151
|
Brown JM, Wasson MCD, Marcato P. The Missing Lnc: The Potential of Targeting Triple-Negative Breast Cancer and Cancer Stem Cells by Inhibiting Long Non-Coding RNAs. Cells 2020; 9:E763. [PMID: 32244924 PMCID: PMC7140662 DOI: 10.3390/cells9030763] [Citation(s) in RCA: 25] [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: 12/31/2019] [Revised: 03/10/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Treatment decisions for breast cancer are based on staging and hormone receptor expression and include chemotherapies and endocrine therapy. While effective in many cases, some breast cancers are resistant to therapy, metastasize and recur, leading to eventual death. Higher percentages of tumor-initiating cancer stem cells (CSCs) may contribute to the increased aggressiveness, chemoresistance, and worse outcomes among breast cancer. This may be particularly true in triple-negative breast cancers (TNBCs) which have higher percentages of CSCs and are associated with worse outcomes. In recent years, increasing numbers of long non-coding RNAs (lncRNAs) have been identified as playing an important role in breast cancer progression and some of these have been specifically associated within the CSC populations of breast cancers. LncRNAs are non-protein-coding transcripts greater than 200 nucleotides which can have critical functions in gene expression regulation. The preclinical evidence regarding lncRNA antagonists for the treatment of cancer is promising and therefore, presents a potential novel approach for treating breast cancer and targeting therapy-resistant CSCs within these tumors. Herein, we summarize the lncRNAs that have been identified as functionally relevant in breast CSCs. Furthermore, our review of the literature and analysis of patient datasets has revealed that many of these breast CSC-associated lncRNAs are also enriched in TNBC. Together, this suggests that these lncRNAs may be playing a particularly important role in TNBC. Thus, certain breast cancer-promoting/CSC-associated lncRNAs could be targeted in the treatment of TNBCs and the CSCs within these tumors should be susceptible to anti-lncRNA therapy.
Collapse
Affiliation(s)
- Justin M Brown
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.M.B.); (M.-C.D.W.)
| | - Marie-Claire D Wasson
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.M.B.); (M.-C.D.W.)
| | - Paola Marcato
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (J.M.B.); (M.-C.D.W.)
- Departments of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
152
|
Yang F, Tang Z, Duan A, Yi B, Shen N, Bo Z, Yin L, Zhu B, Qiu Y, Li J. Long Noncoding RNA NEAT1 Upregulates Survivin and Facilitates Gallbladder Cancer Progression by Sponging microRNA-335. Onco Targets Ther 2020; 13:2357-2367. [PMID: 32256086 PMCID: PMC7093099 DOI: 10.2147/ott.s236350] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/13/2020] [Indexed: 12/24/2022] Open
Abstract
Background Gallbladder cancer (GBC) is the most common cancer of the biliary tract, but molecularly targeted therapies are not available for GBC. Loss of microRNA (miR)-335 expression may be a useful predictor of clinical outcomes and the reversal of its loss of expression may be a useful treatment strategy for GBC. In this study, we investigated whether a long noncoding RNA, nuclear paraspeckle assembly transcript 1 (NEAT1) sponges miR-335 in GBC cells. Materials and Methods Quantitative reverse transcription-polymerase chain reaction (qRT-PCR), Western blotting, and immunohistochemistry were used to determine the expression of miR-335; NEAT1; survivin; and Ki67 in GBC cell lines (GBC-SD and SGC-996) and tissue samples from patients (n = 25). Cell Counting Kit-8, colony-formation, and Transwell migration and invasion assays were performed to measure cell proliferation, migration, and invasion. Bioinformatic analysis and dual-luciferase reporter assays were utilized to analyze correlativity. Results miR-335 overexpression resulted in inhibition of GBC cell proliferation and invasion. In addition, knockdown of NEAT1 resulted in downregulation of survivin expression. As NEAT1 competitively “sponges” miR-335, NEAT1 knockdown resulted in inhibited GBC cell proliferation and invasion in vitro and GBC tumor growth in vivo. Furthermore, NEAT1 was found to be upregulated in GBC samples, and its expression was inversely correlated with miR-335 levels, but positively correlated with survivin levels. Conclusion These findings indicate that NEAT1 promotes survivin expression by functioning as a competitive endogenous RNA for miR-335 in GBC cells; thus, we have identified a potential biomarker and target for GBC diagnosis and therapy.
