351
|
He X, Liu H, Guo F, Feng Y, Gao Y, Sun F, Song B, Lu H, Li Y. Long non-coding RNA Z38 promotes cell proliferation and metastasis in human renal cell carcinoma. Mol Med Rep 2017; 16:5489-5494. [DOI: 10.3892/mmr.2017.7218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 03/20/2017] [Indexed: 11/06/2022] Open
|
352
|
Moschovis D, Gazouli M, Tzouvala M, Vezakis A, Karamanolis G. Long non-coding RNA in pancreatic adenocarcinoma and pancreatic neuroendocrine tumors. Ann Gastroenterol 2017; 30:622-628. [PMID: 29118556 PMCID: PMC5670281 DOI: 10.20524/aog.2017.0185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 07/05/2017] [Indexed: 12/21/2022] Open
Abstract
Interest in non-coding regions of DNA has been increasing since the mapping of the human genome revealed that human DNA contains far fewer genes encoding proteins than previously expected. However, analysis of the derivatives of DNA transcription (transcriptomics) revealed that the majority of the genetic material is transcribed into non-coding RNA (ncRNA), indicating that these molecules probably provide the functional diversity and complexity of the physiology of the human body that cannot be attributed to the proteins. Of these ncRNA, long ncRNA (lncRNA) have a length greater than 200 nucleotides and share many common components with the coding messenger RNA (mRNA): They are transcribed by RNA polymerase II, comprised of multiple exons and subjected to normal RNA splicing giving RNA products of several kilobases. Scientific data reveal the regulatory role of lncRNA in the control of gene expression during cell development and homeostasis. However, to date, very few lncRNAs have been characterized in depth, and lncRNAs are thought to have a wide range of functions in cellular and developmental processes. These molecules will have the possibility to be used as biomarkers and contribute to the development of targeted therapies. Concerning pancreatic cancer, there are limited data in the literature that correlate the growth of these tumors with deregulation of various lncRNA. We herein review the literature regarding the role of lncRNA as a diagnostic and prognostic biomarker and possible therapeutic target in the neoplasms of the pancreas, particularly pancreatic adenocarcinoma and pancreatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Dimitrios Moschovis
- Department of Gastroenterology, Agios Panteleimon General Hospital, Nikea (Dimitrios Moschovis, Maria Tzouvala), Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (Maria Gazouli), Greece
| | - Maria Tzouvala
- Department of Gastroenterology, Agios Panteleimon General Hospital, Nikea (Dimitrios Moschovis, Maria Tzouvala), Greece
| | - Antonios Vezakis
- 2 Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens (Antonios Vezakis), Greece
| | - George Karamanolis
- Gastroenterology Unit, 2 Department of Surgery, Aretaieio University Hospital, National and Kapodistrian University of Athens (George Karamanolis), Greece
| |
Collapse
|
353
|
Jin L, Fu H, Quan J, Pan X, He T, Hu J, Li Y, Li H, Yang Y, Ye J, Zhang F, Ni L, Yang S, Lai Y. Overexpression of long non-coding RNA differentiation antagonizing non-protein coding RNA inhibits the proliferation, migration and invasion and promotes apoptosis of renal cell carcinoma. Mol Med Rep 2017; 16:4463-4468. [PMID: 28765964 DOI: 10.3892/mmr.2017.7135] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 05/15/2017] [Indexed: 11/06/2022] Open
Abstract
Renal cell carcinoma (RCC) is the third most common cancer in the urological system; however, the pathogenesis remains unknown. Accumulating evidence has demonstrated that long non‑coding RNAs are dysregulated in various tumors and serves an important role in tumorigenesis and development. In the present study, expression of lncRNA differentiation antagonizing non‑protein coding RNA (DANCR) in 24 paired RCC and adjacent normal tissues was detected by reverse transcription‑quantitative polymerase chain reaction. The results revealed that DANCR is downregulated in RCC tissues compared with adjacent normal tissues. Subsequent study revealed that overexpression of lncRNA DANCR by transfection of pcDNA3.1‑DANCR could suppress 786‑O and ACHN (RCC cell) proliferation, migration and invasion, and induce apoptosis compared with cells transfected with the pcDNA3.1 vector. The results revealed that DANCR functions as a tumor suppressor in RCC. In conclusion, the present study, to the best of our knowledge, was the first to reveal DANCR as a tumor suppressor. The results implicate DANCR as a potential biomarker of RCC, and further study will be focused on the clinical significance and signaling pathways of DANCR.
