, invasion, and epithelial-mesenchymal transition (EMT). Further experiments revealed that lnc015192 regulated Adam12 expression by functioning as a competing endogenous RNA (ceRNA) for miR-34a. In summary, our study demonstrate that Adam12 and lnc015192 promote breast cancer metastasis partly by sponging miR-34a through the ceRNA mechanism.
Collapse
Affiliation(s)
- Xiaojia Huang
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Xinhua Xie
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Peng Liu
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Lu Yang
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Bo Chen
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Cailu Song
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China
| | - Hailin Tang
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China.
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, China.
| |
Collapse
|
69
|
Deciphering the Far-Reaching Functions of Non-coding RNA in Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2018. [DOI: 10.1007/s11888-018-0408-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
70
|
Serviss JT, Andrews N, Van den Eynden J, Richter FC, Houtman M, Vesterlund M, Schwarzmueller L, Johnsson P, Larsson E, Grandér D, Pokrovskaja Tamm K. An antisense RNA capable of modulating the expression of the tumor suppressor microRNA-34a. Cell Death Dis 2018; 9:736. [PMID: 29970884 PMCID: PMC6030072 DOI: 10.1038/s41419-018-0777-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 01/25/2023]
Abstract
The microRNA-34a is a well-studied tumor suppressor microRNA (miRNA) and a direct downstream target of TP53 with roles in several pathways associated with oncogenesis, such as proliferation, cellular growth, and differentiation. Due to its broad tumor suppressive activity, it is not surprising that miR34a expression is altered in a wide variety of solid tumors and hematological malignancies. However, the mechanisms by which miR34a is regulated in these cancers is largely unknown. In this study, we find that a long noncoding RNA transcribed antisense to the miR34a host gene, is critical for miR34a expression and mediation of its cellular functions in multiple types of human cancer. We name this long noncoding RNA lncTAM34a, and characterize its ability to facilitate miR34a expression under different types of cellular stress in both TP53-deficient and wild-type settings.
Collapse
Affiliation(s)
- Jason T Serviss
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE-17177, Sweden.
| | - Nathanael Andrews
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE-17177, Sweden
| | - Jimmy Van den Eynden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Felix Clemens Richter
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE-17177, Sweden.,Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK
| | - Miranda Houtman
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE-17177, Sweden.,Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Vesterlund
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE-17177, Sweden
| | - Laura Schwarzmueller
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE-17177, Sweden.,Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center, Amsterdam, The Netherlands
| | - Per Johnsson
- Ludwig Institute for Cancer Research, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Erik Larsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Dan Grandér
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE-17177, Sweden
| | - Katja Pokrovskaja Tamm
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE-17177, Sweden
| |
Collapse
|
71
|
Zarkou V, Galaras A, Giakountis A, Hatzis P. Crosstalk mechanisms between the WNT signaling pathway and long non-coding RNAs. Noncoding RNA Res 2018; 3:42-53. [PMID: 30159439 PMCID: PMC6096407 DOI: 10.1016/j.ncrna.2018.04.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/05/2018] [Accepted: 04/05/2018] [Indexed: 12/15/2022] Open
Abstract
The WNT/β-catenin signaling pathway controls a plethora of biological processes throughout animal development and adult life. Because of its fundamental role during animal lifespan, the WNT pathway is subject to strict positive and negative multi-layered regulation, while its aberrant activity causes a wide range of pathologies, including cancer. At present, despite the inroads into the molecules involved in WNT-mediated transcriptional responses, the fine-tuning of WNT pathway activity and the totality of its target genes have not been fully elucidated. Over the past few years, long non-coding RNAs (lncRNAs), RNA transcripts longer that 200nt that do not code for proteins, have emerged as significant transcriptional regulators. Recent studies show that lncRNAs can modulate WNT pathway outcome by affecting gene expression through diversified mechanisms, from the transcriptional to post-translational level. In this review, we selectively discuss those lncRNA-mediated mechanisms we believe the most important to WNT pathway modulation.