Collapse
Affiliation(s)
- Facai Yang
- Department of General Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, People's Republic of China
| | - Zhaohui Tang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Anqi Duan
- Department of Biliary II, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai 200438, People's Republic of China
| | - Bin Yi
- Department of Biliary II, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai 200438, People's Republic of China
| | - Ningjia Shen
- Department of Biliary II, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai 200438, People's Republic of China
| | - Zhiyuan Bo
- Department of Biliary II, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai 200438, People's Republic of China
| | - Lei Yin
- Department of Biliary II, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai 200438, People's Republic of China
| | - Bin Zhu
- Department of Biliary II, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai 200438, People's Republic of China
| | - Yinghe Qiu
- Department of Biliary II, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Military Medical University, Shanghai 200438, People's Republic of China
| | - Jingdong Li
- Department of General Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, People's Republic of China
| |
Collapse
|
153
|
Tang L, Chen Y, Chen H, Jiang P, Yan L, Mo D, Tang X, Yan F. DCST1-AS1 Promotes TGF-β-Induced Epithelial-Mesenchymal Transition and Enhances Chemoresistance in Triple-Negative Breast Cancer Cells via ANXA1. Front Oncol 2020; 10:280. [PMID: 32226772 PMCID: PMC7080863 DOI: 10.3389/fonc.2020.00280] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/18/2020] [Indexed: 12/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly metastatic breast cancer subtype, and the primary systemic treatment strategy involves conventional chemotherapy. DC-STAMP domain containing 1-antisense 1 (DCST1-AS1) is a long non-coding RNA that promotes TNBC migration and invasion. Studying the role of DCST1-AS1 in promoting epithelial–mesenchymal transition (EMT) and chemoresistance will provide a new strategy for TNBC therapy. In the present study, we found that DCST1-AS1 regulates the expression or secretion of EMT-related proteins E-cadherin, snail family zinc finger 1 (SNAI1), vimentin, matrix metallopeptidase 2 (MMP2), and matrix metallopeptidase 9 (MMP9). Interference with DCST1-AS1 impaired TGF-β-induced TNBC cell invasion and migration. DCST1-AS1 directly binds to ANXA1 in BT-549 cells and affects the expression of ANXA1. DCST1-AS1 enhances TGF-β/Smad signaling in BT-549 cells through ANXA1 to promote EMT. The combination of DCST1-AS1 and ANXA1 also contributes to enhancement of the resistance of BT-549 cells to doxorubicin and paclitaxel. In conclusion, DCST1-AS1 promotes TGF-β-induced EMT and enhances chemoresistance in TNBC cells through ANXA1, and therefore represents a potentially promising target for metastatic breast cancer therapy.
Collapse
Affiliation(s)
- Li Tang
- Department of Clinical Laboratory, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yuli Chen
- Department of Clinical Laboratory, Nanjing Qixia District Hospital, Nanjing, China
| | - Huanhuan Chen
- The Fourth Clinical Medical School, Nanjing Medical University, Nanjing, China
| | - Pan Jiang
- The Fourth Clinical Medical School, Nanjing Medical University, Nanjing, China
| | - Linping Yan
- Department of Clinical Laboratory, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Dongping Mo
- Department of Clinical Laboratory, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xun Tang
- Department of Clinical Laboratory, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Yan
- Department of Clinical Laboratory, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
154
|
Agostini M, Ganini C, Candi E, Melino G. The role of noncoding RNAs in epithelial cancer. Cell Death Discov 2020; 6:13. [PMID: 32194993 PMCID: PMC7067833 DOI: 10.1038/s41420-020-0247-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
Regulatory noncoding RNAs (ncRNAs) are a class of RNAs transcribed by regions of the human genome that do not encode for proteins. The three main members of this class, named microRNA, long noncoding RNA, and circular RNA play a key role in the regulation of gene expression, eventually shaping critical cellular processes. Compelling experimental evidence shows that ncRNAs function either as tumor suppressors or oncogenes by participating in the regulation of one or several cancer hallmarks, including evading cell death, and their expression is frequently deregulated during cancer onset, progression, and dissemination. More recently, preclinical and clinical studies indicate that ncRNAs are potential biomarkers for monitoring cancer progression, relapse, and response to cancer therapy. Here, we will discuss the role of noncoding RNAs in regulating cancer cell death, focusing on those ncRNAs with a potential clinical relevance.
Collapse
Affiliation(s)
- Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Carlo Ganini
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
- IDI-IRCCS, Via Monti di Creta 106, 00166 Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
- MRC Toxicology Unit, University of Cambridge, Department of Pathology, Tennis Court Road, Cambridge, CB2 1QP UK
| |
Collapse
|
155
|
Prabhu KS, Raza A, Karedath T, Raza SS, Fathima H, Ahmed EI, Kuttikrishnan S, Therachiyil L, Kulinski M, Dermime S, Junejo K, Steinhoff M, Uddin S. Non-Coding RNAs as Regulators and Markers for Targeting of Breast Cancer and Cancer Stem Cells. Cancers (Basel) 2020; 12:cancers12020351. [PMID: 32033146 PMCID: PMC7072613 DOI: 10.3390/cancers12020351] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/27/2020] [Accepted: 02/02/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is regarded as a heterogeneous and complicated disease that remains the prime focus in the domain of public health concern. Next-generation sequencing technologies provided a new perspective dimension to non-coding RNAs, which were initially considered to be transcriptional noise or a product generated from erroneous transcription. Even though understanding of biological and molecular functions of noncoding RNA remains enigmatic, researchers have established the pivotal role of these RNAs in governing a plethora of biological phenomena that includes cancer-associated cellular processes such as proliferation, invasion, migration, apoptosis, and stemness. In addition to this, the transmission of microRNAs and long non-coding RNAs was identified as a source of communication to breast cancer cells either locally or systemically. The present review provides in-depth information with an aim at discovering the fundamental potential of non-coding RNAs, by providing knowledge of biogenesis and functional roles of micro RNA and long non-coding RNAs in breast cancer and breast cancer stem cells, as either oncogenic drivers or tumor suppressors. Furthermore, non-coding RNAs and their potential role as diagnostic and therapeutic moieties have also been summarized.