Collapse
Affiliation(s)
- Lu Jin
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Huifang Fu
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Shenzhen, Guangdong 518036, P.R. China
| | - Jing Quan
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Xiang Pan
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Tao He
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Jia Hu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Yifan Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Hang Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Yu Yang
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Jing Ye
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Shenzhen, Guangdong 518036, P.R. China
| | - Fangting Zhang
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Shenzhen, Guangdong 518036, P.R. China
| | - Liangchao Ni
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Shangqi Yang
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Yongqing Lai
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
354
|
Wang Y, Zhang Y, Yang T, Zhao W, Wang N, Li P, Zeng X, Zhang W. Long non-coding RNA MALAT1 for promoting metastasis and proliferation by acting as a ceRNA of miR-144-3p in osteosarcoma cells. Oncotarget 2017; 8:59417-59434. [PMID: 28938647 PMCID: PMC5601743 DOI: 10.18632/oncotarget.19727] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/29/2017] [Indexed: 12/30/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are involved in various biological processes and diseases including osteosarcoma. Long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is overly expressed in osteosarcoma. But the function and mechanism it works on in osteosarcoma proliferation and metastasis mediated by Rho associated coiled-coil containing protein kinase 1 (ROCK1) and Rho associated coiled-coil containing protein kinase 2 (ROCK2) remain unclear. In the present study, an elevated MALAT1 was found in osteosarcoma tissues and cell lines, and the elevated MALAT1 was correlated with a poor prognosis in osteosarcoma patients. The functional experiments show that a decreased MALAT1 could remarkably inhibit osteosarcoma cell metastasis and proliferation but induce cell cycle arrest, indicating that MALAT1 functioned as an oncogene in osteosarcoma. Furthermore, we confirmed that MALAT1 and ROCK1/ROCK2 which were targeted by microRNA-144-3p (miR-144-3p) shared the same miR-144-3p combining site. Furthermore, the constructed luciferase assay verified that MALAT1 was a target of miR-144-3p. Additionally, the results of a qRT-PCR demonstrated that MALAT1 and miR-144-3p repressed each other's expression in a reciprocal manner. Finally, we affirmed that an overexpression of MALAT1 inhibited ROCK1/ROCK2 expression and its mediated metastasis and proliferation by working as a competitive endogenous RNA (ceRNA) via miR-144-3p. In summary, the findings of this study based on the ceRNA theory, combining the research foundation of miR-144-3p, ROCK1 and ROCK2, taking MALAT1 as a new point of study, provided new insights into molecular level proliferation reversal and metastasis of osteosarcoma.