Collapse
Affiliation(s)
- Vasiliki Zarkou
- Biomedical Sciences Research Center ‘Alexander Fleming’, 16672 Vari, Greece
- School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Alexandros Galaras
- Biomedical Sciences Research Center ‘Alexander Fleming’, 16672 Vari, Greece
- Department of Medicine, National and Kapodistrian University of Athens, 11527 Goudi, Greece
| | - Antonis Giakountis
- Biomedical Sciences Research Center ‘Alexander Fleming’, 16672 Vari, Greece
- Department of Biochemistry and Biotechnology, University of Thessaly, 41500 Larissa, Greece
| | - Pantelis Hatzis
- Biomedical Sciences Research Center ‘Alexander Fleming’, 16672 Vari, Greece
- Corresponding author.
| |
Collapse
|
72
|
Zhou W, Wang L, Miao Y, Xing R. Novel long noncoding RNA GACAT3 promotes colorectal cancer cell proliferation, invasion, and migration through miR-149. Onco Targets Ther 2018; 11:1543-1552. [PMID: 29593420 PMCID: PMC5865577 DOI: 10.2147/ott.s144103] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim To explore the expression and clinical significance of long noncoding RNA (lncRNA) gastric cancer-associated transcript 3 (GACAT3) in human colorectal cancer (CRC). Methods Expression of GACAT3 in CRC tissues and cell lines was measured using quantitative real-time PCR. CCK-8 and colony formation assays were used to assess the effect of GACAT3 on CRC cell line proliferation. Transwell invasion and migration assays were performed to detect the effect of GACAT3 on CRC cell line invasion and migration. Bioinformatics prediction, luciferase reporter assay, and pull-down assay were used to determine if miR-149 was a target of GACAT3. In addition, we also conducted colony formation assays and invasion assays to verify that GACAT3 promotes tumor progression through miR-149. Finally, in vivo tumorigenesis studies were used to demonstrate subcutaneous tumor growth. Results In the present study, we found that GACAT3 was highly expressed in CRC tissues and cell lines. Si-GACAT3 significantly decreased cell proliferation, motility, and invasiveness both in vitro and in vivo. We confirmed that downregulated GACAT3 significantly increased the expression of miR-149, and miR-149 binds to GACAT3 in a sequence-specific manner using luciferase reporter assays and pull-down assay. Further functional experiments indicated that GACAT3 could directly upregulate SP1 and STAT3 expressions by functioning as a competing endogenous RNA for miR-149, and consequentially, promoting CRC cell proliferation and invasion in vitro. Conclusion This study demonstrated that GACAT3 promotes tumor progression through competitive binding to miR-149 and suggests a promising new strategy for anti-CRC therapy.
Collapse
Affiliation(s)
- Weiwei Zhou
- Department of Pathology, Central Hospital of Cangzhou, Hebei
| | - Lei Wang
- Department of Pharmacy, Cangzhou Medical College, Hebei, China
| | - Yu Miao
- Department of Pathology, Central Hospital of Cangzhou, Hebei
| | - Rongge Xing
- Department of Pathology, Central Hospital of Cangzhou, Hebei
| |
Collapse
|
73
|
Fu Y, Li C, Luo Y, Li L, Liu J, Gui R. Silencing of Long Non-coding RNA MIAT Sensitizes Lung Cancer Cells to Gefitinib by Epigenetically Regulating miR-34a. Front Pharmacol 2018; 9:82. [PMID: 29487526 PMCID: PMC5816758 DOI: 10.3389/fphar.2018.00082] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/25/2018] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNA (lncRNA) myocardial infarction associated transcript (MIAT) was recently identified as oncogene in several cancers. However, the role of MIAT on acquired resistance in lung cancer and the underlying mechanisms remain unclear. Here, we showed that the expression of MIAT in lung cancer tissues was upregulated compared with adjacent tissues. LncRNA MIAT expression was associated with tumor size, lymph node metastasis, distant metastasis and TNM stage. Univariate analysis and multivariate analysis revealed that the lncRNA MIAT to be an independent factor for predicating the prognosis of lung cancer patients. Low lncRNA MIAT have longer overall survival time and progression-free survival time than patients with high lncRNA MIAT expression. Moreover, the knockdown of MIAT significantly sensitized PC9 and gefitinib-resistant PC9 cells to gefitinib in vitro and in vivo, and increased the expression of miR-34a and inactivated PI3K/Akt signaling. MIAT interacted with miR-34a and epigenetically controlled the miR-34a expression by hyper-methylating its promotor. Taken together, our findings demonstrated that knockdown of MIAT by siRNA enhances lung cancer cells to gefitinib through the PI3K/Akt signaling pathway by epigenetically regulating miR-34a. Thus, MIAT may be a useful prognostic marker and therapeutic target for lung cancer patients.