Collapse
Affiliation(s)
- Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (H.F.); (E.I.A.); (S.K.); (L.T.); (M.K.); (M.S.); (S.U.)
- Correspondence: ; Tel.: +974-4439-0966
| | - Afsheen Raza
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha 3050, Qatar; (A.R.); (S.D.)
| | | | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow 226003, India;
| | - Hamna Fathima
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (H.F.); (E.I.A.); (S.K.); (L.T.); (M.K.); (M.S.); (S.U.)
| | - Eiman I. Ahmed
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (H.F.); (E.I.A.); (S.K.); (L.T.); (M.K.); (M.S.); (S.U.)
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (H.F.); (E.I.A.); (S.K.); (L.T.); (M.K.); (M.S.); (S.U.)
- Qatar College of Pharmacy, Qatar University, Doha 3050, Qatar
| | - Lubna Therachiyil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (H.F.); (E.I.A.); (S.K.); (L.T.); (M.K.); (M.S.); (S.U.)
- Qatar College of Pharmacy, Qatar University, Doha 3050, Qatar
| | - Michal Kulinski
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (H.F.); (E.I.A.); (S.K.); (L.T.); (M.K.); (M.S.); (S.U.)
| | - Said Dermime
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha 3050, Qatar; (A.R.); (S.D.)
| | - Kulsoom Junejo
- General Surgery Department, Hamad General Hospital, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (H.F.); (E.I.A.); (S.K.); (L.T.); (M.K.); (M.S.); (S.U.)
- Department of Dermatology Venereology, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology, Weill Cornell Medicine, Qatar Foundation, Education City, Doha 24144, Qatar
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (H.F.); (E.I.A.); (S.K.); (L.T.); (M.K.); (M.S.); (S.U.)
| |
Collapse
|
156
|
Knutsen E, Lellahi SM, Aure MR, Nord S, Fismen S, Larsen KB, Gabriel MT, Hedberg A, Bjørklund SS, Bofin AM, Mælandsmo GM, Sørlie T, Mortensen ES, Perander M. The expression of the long NEAT1_2 isoform is associated with human epidermal growth factor receptor 2-positive breast cancers. Sci Rep 2020; 10:1277. [PMID: 31992741 PMCID: PMC6987222 DOI: 10.1038/s41598-020-57759-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/21/2019] [Indexed: 12/21/2022] Open
Abstract
The long non-coding RNA NEAT1 locus is transcribed into two overlapping isoforms, NEAT1_1 and NEAT1_2, of which the latter is essential for the assembly of nuclear paraspeckles. NEAT1 is abnormally expressed in a wide variety of human cancers. Emerging evidence suggests that the two isoforms have distinct functions in gene expression regulation, and recently it was shown that NEAT1_2, but not NEAT1_1, expression predicts poor clinical outcome in cancer. Here, we report that NEAT1_2 expression correlates with HER2-positive breast cancers and high-grade disease. We provide evidence that NEAT1_1 and NEAT1_2 have distinct expression pattern among different intrinsic breast cancer subtypes. Finally, we show that NEAT1_2 expression and paraspeckle formation increase upon lactation in humans, confirming what has previously been demonstrated in mice.
Collapse
Affiliation(s)
- Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Seyed Mohammad Lellahi
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Miriam Ragle Aure
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Silje Nord
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Silje Fismen
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Kenneth Bowitz Larsen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Marta Tellez Gabriel
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Annica Hedberg
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Sunniva Stordal Bjørklund
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | | | - Anna Mary Bofin
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Therese Sørlie
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Elin Synnøve Mortensen
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.,Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Maria Perander
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
157
|
Rahmani Z, Mojarrad M, Moghbeli M. Long non-coding RNAs as the critical factors during tumor progressions among Iranian population: an overview. Cell Biosci 2020; 10:6. [PMID: 31956395 PMCID: PMC6961246 DOI: 10.1186/s13578-020-0373-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cancer is associated with various genetic and environmental risk factors. Beside the mutations or aberrant expression of protein-coding genes, the genetic deregulation of non-coding RNAs has also an important role during tumor progression and metastasis. Long non-coding RNAs (lncRNAs) are a class of ncRNAs larger than 200 nucleotides that may function as tumor-suppressor or oncogene. MAIN BODY There is a raising trend of cancer incidence among Iranian population during the last decades. Therefore, it is required to prepare a general population specific panel of genetic markers for the early detection of cancer in this population. The tissue-specific expression characteristics and high stability in body fluids highlight the lncRNAs as efficient diagnostic and prognostic noninvasive biomarkers in cancer. In present review we summarized all of the lncRNAs which have been reported until now in different tumors among Iranian patients. CONCLUSIONS This review paves the way of introducing a population based noninvasive diagnostic panel of lncRNAs for the early detection of tumor cells among Iranian population.