Collapse
Affiliation(s)
- Yong Wang
- The 4th Department of Orthopedic Surgery, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
| | - Yueyang Zhang
- Department of Pathology, Liaoning Cancer Hospital & Institute, Shenyang, P. R. China
| | - Tao Yang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, P. R. China
| | - Wei Zhao
- The 4th Department of Orthopedic Surgery, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
| | - Ningning Wang
- The 2nd Department of Cardiology, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
| | - Pengcheng Li
- The 4th Department of Orthopedic Surgery, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
| | - Xiandong Zeng
- Department of Surgical Oncology, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
| | - Weiguo Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, P. R. China
| |
Collapse
|
355
|
Grimaldi A, Zarone MR, Irace C, Zappavigna S, Lombardi A, Kawasaki H, Caraglia M, Misso G. Non-coding RNAs as a new dawn in tumor diagnosis. Semin Cell Dev Biol 2017; 78:37-50. [PMID: 28765094 DOI: 10.1016/j.semcdb.2017.07.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 12/12/2022]
Abstract
The current knowledge about non-coding RNAs (ncRNAs) as important regulators of gene expression in both physiological and pathological conditions, has been the main engine for the design of innovative platforms to finalize the pharmacological application of ncRNAs as either therapeutic tools or as molecular biomarkers in cancer. Biochemical alterations of cancer cells are, in fact, largely supported by ncRNA disregulation in the tumor site, which, in turn, reflects the cancer-associated specific modification of circulating ncRNA expression pattern. The aim of this review is to describe the state of the art of pre-clinical and clinical studies that analyze the involvement of miRNAs and lncRNAs in cancer-related processes, such as proliferation, invasion and metastases, giving emphasis to their functional role. A central node of our work has been also the examination of advantages and criticisms correlated with the clinical use of ncRNAs, taking into account the pressing need to refine the profiling methods aimed at identify novel diagnostic and prognostic markers and the request to optimize the delivery of such nucleic acids for a therapeutic use in an imminent future.
Collapse
Affiliation(s)
- Anna Grimaldi
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Mayra Rachele Zarone
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Carlo Irace
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Silvia Zappavigna
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Angela Lombardi
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Hiromichi Kawasaki
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy; Wakunaga Pharmaceutical Co. LTD, 4-5-36 Miyahara, Yodogawa-ku, Osaka 532-0003 Japan
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy.
| | - Gabriella Misso
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy.
| |
Collapse
|
356
|
Lu Q, Zhao N, Zha G, Wang H, Tong Q, Xin S. LncRNA HOXA11-AS Exerts Oncogenic Functions by Repressing p21 and miR-124 in Uveal Melanoma. DNA Cell Biol 2017; 36:837-844. [PMID: 28749709 DOI: 10.1089/dna.2017.3808] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have been reported to play vital roles in various human cancers. The aim of this study was to explore the critical role of lncRNA HOXA11-AS in uveal melanoma (UM) progression. Briefly, we found that HOXA11-AS is overexpressed in UM tissues and cells; HOXA11-AS could regulate UM cell growth, invasion, and apoptosis. Mechanistically, RNA immunoprecipitation demonstrated that HOXA11-AS could simultaneously interact with enhancer of zeste homolog 2 (EZH2) to suppress its target p21 protein expression. In addition, we demonstrated that HOXA11-AS functioned as a molecular sponge for miR-124, and overexpression of miR-124 attenuated the proliferation and invasion-promoting effect of HOXA11-AS. Collectively, our findings reveal an oncogenic role for HOXA11-AS in UM tumorigenesis.
Collapse
Affiliation(s)
- Qinkang Lu
- Ophthalmology Center, Yinzhou Hospital Affiliated to Medical School of Ningbo University , Ningbo, China
| | - Na Zhao