Collapse
Affiliation(s)
- Yunfeng Fu
- The Third Xiangya Hospital of Central South University, Changsha, China
| | - Chengyuan Li
- The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yanwei Luo
- The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lian Li
- The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jing Liu
- The Third Xiangya Hospital of Central South University, Changsha, China
| | - Rong Gui
- The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
74
|
Rossi M, Jahanzaib Anwar M, Usman A, Keshavarzian A, Bishehsari F. Colorectal Cancer and Alcohol Consumption-Populations to Molecules. Cancers (Basel) 2018; 10:E38. [PMID: 29385712 PMCID: PMC5836070 DOI: 10.3390/cancers10020038] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is a major cause of morbidity and mortality, being the third most common cancer diagnosed in both men and women in the world. Several environmental and habitual factors have been associated with the CRC risk. Alcohol intake, a common and rising habit of modern society, is one of the major risk factors for development of CRC. Here, we will summarize the evidence linking alcohol with colon carcinogenesis and possible underlying mechanisms. Some epidemiologic studies suggest that even moderate drinking increases the CRC risk. Metabolism of alcohol involves ethanol conversion to its metabolites that could exert carcinogenic effects in the colon. Production of ethanol metabolites can be affected by the colon microbiota, another recently recognized mediating factor to colon carcinogenesis. The generation of acetaldehyde and alcohol's other metabolites leads to activation of cancer promoting cascades, such as DNA-adduct formation, oxidative stress and lipid peroxidation, epigenetic alterations, epithelial barrier dysfunction, and immune modulatory effects. Not only does alcohol induce its toxic effect through carcinogenic metabolites, but alcoholics themselves are predisposed to a poor diet, low in folate and fiber, and circadian disruption, which could further augment alcohol-induced colon carcinogenesis.
Collapse
Affiliation(s)
- Marco Rossi
- Division of Digestive Diseases, Hepatology, and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Muhammad Jahanzaib Anwar
- Division of Digestive Diseases, Hepatology, and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Ahmad Usman
- Division of Digestive Diseases, Hepatology, and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Ali Keshavarzian
- Division of Digestive Diseases, Hepatology, and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Faraz Bishehsari
- Division of Digestive Diseases, Hepatology, and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
75
|
Loss of p53-inducible long non-coding RNA LINC01021 increases chemosensitivity. Oncotarget 2017; 8:102783-102800. [PMID: 29262524 PMCID: PMC5732690 DOI: 10.18632/oncotarget.22245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 12/19/2022] Open
Abstract
We have previously identified the long non-coding RNA LINC01021 as a direct p53 target (Hünten et al. Mol Cell Proteomics. 2015; 14:2609-2629). Here, we show that LINC01021 is up-regulated in colorectal cancer (CRC) cell lines upon various p53-activating treatments. The LINC01021 promoter and the p53 binding site lie within a MER61C LTR, which originated from insertion of endogenous retrovirus 1 (ERV1) sequences. Deletion of this MER61C element by a CRISPR/Cas9 approach, as well as siRNA-mediated knockdown of LINC01021 RNA significantly enhanced the sensitivity of the CRC cell line HCT116 towards the chemotherapeutic drugs doxorubicin and 5-FU, suggesting that LINC01021 is an integral part of the p53-mediated response to DNA damage. Inactivation of LINC01021 and also its ectopic expression did not affect p53 protein expression and transcriptional activity, implying that LINC01021 does not feedback to p53. Furthermore, in CRC patient samples LINC01021 expression positively correlated with a wild-type p53-associated gene expression signature. LINC01021 expression was increased in primary colorectal tumors and displayed a bimodal distribution that was particularly pronounced in the mesenchymal CMS4 consensus molecular subtype of CRCs. CMS4 tumors with low LINC01021 expression were associated with poor patient survival. Our results suggest that the genomic redistribution of ERV1-derived p53 response elements and generation of novel p53-inducible lncRNA-encoding genes was selected for during primate evolution as integral part of the cellular response to various forms of genotoxic stress.