Collapse
Affiliation(s)
- Zahra Rahmani
- Department of Medical Genetics, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
158
|
Xu Z, Liu C, Zhao Q, Lü J, Ding X, Luo A, He J, Wang G, Li Y, Cai Z, Wang Z, Liu J, Liu S, Li W, Yu Z. Long non-coding RNA CCAT2 promotes oncogenesis in triple-negative breast cancer by regulating stemness of cancer cells. Pharmacol Res 2020; 152:104628. [PMID: 31904506 DOI: 10.1016/j.phrs.2020.104628] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/22/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancers (TNBC) are more aggressive due to lacking receptors for hormone therapy and maintaining stemness features in cancer cells. Herein we found long non-coding RNA CCAT2 overexpressed specially in TNBC, and in breast cancer stem cells (BCSC) as well. Enforced overexpression and targeted knockdown demonstrated the oncogenic function of CCAT2 both in vitro and in vivo. CCAT2 promoted the expression of stemness markers including OCT4, Nanog and KLF4, increased mammosphere formation and induced ALDH+ cancer stem cell population in TNBC. A chromosomally adjacent gene OCT4-PG1, as a pseudogene of OCT4, was upregulated by CCAT2, and positively regulated the stemness features of TNBC cells. miR-205 was identified as a target gene of CCAT2 in TNBC. Point-mutation in CCAT2 impaired the sponge inhibition of miR-205. Overexpression of miR-205 rescued the oncogenic phenotypes induced by CCAT2. In addition, Notch2, as a target gene of miR-205, was downregulated by miR-205 and upregulated by CCAT2 in TNBC. Collectively, the current study revealed a novel function of CCAT2 in promoting tumor initiation and progression in TNBC through upregulating OCT4-PG1 expression and activating Notch signaling. These findings not only demonstrated a lncRNA-based therapeutic strategy in treatment of TNBC, but also added a node to the regulatory network of CCAT2 that controls aggressiveness of breast cancer stem cells.
Collapse
Affiliation(s)
- Zhen Xu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China; Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cuiui Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China; Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qian Zhao
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jinhui Lü
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xin Ding
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - An Luo
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jia He
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Guangxue Wang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yuan Li
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhaoqing Cai
- Tongji University School of Life Science and Technology, Shanghai, China
| | - Zhongrui Wang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China; Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Junjun Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China; Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Suling Liu
- Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenshu Li
- Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Zuoren Yu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
159
|
Jiang X, Guo S, Zhang Y, Zhao Y, Li X, Jia Y, Xu Y, Ma B. LncRNA NEAT1 promotes docetaxel resistance in prostate cancer by regulating ACSL4 via sponging miR-34a-5p and miR-204-5p. Cell Signal 2020; 65:109422. [DOI: 10.1016/j.cellsig.2019.109422] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/14/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022]
|
160
|
Lin LC, Lee HT, Chien PJ, Huang YH, Chang MY, Lee YC, Chang WW. NAD(P)H:quinone oxidoreductase 1 determines radiosensitivity of triple negative breast cancer cells and is controlled by long non-coding RNA NEAT1. Int J Med Sci 2020; 17:2214-2224. [PMID: 32922184 PMCID: PMC7484675 DOI: 10.7150/ijms.45706] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022] Open
Abstract
Radioresistant cells cause recurrence in patients with breast cancer after they undergo radiation therapy. The molecular mechanisms by which cancer cells obtain radioresistance should be understood to develop radiation-sensitizing agents. Results showed that the protein expression and activity of NAD(P)H:quinone oxidoreductase 1 (NQO1) were upregulated in radioresistant MDA-MB-231 triple-negative breast cancer (TNBC) cells. NQO1 knockdown inhibited the proliferation of NQO1 expressing Hs578t TNBC cells or the radioresistant MDA-MB-231 cells, whereas NOQ1 overexpression increased the survival of MDA-MB-231 cells, which lack of NQO1 expression originally, under irradiation. The cytotoxicity of β-lapachone, an NQO1-dependent bioactivatable compound, was greater in radioresistant MDA-MB-231 cells than in parental cells. β-lapachone displayed a radiosensitization effect on Hs578t or radioresistant MBDA-MB-231 cells. The expression of the long noncoding RNA NEAT1 positively regulated the NQO1 expression in radioresistant MDA-MB-231 cells at a translational level rather than at a transcription level. The inhibition of the NEAT1 expression through the CRISPR-Cas9 method increased the sensitivity of radioresistant MDA-MB-231 cells to radiation and decreased their proliferation, the activity of cancer stem cells, and the expression of stemness genes, including BMI1, Oct4, and Sox2. In conclusion, the NQO1 expression in triple-negative breast cancer cells determined their radiosensitivity and was controlled by NEAT1. In addition, NOQ1 bioactivatable compounds displayed potential for application in the development of radiation sensitizers in breast cancer.