- Ophthalmology Center, Yinzhou Hospital Affiliated to Medical School of Ningbo University , Ningbo, China
| | - Guiping Zha
- Ophthalmology Center, Yinzhou Hospital Affiliated to Medical School of Ningbo University , Ningbo, China
| | - Huiyun Wang
- Ophthalmology Center, Yinzhou Hospital Affiliated to Medical School of Ningbo University , Ningbo, China
| | - Qihu Tong
- Ophthalmology Center, Yinzhou Hospital Affiliated to Medical School of Ningbo University , Ningbo, China
| | - Shuanghua Xin
- Ophthalmology Center, Yinzhou Hospital Affiliated to Medical School of Ningbo University , Ningbo, China
| |
Collapse
|
357
|
Cao Y, Shi H, Ren F, Jia Y, Zhang R. Long non-coding RNA CCAT1 promotes metastasis and poor prognosis in epithelial ovarian cancer. Exp Cell Res 2017; 359:185-194. [PMID: 28754469 DOI: 10.1016/j.yexcr.2017.07.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/11/2017] [Accepted: 07/24/2017] [Indexed: 01/17/2023]
Abstract
In this study, we reported that long non-coding RNA (lncRNA) CCAT1 was upregulated in epithelial ovarian cancer (EOC) tissues, and was associated with FIGO stage, histological grade, lymph node metastasis and poor survival of EOC patients. Multivariate Cox regression analysis showed that CCAT1 was an independent prognostic indicator. While CCAT1 downregulation inhibited EOC cell epithelial-mesenchymal transition (EMT), migration and invasion, CCAT1 upregulation promoted EOC cell EMT, migration and invasion. We further identified and confirmed that miR-152 and miR-130b were the targets of CCAT1, and CCAT1 functioned by targeting miR-152 and miR-130b. Subsequently, ADAM17 and WNT1, and STAT3 and ZEB1 were confirmed to be the targets of miR-152 and miR-130b, respectively, and could be regulated by CCAT1 in EOC cells. Knockdown of anyone of these four proteins inhibited EOC cell EMT, migration and invasion. Taken together, our study first revealed a critical role of CCAT1-miR-152/miR-130b-ADAM17/WNT1/STAT3/ZEB1 regulatory network in EOC cell metastasis. These findings provide great insights into EOC initiation and progression, and novel potential therapeutic targets and biomarkers for diagnosis and prognosis for EOC.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Gynaecology, the First Affiliated Hospital of Zhengzhou University, China
| | - Huirong Shi
- Department of Gynaecology, the First Affiliated Hospital of Zhengzhou University, China.
| | - Fang Ren
- Department of Gynaecology, the First Affiliated Hospital of Zhengzhou University, China
| | - Yanyan Jia
- Department of Gynaecology, the First Affiliated Hospital of Zhengzhou University, China
| | - Ruitao Zhang
- Department of Gynaecology, the First Affiliated Hospital of Zhengzhou University, China
| |
Collapse
|
358
|
Sha L, Huang L, Luo X, Bao J, Gao L, Pan Q, Guo M, Zheng F, Wang H. Long non-coding RNA LINC00261 inhibits cell growth and migration in endometriosis. J Obstet Gynaecol Res 2017; 43:1563-1569. [PMID: 28707780 DOI: 10.1111/jog.13427] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/17/2017] [Accepted: 05/14/2017] [Indexed: 01/22/2023]
Abstract
AIM A previous study reported that LINC00261 is significantly downregulated in human ectopic endometrial tissues. The present study aimed to explore whether LINC00261 is functional in endometriosis cell proliferation, apoptosis and migration. METHODS By transfecting human endometriosis cell line CRL-7566 with plasmids containing LINC00261, we successfully established the cell CRL-7566/LINC00261 with a high LINC00261 expression level. Cell-counting kit-8 and colony formation assays were conducted to evaluate the effect of LINC00261 on cell proliferation, and flow cytometry analysis and transwell migration assay were conducted to evaluate its effect on cell apoptosis and cell migration, respectively. RESULTS Cell-counting kit-8 and colony formation assays both indicated that LINC00261 could inhibit cell proliferation in CRL-7566. Flow cytometry analysis confirmed that LINC00261 mediated inhibition of cell proliferation, which might be a consequence of inducting apoptosis. Furthermore, transwell migration assay indicated that LINC00261 could inhibit cell migration in endometriosis. CONCLUSION LncRNA LINC00261 is capable of inhibiting cell growth and migration in endometriosis.