Collapse
|
76
|
Luo J, Qu J, Wu DK, Lu ZL, Sun YS, Qu Q. Long non-coding RNAs: a rising biotarget in colorectal cancer. Oncotarget 2017; 8:22187-22202. [PMID: 28108736 PMCID: PMC5400657 DOI: 10.18632/oncotarget.14728] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/09/2017] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a common gastrointestinal cancer, with a high incidence and high mortality. Long non-coding RNAs (lncRNAs) are involved in the development, invasion and metastasis, early diagnosis, prognosis, the chemoresistance and radioresistance of CRC through interference with mRNA activity, directly combining with proteins to regulate their activity or alter their localization, influencing downstream gene expression by inhibiting RNA polymerase and regulating gene expression as competing endogenous RNAs. Recent progress in next generation sequencing and transcriptome analysis has revealed that tissue and cancer-type specific lncRNAs could be useful prognostic markers. Here, the CRC-associated lncRNAs from recent studies until October 2016 are reviewed and multiple studies that have confirmed CRC-associated lncRNAs are summarized. This review may be helpful in understanding the overall relationships between the lncRNAs involved in CRC.
Collapse
Affiliation(s)
- Jian Luo
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Institute of Clinical Pharmacy, Central South University, Changsha, P. R. China
| | - Dong-Kai Wu
- Department of Cardiothoracic Surgery, Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Zhi-Li Lu
- Department of Pathology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, P. R. China
| | - Yue-Sheng Sun
- Department of General Surgery, The Third Clinical College of Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, P. R. China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, P. R. China
| |
Collapse
|
77
|
Han M, Dai D, Yousafzai NA, Wang F, Wang H, Zhou Q, Lu H, Xu W, Feng L, Jin H, Wang X. CXXC4 activates apoptosis through up-regulating GDF15 in gastric cancer. Oncotarget 2017; 8:103557-103567. [PMID: 29262584 PMCID: PMC5732750 DOI: 10.18632/oncotarget.21581] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/05/2017] [Indexed: 12/19/2022] Open
Abstract
Worldwide, gastric cancer is one of the most fatal cancers. Epigenetic alterations in gastric cancer play important roles in silencing of tumor suppressor genes. We previously found that CXXC finger protein 4 (CXXC4) was a novel tumor suppressor in gastric cancer. In this report, we demonstrated that CXXC4 inhibited growth of gastric cancer cells as a pro-apoptotic factor. This inhibition could be reversed by the pan-caspase inhibitor called Z-VAD-FMK. However, CXXC4 with mutations in its DNA binding domain failed to induce apoptosis. Growth differentiation factor 15 (GDF15) was identified as one of potential targets responsible for CXXC4-induced apoptosis. CXXC4 activated GDF15 transcription through enhancing the interaction of transcription factor Sp1 with GDF15 promoter. In summary, the nuclear protein CXXC4 activated apoptosis in gastric cancer through up-regulating its novel potential downstream target GDF15. GDF15 might be a promising target for clinical treatment of gastric cancer with CXXC4 deficiency.