Collapse
Affiliation(s)
- Li-Ching Lin
- Department of Radiation Oncology, Chi-Mei Foundation Medical Center, Tainan, Taiwan.,School of Medicine, Taipei Medical University, Taipei, Taiwan.,Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Hsueh-Te Lee
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming University, Taipei City, Taiwan
| | - Peng-Ju Chien
- School of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Hao Huang
- School of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Mu-Ya Chang
- School of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Yueh-Chun Lee
- Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Wen-Wei Chang
- School of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
161
|
Feng Y, Gao L, Cui G, Cao Y. LncRNA NEAT1 facilitates pancreatic cancer growth and metastasis through stabilizing ELF3 mRNA. Am J Cancer Res 2020; 10:237-248. [PMID: 32064164 PMCID: PMC7017733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/22/2019] [Indexed: 06/10/2023] Open
Abstract
Recently, increasing evidence has revealed that long noncoding RNAs (lncRNAs) play important roles in the pathogenesis of multiple cancers. Although the oncogenic effects of lncRNA nuclear-enriched abundant transcript 1 (NEAT1) in some cancers have been reported, the functional significance and molecular mechanism of NEAT1 in pancreatic cancer (PC) progression remains elusive. In this study, our findings showed that NEAT1 expression was upregulated in PC tissues and cell lines; high NEAT1 expression was associated with tumor size, TNM stage, lymph node and distant metastasis, and also predicted poor prognosis. Functional experiments demonstrated that NEAT1 could promote PC cell proliferation and metastasis both in vitro and in vivo. Mechanistically, NEAT1 could associate with E74 like ETS transcription factor 3 (ELF3) mRNA and enhance the combination of Insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) and ELF3 mRNA, subsequently suppressing the degradation of ELF3 mRNA. Overall, our research indicates that NEAT1 might be a potential therapeutic target for patients with PC.
Collapse
Affiliation(s)
- Yaguang Feng
- Department of Gastrointestinal and Hepatobiliary Surgery, The First People's Hospital of Shangqiu Shangqiu 476000, Henan Province, China
| | - Ling Gao
- Department of Gastrointestinal and Hepatobiliary Surgery, The First People's Hospital of Shangqiu Shangqiu 476000, Henan Province, China
| | - Guangfei Cui
- Department of Gastrointestinal and Hepatobiliary Surgery, The First People's Hospital of Shangqiu Shangqiu 476000, Henan Province, China
| | - Yan Cao
- Department of Gastrointestinal and Hepatobiliary Surgery, The First People's Hospital of Shangqiu Shangqiu 476000, Henan Province, China
| |
Collapse
|
162
|
Huang QY, Liu GF, Qian XL, Tang LB, Huang QY, Xiong LX. Long Non-Coding RNA: Dual Effects on Breast Cancer Metastasis and Clinical Applications. Cancers (Basel) 2019; 11:E1802. [PMID: 31744046 PMCID: PMC6896003 DOI: 10.3390/cancers11111802] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
As a highly heterogeneous malignancy, breast cancer (BC) has become the most significant threat to female health. Distant metastasis and therapy resistance of BC are responsible for most of the cases of mortality and recurrence. Distant metastasis relies on an array of processes, such as cell proliferation, epithelial-to-mesenchymal transition (EMT), mesenchymal-to-epithelial transition (MET), and angiogenesis. Long non-coding RNA (lncRNA) refers to a class of non-coding RNA with a length of over 200 nucleotides. Currently, a rising number of studies have managed to investigate the association between BC and lncRNA. In this study, we summarized how lncRNA has dual effects in BC metastasis by regulating invasion, migration, and distant metastasis of BC cells. We also emphasize that lncRNA has crucial regulatory effects in the stemness and angiogenesis of BC. Clinically, some lncRNAs can regulate chemotherapy sensitivity in BC patients and may function as novel biomarkers to diagnose or predict prognosis for BC patients. The exact impact on clinical relevance deserves further study. This review can be an approach to understanding the dual effects of lncRNAs in BC, thereby linking lncRNAs to quasi-personalized treatment in the future.
Collapse
Affiliation(s)
- Qi-Yuan Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Guo-Feng Liu
- First Clinical Medical College, Nanchang University, Nanchang 330006, China;
| | - Xian-Ling Qian
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
- First Clinical Medical College, Nanchang University, Nanchang 330006, China;
| | - Li-Bo Tang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qing-Yun Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
| | - Li-Xia Xiong
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (Q.-Y.H.); (X.-L.Q.); (L.-B.T.); (Q.-Y.H.)
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang 330006, China
| |
Collapse
|
163
|
Quan D, Chen K, Zhang J, Guan Y, Yang D, Wu H, Wu S, Lv L. Identification of lncRNA NEAT1/miR-21/RRM2 axis as a novel biomarker in breast cancer. J Cell Physiol 2019; 235:3372-3381. [PMID: 31621912 DOI: 10.1002/jcp.29225] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/03/2019] [Indexed: 01/01/2023]
Abstract
As a major disease that threatens the health of women worldwide, breast cancer (BC) lacks effective molecular markers in the clinic at the same time. We aim at finding a new biomarker of BC. In our study, through the Gene Expression Omnibus database chip, a total of 1393 pairs of microRNA-messenger RNA (miRNA-mRNA) networks and 35754 pairs of long noncoding RNA-miRNA networks were obtained. We found out that NEAT1/miR-21/RRM2 axis may play a role in BC diagnosis and prognosis. The real-time quantitative reverse transcription-polymerase chain reaction test was used to analyze the mRNA level of NEAT1, miR-21, and RRM2. Western blot was used to detect the protein level of RRM2. Through the 5-ethynyl-2'-deoxyuridine assay, the proliferation of MDA-MB-231 cells was detected. Through wound healing and transwell assay, the migration of MDA-MB-231 cells was detected. Altogether, our data indicated that NEAT1, miR-21, and RRM2 were upregulated in several BC cell lines. Overexpressed of miR-21 in MDA-MB-231 cells promote proliferation and migration. Besides, our results demonstrated that overexpressed of miR-21 upregulated the level of RRM2. Accordingly, miR-21/RRM2 might be a new diagnosis and treatment target of BC.
Collapse
Affiliation(s)
- Dongling Quan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Kai Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingru Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiqing Guan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Danni Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Huanxian Wu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Shaoyu Wu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Lin Lv
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
164
|
Yang Y, Hao E, Pan X, Tan D, Du Z, Xie J, Hou X, Deng J, Wei K. Gomisin M2 from Baizuan suppresses breast cancer stem cell proliferation in a zebrafish xenograft model. Aging (Albany NY) 2019; 11:8347-8361. [PMID: 31612865 PMCID: PMC6814583 DOI: 10.18632/aging.102323] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/22/2019] [Indexed: 12/24/2022]
Abstract
Gomisin M2 isolated from Schisandra viridis A. C. Smith has potential anti-tumor effects on certain cancers, including breast cancer. However, only a few investigations have been conducted on the effects of Gomisin M2 on breast cancer stem cells (CSCs), which have the ability to self-renew and differentiate, as a possible strategy to resolve cancer cell resistance to apoptosis and to improve treatments. It is essential to investigate the effects of Gomisin M2 on breast cancer stem cells (BCSCs). In this study, we enriched breast cancer stem cells with CD44+/CD24- from MDA-MB-231 and HCC1806 cells through magnetic-activated cell sorting and cultured these in serum-free medium. The ability of Gomisin M2 to kill breast cancer stem cells was evaluated in vitro and in vivo. Gomisin M2 significantly inhibited the proliferation of the triple-negative breast cancer cell lines and mammosphere formation in breast CSCs and downregulated the Wnt/β-catenin self-renewal pathway. Moreover, Gomisin M2 induced apoptosis and blocked the mitochondrial membrane potential of BCSCs. Gomisin M2 suppressed the proliferation of MDA-MB-231 and HCC1806 xenografts in zebrafish. Together, these findings suggest that the anti-BCSC activity of Gomisin M2 could become a promising starting point for the discovery of novel BCSC-targeting drugs.
Collapse
Affiliation(s)
- Yeguo Yang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Xianglong Pan
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Dechao Tan
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Zhengcai Du
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Jinling Xie
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Xiaotao Hou
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Sino-Canada Joint Zebrafish Lab for Chinese Herbal Drug Screening, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Kun Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
165
|
Yousefi H, Maheronnaghsh M, Molaei F, Mashouri L, Reza Aref A, Momeny M, Alahari SK. Long noncoding RNAs and exosomal lncRNAs: classification, and mechanisms in breast cancer metastasis and drug resistance. Oncogene 2019; 39:953-974. [PMID: 31601996 DOI: 10.1038/s41388-019-1040-y] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/04/2019] [Accepted: 09/20/2019] [Indexed: 12/16/2022]
Abstract
Breast cancer is the most common cancer, and the second cause of cancer-related deaths (after lung cancer) among women. Developing tumor metastasis and invasion is the most important cause of death in breast cancer patients. Several key factors participate in breast cancer metastasis including long noncoding RNAs (lncRNAs). lncRNAs are a category of cellular RNAs that are longer than 200 nucleotides in length. Accumulating evidence suggests that lncRNAs have the potential to be promising diagnostic, prognostic biomarkers and therapeutic targets in breast cancer. Understanding the role of lncRNAs and their mechanisms of functions might help to further discovery of breast cancer biological characteristics. In this review, we discuss physiological functions, epigenetic regulation, transcriptional regulation of lncRNAs, and their important role in tumor progression and metastasis. Some lncRNAs function as oncogenes and some function as tumor suppressors. Interestingly, recent reports depict that hypomethylation of promoters of lncRNAs play a pivotal role in cancer progression, suggesting the importance of epigenetic regulation. Furthermore, we discuss the role of lncRNAs in exosomes and their function in drug resistance, and therapeutic importance of exosomal lncRNAs in cancer biology. In summary, lncRNAs have a great potential to consider them as novel prognostic biomarkers as well as new therapeutic targets in breast cancer.