Collapse
Affiliation(s)
- Mengjiao Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Dongjun Dai
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Neelum Aziz Yousafzai
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Faliang Wang
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Hanying Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Qiying Zhou
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Haiqi Lu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Wenxia Xu
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Lifeng Feng
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Hongchuan Jin
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| |
Collapse
|
78
|
lncRNA DANCR promotes tumor progression and cancer stemness features in osteosarcoma by upregulating AXL via miR-33a-5p inhibition. Cancer Lett 2017. [DOI: 10.1016/j.canlet.2017.06.009] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
79
|
Shen P, Pichler M, Chen M, Calin GA, Ling H. To Wnt or Lose: The Missing Non-Coding Linc in Colorectal Cancer. Int J Mol Sci 2017; 18:ijms18092003. [PMID: 28930145 PMCID: PMC5618652 DOI: 10.3390/ijms18092003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/09/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequent cancer and one of the leading causes for cancer-related mortality. Aberrant activation of the Wnt signaling is an essential initiating factor in colon carcinogenesis, and a driving force of CRC progression. Recently, long non-coding RNAs (lncRNAs) have emerged as significant players in CRC pathogenesis through diversified mechanisms. Although both Wnt signaling and lncRNAs represent interesting research areas for CRC, an effort of directly connecting these two areas is lacking. To fill in the knowledge gap, we focus on the reported findings of lncRNAs that regulate Wnt signaling or essential Wnt signaling targets. These include several newly discovered lncRNAs originated from the amplified cancer-associated chromosome 8q24 region that surrounds the essential Wnt target MYC gene, lncRNAs reported to be involved in CRC stem cells, and several individual lncRNAs connected to Wnt signaling through other mechanisms. This review will provide essential information that assists in understanding the missing link of lncRNAs to the classical Wnt signaling in CRC.
Collapse
Affiliation(s)
- Peng Shen
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
- Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - Martin Pichler
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
- Research Unit of Non-Coding RNA and Genome Editing in Cancer, Division of Oncology, Medical University of Graz, Graz 8010, Austria.
| | - Meng Chen
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
- The Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Hui Ling
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
80
|
Dashzeveg NK, Taftaf R, Ramos EK, Torre-Healy L, Chumakova A, Silver DJ, Alban TJ, Sinyuk M, Thiagarajan PS, Jarrar AM, Turaga SM, Saygin C, Mulkearns-Hubert E, Hitomi M, Rich JN, Gerson SL, Lathia JD, Liu H. New Advances and Challenges of Targeting Cancer Stem Cells. Cancer Res 2017; 77:5222-5227. [PMID: 28928129 DOI: 10.1158/0008-5472.can-17-0054] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/25/2017] [Accepted: 07/18/2017] [Indexed: 02/04/2023]
Abstract
The second International Cancer Stem Cell Conference in Cleveland, Ohio, on September 20-23, 2016, convened 330 attendees from academic, industrial, and clinical organizations. It featured a debate on the concepts and challenges of the cancer stem cells (CSC) as well as CSC-centered scientific sessions on clinical trials, genetics and epigenetics, tumor microenvironment, immune suppression, metastasis, therapeutic resistance, and emerging novel concepts. The conference hosted 35 renowned speakers, 100 posters, 20 short talks, and a preconference workshop. The reported advances of CSC research and therapies fostered new collaborations across national and international borders, and inspired the next generation's young scientists. Cancer Res; 77(19); 5222-7. ©2017 AACR.