Collapse
Affiliation(s)
- Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, LA, USA
| | - Maryam Maheronnaghsh
- Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Molaei
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ladan Mashouri
- Department of Genetics, Faculty of Science, Shahrekord University, Shahrekord, Iran
| | - Amir Reza Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Majid Momeny
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
166
|
Li X, Zhou Y, Yang L, Ma Y, Peng X, Yang S, Li H, Liu J. LncRNA NEAT1 promotes autophagy via regulating miR-204/ATG3 and enhanced cell resistance to sorafenib in hepatocellular carcinoma. J Cell Physiol 2019; 235:3402-3413. [PMID: 31549407 DOI: 10.1002/jcp.29230] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Long noncoding RNAs (lncRNAs) has been acknowledged in tumorigenesis gradually because of the great importance in different cancers. LncRNA nuclear enriched abundant transcript 1 (NEAT1) is a novel lncRNA and has been reported to promote multiple cancer progression. However, the biological roles of NEAT1 in hepatocellular carcinoma (HCC) is not cleared nowadays. In the present research, the level of NEAT1 was found to be upregulated in HCC by The Cancer Genome Atlas. In addition, NEAT1 expression is negatively correlated with the survival rate in HCC. Further investigation revealed that NEAT1 upregulation inhibited sorafenib efficacy and promoted autophagy. We found that NEAT1 could be a sponge for microRNA-204 (miR-204) and inhibits its level to upregulate ATG3 expression. In addition to the above, we demonstrated that miR-204 mimics also attenuated tumor autophagy. And rescue assays demonstrated that NEAT1 promotes HCC autophagy through modulating miR-204/ATG3 pathway. Collectively, this study first demonstrated that a novel NEAT1/miR-204/ATG3 signaling regulates HCC progression.
Collapse
Affiliation(s)
- Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Yong Zhou
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Yingbo Ma
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Jingang Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
167
|
Li C, Hu G, Wei B, Wang L, Liu N. lncRNA LINC01494 Promotes Proliferation, Migration And Invasion In Glioma Through miR-122-5p/CCNG1 Axis. Onco Targets Ther 2019; 12:7655-7662. [PMID: 31571916 PMCID: PMC6756415 DOI: 10.2147/ott.s213345] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022] Open
Abstract
Background Long noncoding RNAs (lncRNAs) are recognized as key effectors in tumor, including glioma. LINC01494 is an uncharacterized novel lncRNA. In this research, we aimed to investigate the function of LINC01494 in glioma. Methods Gene relative expression was analyzed by qRT-PCR method. CCK8, colony formation and Transwell assay was used to determine cell proliferation, migration and invasion. Bioinformatics analyses were used to predict the target of LINC01494 and miR-122-5p. Luciferase reporter assay was utilized to validate the interactions between LINC01494 and miR-122-5p or CCNG1 and miR-122-5p. Results LINC01494 was identified as a significantly upregulated lncRNA in glioma through bioinformatics analysis. Furthermore, LINC01494 upregulation indicated poor prognosis. Meanwhile, in vitro investigation indicated that silencing LINC01494 with siRNAs obviously inhibited the proliferation, cell cycle, migration and invasion of glioma cells. Besides, it is found that LINC01494 expression was negatively correlated with miR-122-5p. We demonstrated that LINC01494 inhibited miR-122-5p to upregulate CCNG1 expression through direct interaction. Rescue assay further demonstrated that LINC01494/miR-122-5p/CCNG1 signaling cascade plays a critical role in regulating glioma cell proliferation, migration and invasion. Conclusion Taken together, our findings demonstrated the essential function and molecular mechanism of LINC01494 in glioma progression.
Collapse
Affiliation(s)
- Chang Li
- Department of VIP Unit, China-Japan Union Hospital of Jilin University, Changchun 130031, People's Republic of China
| | - Guozhang Hu
- Department of First-aid Medicine, China-Japan Union Hospital of Jilin University, Changchun 130031, People's Republic of China
| | - Bo Wei
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130031, People's Republic of China
| | - Le Wang
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Naijie Liu
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130031, People's Republic of China
| |
Collapse
|
168
|
Najafi M, Mortezaee K, Majidpoor J. Cancer stem cell (CSC) resistance drivers. Life Sci 2019; 234:116781. [PMID: 31430455 DOI: 10.1016/j.lfs.2019.116781] [Citation(s) in RCA: 263] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/02/2019] [Accepted: 08/16/2019] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSCs) are a population of self-renewal cells with high tumorigenic potency. CSCs can adopt easily with changes in the nearby milieu, and are more resistant to conventional therapies than other cells within a tumor. CSC resistance can be induced secondary to radio- and chemotherapy, or even after chemotherapy secession. A combination of both intrinsic and extrinsic factors is contributed to CSC-mediated therapy resistance. CSCs represent protective autophagy and efficient cell cycling, along with highly qualified epithelial-mesenchymal transition (EMT) regulators, reactive oxygen species (ROS) scavengers, drug transporters, and anti-apoptotic and DNA repairing systems. In addition, CSCs develop cross-talking and share some characteristics with other cells within the tumor microenvironment (TME) being more intense in higher stage tumors, and thereby sophisticating tumor-targeted therapies. TME, in fact, is a nest for aggravating resistance mechanisms in CSCs. TME is exposed constantly to the nutritional, metabolic and oxygen deprivation; these conditions promote CSC adaptation. This review is aimed to discuss main (intrinsic and extrinsic) mechanisms of CSC resistance and suggest some strategies to revoke this important promoter of therapy failure.