Collapse
Affiliation(s)
- Nurmaa K Dashzeveg
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Rokana Taftaf
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Erika K Ramos
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Luke Torre-Healy
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Anastasia Chumakova
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Daniel J Silver
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Tyler J Alban
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Maksim Sinyuk
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Praveena S Thiagarajan
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Awad M Jarrar
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Soumya M Turaga
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Caner Saygin
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Erin Mulkearns-Hubert
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Masahiro Hitomi
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Jeremy N Rich
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio.,The Case Comprehensive Cancer Center, Cleveland, Ohio.,The National Center for Regenerative Medicine, Cleveland, Ohio
| | - Stanton L Gerson
- The Case Comprehensive Cancer Center, Cleveland, Ohio.,The National Center for Regenerative Medicine, Cleveland, Ohio.,The University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio.,The Case Comprehensive Cancer Center, Cleveland, Ohio.,The National Center for Regenerative Medicine, Cleveland, Ohio
| | - Huiping Liu
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois. .,The Case Comprehensive Cancer Center, Cleveland, Ohio.,The National Center for Regenerative Medicine, Cleveland, Ohio.,Department of Medicine (Hematology and Oncology Division) and Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Deparmtent of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
81
|
Chi HC, Tsai CY, Tsai MM, Yeh CT, Lin KH. Roles of Long Noncoding RNAs in Recurrence and Metastasis of Radiotherapy-Resistant Cancer Stem Cells. Int J Mol Sci 2017; 18:1903. [PMID: 28872613 PMCID: PMC5618552 DOI: 10.3390/ijms18091903] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy is a well-established therapeutic regimen applied to treat at least half of all cancer patients worldwide. Radioresistance of cancers or failure to treat certain tumor types with radiation is associated with enhanced local invasion, metastasis and poor prognosis. Elucidation of the biological characteristics underlying radioresistance is therefore critical to ensure the development of effective strategies to resolve this issue, which remains an urgent medical problem. Cancer stem cells (CSCs) comprise a small population of tumor cells that constitute the origin of most cancer cell types. CSCs are virtually resistant to radiotherapy, and consequently contribute to recurrence and disease progression. Metastasis is an increasing problem in resistance to cancer radiotherapy and closely associated with the morbidity and mortality rates of several cancer types. Accumulating evidence has demonstrated that radiation induces epithelial-mesenchymal transition (EMT) accompanied by increased cancer recurrence, metastasis and CSC generation. CSCs are believed to serve as the basis of metastasis. Previous studies indicate that CSCs contribute to the generation of metastasis, either in a direct or indirect manner. Moreover, the heterogeneity of CSCs may be responsible for organ specificity and considerable complexity of metastases. Long noncoding RNAs (lncRNAs) are a class of noncoding molecules over 200 nucleotides in length involved in the initiation and progression of several cancer types. Recently, lncRNAs have attracted considerable attention as novel critical regulators of cancer progression and metastasis. In the current review, we have discussed lncRNA-mediated regulation of CSCs following radiotherapy, their association with tumor metastasis and significance in radioresistance of cancer.
Collapse
Affiliation(s)
- Hsiang-Cheng Chi
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan.
| | - Chung-Ying Tsai
- Kidney Research Center and Department of Nephrology, Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan.
| | - Ming-Ming Tsai
- Department of Nursing, Chang-Gung University of Science and Technology, Taoyuan 333, Taiwan.
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi 613, Taiwan.
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan.
| | - Kwang-Huei Lin
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan.
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan.
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan.
| |
Collapse
|
82
|
Yang Y, Zhao L, Lei L, Lau WB, Lau B, Yang Q, Le X, Yang H, Wang C, Luo Z, Xuan Y, Chen Y, Deng X, Xu L, Feng M, Yi T, Zhao X, Wei Y, Zhou S. LncRNAs: the bridge linking RNA and colorectal cancer. Oncotarget 2017; 8:12517-12532. [PMID: 27888635 PMCID: PMC5355361 DOI: 10.18632/oncotarget.13573] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 11/12/2016] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are transcribed by genomic regions (exceeding 200 nucleotides in length) that do not encode proteins. While the exquisite regulation of lncRNA transcription can provide signals of malignant transformation, lncRNAs control pleiotropic cancer phenotypes through interactions with other cellular molecules including DNA, protein, and RNA. Recent studies have demonstrated that dysregulation of lncRNAs is influential in proliferation, angiogenesis, metastasis, invasion, apoptosis, stemness, and genome instability in colorectal cancer (CRC), with consequent clinical implications. In this review, we explicate the roles of different lncRNAs in CRC, and the potential implications for their clinical application.