Collapse
Affiliation(s)
- Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran; Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
169
|
Luo Y, Hao T, Zhang J, Zhang M, Sun P, Wu L. MicroRNA-592 suppresses the malignant phenotypes of thyroid cancer by regulating lncRNA NEAT1 and downregulating NOVA1. Int J Mol Med 2019; 44:1172-1182. [PMID: 31524231 DOI: 10.3892/ijmm.2019.4278] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 07/05/2019] [Indexed: 11/05/2022] Open
Abstract
Numerous studies have demonstrated that various microRNAs (miRs) are aberrantly expressed in thyroid cancer and play critical roles in thyroid cancer malignancy. The aberrant expression of miR‑592 has frequently been reported in multiple human cancer types; however, its expression profile and functions in thyroid cancer remain poorly understood. Reverse transcription‑quantitative polymerase chain reaction was carried out to determine the expression profile of miR‑592 in thyroid cancer tissues and cell lines. The regulatory effects of miR‑592 upregulation on thyroid cancer cell proliferation, migration, and invasion in vitro, and tumor growth in vivo were investigated using a CCK‑8 assay, migration and invasion assays, and a xenograft tumor model, respectively. Furthermore, the mechanisms underlying miR‑592‑mediated suppression of the aggressive phenotypes of thyroid cancer cells were explored in detail. The results indicated that miR‑592 was significantly downregulated in thyroid cancer samples, and its downregulation was associated with lymph node metastasis and tumor‑node‑metastasis stage. Patients with thyroid cancer and low miR‑592 expression exhibited shorter overall survival than patients with high miR‑592 expression. Overexpression of miR‑592 resulted in decreased cell proliferation, migration, and invasion in thyroid cancer. In addition, neuro‑oncological ventral antigen 1 (NOVA1) was identified as a novel target gene of miR‑592 in thyroid cancer cells. Furthermore, ectopic NOVA1 expression may effectively abolish the tumor‑suppressing effects of miR‑592 overexpression in thyroid cancer cells. Notably, the lncRNA NEAT1 was proposed to function as a sponge of miR‑592 in thyroid cancer cells, thereby regulating NOVA1 expression. Finally, resuming miR‑592 expression significantly impaired thyroid cancer tumor growth in vivo. The results indicated that the NEAT1/miR‑592/NOVA1 pathway may play regulatory roles in thyroid cancer malignancy in vitro and in vivo. Our findings may provide novel insight into the pathogenesis of thyroid cancer. Therefore, this pathway may be an effective target for treating patients with this disease.
Collapse
Affiliation(s)
- Yiqiang Luo
- Department of Breast and Thyroid Surgery, Jilin Central General Hospital, Chuanying, Jilin 132000, P.R. China
| | - Tianwei Hao
- Department of Breast and Thyroid Surgery, Jilin Central General Hospital, Chuanying, Jilin 132000, P.R. China
| | - Jian Zhang
- Department of Breast and Thyroid Surgery, Jilin Central General Hospital, Chuanying, Jilin 132000, P.R. China
| | - Ming Zhang
- Department of Breast and Thyroid Surgery, Jilin Central General Hospital, Chuanying, Jilin 132000, P.R. China
| | - Peng Sun
- Department of Breast and Thyroid Surgery, Jilin Central General Hospital, Chuanying, Jilin 132000, P.R. China
| | - Lei Wu
- Department of Breast and Thyroid Surgery, Jilin Central General Hospital, Chuanying, Jilin 132000, P.R. China
| |
Collapse
|
170
|
Du Z, Gao W, Sun J, Li Y, Sun Y, Chen T, Ge S, Guo W. Identification of long non‑coding RNA‑mediated transcriptional dysregulation triplets reveals global patterns and prognostic biomarkers for ER+/PR+, HER2‑ and triple negative breast cancer. Int J Mol Med 2019; 44:1015-1025. [PMID: 31257479 PMCID: PMC6657959 DOI: 10.3892/ijmm.2019.4261] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BRCA) is the most common type of cancer in adult females. Estrogen receptor (ER)+/progesterone receptor (PR)+, human epidermal-growth factor receptor 2 (HER2)− BRCA and triple-negative breast cancer (TNBC) are two important subtypes of this disease. Long non-coding RNA (lncRNA)-mediated transcriptional dysregulation triplets (lncTDTs) may contribute to the development of cancer; however, the precise functional roles of lncTDTs in ER+/PR+, HER2− BRCA and TNBC require further investigation. In the present study, an integrated and computational approach was conducted to identify lncTDTs based on transcription factor (TF), gene, lncRNA expression profiles and experimentally verified TF-gene interactions. The regulatory patterns of these lncTDTs are complex and differed in ER+/PR+, HER2− BRCA and TNBC. Of note, five common lncTDTs were reported for these BRCA subtypes. Functional analysis revealed lncTDTs to be enriched in the PI3K/AKT signaling pathway within the two BRCA subtypes. Additionally, certain lncTDTs were associated with survival and may be considered candidate prognostic biomarkers for BRCA subtypes. Collectively, the results of the present study provide novel insight into the functions and mechanisms of lncRNAs in ER+/PR+, HER2− BRCA and TNBC, and may aid the development of targeted treatments against certain subtypes of BRCA.
Collapse
Affiliation(s)
- Zhengwei Du
- Department of Thyroid Breast Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Wei Gao
- Department of Thyroid Breast Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Jiayi Sun
- Department of Thyroid Breast Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Yujing Li
- Department of Thyroid Breast Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Yu Sun
- Department of Thyroid Breast Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Tong Chen
- Department of Thyroid Breast Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Shuke Ge
- Department of Thyroid Breast Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Wenbin Guo
- Department of Thyroid Breast Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| |
Collapse
|