Collapse
Affiliation(s)
- Yanfei Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Linjie Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Lingzi Lei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University Hospital, U.S.A
| | - Bonnie Lau
- Department of Emergency Medicine, Kaiser Permanente Santa Clara Medical Center, Affiliate of Stanford University, U.S.A
| | - Qilian Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Xiaobing Le
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Huiliang Yang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Chenlu Wang
- College of Life Sciences, Sichuan University, Chengdu, P. R. China
| | - Zhongyue Luo
- College of Life Sciences, Sichuan University, Chengdu, P. R. China
| | - Yu Xuan
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Yi Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiangbing Deng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Lian Xu
- Department of Pathology, West China Second Hospital, Sichuan University, Chengdu, P. R. China
| | - Min Feng
- Department of Pathology, West China Second Hospital, Sichuan University, Chengdu, P. R. China
| | - Tao Yi
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Xia Zhao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Yuquan Wei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second Hospital and State Key Laboratory of Biotherapy/Collaborative Innovation Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
83
|
The crosstalk between long non-coding RNAs and PI3K in cancer. Med Oncol 2017; 34:39. [PMID: 28176240 DOI: 10.1007/s12032-017-0897-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 01/27/2017] [Indexed: 12/21/2022]
Abstract
Long non-coding RNAs (lncRNAs) are able to positively or negatively regulate other genes expression in cis or in trans. Their effect can be achieved through RNA-protein, RNA-DNA, or RNA-RNA interactions. They can recruit transcription factors and act as scaffolds or guides for chromatin-modifying enzymes. PI3K kinases transform external stimuli to intracellular signals regulating cell growth, differentiation, proliferation, survival, intracellular trafficking, cytoskeletal changes, cell migration and motility, and metabolism. PI3K is activated in cancer and affects several aspects of oncogenesis. LncRNAs and PI3K have been shown to be interconnected in several different cancer subtypes enhancing aberrant cell proliferation, epithelial-to-mesenchymal transition, migration and invasion, and also cancer cell metabolism. In this review, we have assembled recent data describing the interaction between lncRNAs and PI3K and the results of such interaction.
Collapse
|
84
|
Manna A, Banerjee S, Khan P, Bhattacharya A, Das T. Contribution of nuclear events in generation and maintenance of cancer stem cells: revisiting chemo-resistance. THE NUCLEUS 2017. [DOI: 10.1007/s13237-017-0193-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
85
|
Ezzatizadeh V. Cancer Stem Cell: From Conjecture to Reality. CANCER GENETICS AND PSYCHOTHERAPY 2017:757-787. [DOI: 10.1007/978-3-319-64550-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
86
|
Mining, Validation, and Clinical Significance of Colorectal Cancer (CRC)-Associated lncRNAs. PLoS One 2016; 11:e0164590. [PMID: 27788162 PMCID: PMC5082825 DOI: 10.1371/journal.pone.0164590] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 09/27/2016] [Indexed: 12/11/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the deadliest tumours, but its pathogenesis remains unclear. The involvement of differentially expressed long non-coding RNAs (lncRNAs) in CRC tumorigenesis makes them suitable tumour biomarkers. Methods/Findings Here, we screened 150 cases of CRC and 85 cases of paracancerous tissues in the GEO database for differentially expressed lncRNAs. The levels of lncRNA candidates in 84 CRC and paracancerous tissue samples were validated by qRT-PCR and their clinical significance was analyzed. We identified 15 lncRNAs with differential expression in CRC tumours; among them, AK098081 was significantly up-regulated, whereas AK025209, BC040303, BC037331, AK026659, and CR749831 were down-regulated in CRC. In a receiver operating characteristic curve analysis, the area under the curve for the six lncRNAs was 0.914. High expression of AK098081 and low expression of BC040303, CR749831, and BC037331 indicated poor CRC differentiation. CRC patients with lymph node metastasis had lower expression of BC037331. In addition, the group with high AK098081 expression presented significantly lower overall survival and disease-free survival rates than the low-expression group, confirming AK098081 as an independent risk factor for CRC patients. Conclusion/Significance In conclusion, we have identified multiple CRC-associated lncRNAs from microarray expression profiles that can serve as novel biomarkers for the diagnosis and prognosis of CRC.
Collapse
|
87
|
Combined expression of miR-34a and Smac mediated by oncolytic vaccinia virus synergistically promote anti-tumor effects in Multiple Myeloma. Sci Rep 2016; 6:32174. [PMID: 27552933 PMCID: PMC5001249 DOI: 10.1038/srep32174] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/03/2016] [Indexed: 02/07/2023] Open
Abstract
Despite great progress made in the treatment of multiple myeloma (MM), it is still incurable. Promising phase II clinical results have been reported recently for oncolytic vaccinia virus (OVV) clinic therapeutics. One reason for this has focused on the critical therapeutic importance of the immune response raised by these viruses. However, few studies have performed their applications as an optimal delivery system for therapeutic gene, especially miRNA in MM. In this study, we constructed two novel OVVs (TK deletion) that express anti-tumor genes, miR-34a and Smac, respectively, in MM cell lines and xenograft model. The results demonstrated that the novel OVV can effectively infect MM cell lines, and forcefully enhance the exogenous gene (miR-34a or Smac) expression. Furthermore, utilization of VV-miR-34a combined with VV-Smac synergistically inhibited tumor growth and induced apoptosis in vitro and in vivo. The underlying mechanism is proposed that blocking of Bcl-2 by VV-miR-34a increases the release of cytochrome c from mitochondria and then synergistically amplifies the antitumor effects of Smac-induced cell apoptosis. Our study is the first to utilize OVV as the vector for miR-34a or Smac expression to treat MM, and lays the groundwork for future clinical therapy for MM.
Collapse
|
88
|
Häfner SJ, Talvard TG, Lund AH. Long noncoding RNAs in normal and pathological pluripotency. Semin Cell Dev Biol 2016; 65:1-10. [PMID: 27438587 DOI: 10.1016/j.semcdb.2016.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/05/2016] [Accepted: 07/10/2016] [Indexed: 11/29/2022]
Abstract
The striking similarities between pluripotent and cancer cells, such as immortality and increased stress resistance, have long been acknowledged. Numerous studies searched for and successfully identified common molecular players and pathways, thus providing an entirely new challenge and potential therapeutic angle by targeting cancer cells or a specific stem population of the tumor via pluripotency associated processes. However, these strategies have until now mainly been restricted to proteins. Nonetheless, it has become clear over the past decade that the overwhelming majority of the genome produces noncoding transcripts, many of which have proven both functional and crucial for key cellular processes, including stemness maintenance. Moreover, numerous long noncoding RNAs are deregulated in cancer, but little is known concerning their functions and molecular mechanisms. Consequently, it seems essential to integrate the noncoding transcripts into the picture of the stemness-cancer connection. Whereas a number of studies have addressed the expression of lncRNAs in cancer stem cells, no systematic approach has yet been undertaken to identify lncRNAs implicated in the maintenance of the embryonic stemness state that is hijacked by cancer cells. The aim of this review is to highlight long noncoding RNAs with shared functions in stemness and cancer and to outline the current state of a field in its infancy, the search for long noncoding transcripts in cancer stem cells.
Collapse
Affiliation(s)
- Sophia J Häfner
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, DK-2200, Copenhagen, Denmark.
| | - Thomas G Talvard
- Dansk Fundamental Metrologi, Matematiktorvet 307, DK-2600, Lyngby, Denmark
| | - Anders H Lund
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaloes Vej 5, DK-2200, Copenhagen, Denmark.
| |
Collapse
|