Collapse
Affiliation(s)
- Lixiao Sha
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, China
| | - Lingxiao Huang
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, China
| | - Xishao Luo
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiaping Bao
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lijun Gao
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qionghui Pan
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, China
| | - Min Guo
- Department of Obstetrics and Gynecology, Wenzhou People's Hospital, Wenzhou, China
| | - Feiyun Zheng
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hanchu Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
359
|
Fang J, Qiao F, Tu J, Xu J, Ding F, Liu Y, Akuo BA, Hu J, Shao S. High expression of long non-coding RNA NEAT1 indicates poor prognosis of human cancer. Oncotarget 2017; 8:45918-45927. [PMID: 28507281 PMCID: PMC5542237 DOI: 10.18632/oncotarget.17439] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 04/11/2017] [Indexed: 01/28/2023] Open
Abstract
The nuclear paraspeckle assembly transcript 1 (NEAT1) is a long non-coding RNA. Many studies have reported that NEAT1 plays critical oncogenic roles and facilitates tumorigenesis of various human cancers. High NEAT1 expression is associated with a poor prognosis in cancer patients. This meta-analysis was conducted to assess the association between NEAT1 levels and survival times of cancer patients. Overall survival (OS) was the primary endpoint. Thirteen publications with 1,496 cancer patients from 5 databases (PubMed, EMBASE, Cochrane Library, Wiley Online Library, and Medline) met the criteria for this meta-analysis. Results of the analysis showed that NEAT1 expression in human cancer was significantly associated with OS (hazard ratio [HR]=1.53, 95% confidence interval [CI]: 1.39-1.68), including digestive system tumor (HR=1.54, 95% CI: 1.37-1.73) and respiratory carcinomas (HR=1.44, 95% CI: 1.11-1.85). The results also indicated that NEAT1 expression was highly associated with tumor size (>3 cm vs. ≤3 cm; odds ratio [OR]=2.51, 95% CI: 1.27-4.99; p=0.009), TNM stage (III+IV vs. I+II; OR=4.17, 95% CI: 2.42-7.18; p=0.00001), and distant metastasis (OR=2.73, 95% CI: 1.28-5.79; p=0.01). However, there was no significant association with differentiation (poor vs. well + moderate, OR=1.45, 95% CI: 0.72-2.91) and lymph node metastasis (OR=1.39, 95% CI: 0.54-3.60). This meta-analysis showed that NEAT1 expression may be a useful biomarker for predicting a poor prognosis in patients with cancer.
Collapse
Affiliation(s)
- Jian Fang
- Department of Pathogenic Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Fuhao Qiao
- Department of Pathogenic Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jingjing Tu
- Department of Pathogenic Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jinfeng Xu
- Department of Pathogenic Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Fangfang Ding
- Department of Pathogenic Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yun Liu
- Department of Pathogenic Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Bufugdi Andreas Akuo
- Department of Pathogenic Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jianpeng Hu
- Department of Urinary Surgery, Zhenjiang First People's Hospital, Zhenjiang, Jiangsu 212013, China
| | - Shihe Shao
- Department of Pathogenic Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| |
Collapse
|
360
|
Lu W, Zhang H, Niu Y, Wu Y, Sun W, Li H, Kong J, Ding K, Shen HM, Wu H, Xia D, Wu Y. Long non-coding RNA linc00673 regulated non-small cell lung cancer proliferation, migration, invasion and epithelial mesenchymal transition by sponging miR-150-5p. Mol Cancer 2017; 16:118. [PMID: 28697764 PMCID: PMC5504775 DOI: 10.1186/s12943-017-0685-9] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 06/22/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The function of a new long non-coding RNA linc00673 remains unclear. While identified as an oncogenic player in non-small cell lung cancer (NSCLC), linc00673 was found to be anti-oncogenic in pancreatic ductal adenocarcinoma (PDAC). However whether linc00673 regulated malignancy and epithelial mesenchymal transition (EMT) has not been characterized. METHODS Cell proliferation was assessed using CCK-8 and EdU assays, and cell migration and invasion were assessed using scratch assays and transwell invasion assays. Epithelial mesenchymal transition was examined using western blot, qRT-PCR and immunofluorescence staining. Interaction between miRNA and linc00673 was determined using luciferase reporter assays. In vivo experiments were performed to assess tumor formation. In addition, the expression data of NSCLC specimens of TCGA and patient survival data were utilized to explore the prognostic significance of linc00673. RESULTS In the present study, we found high linc00673 expression was associated with poor prognosis of NSCLC patients. In vitro experiments showed linc00673 knockdown reversed TGF-β induced EMT, and miR-150-5p was predicted to target linc00673 through bioinformatics tools. Overexpression of miR-150-5p suppressed lin00673's expression while inhibition of miR-150-5p led to significant upregulation of lin00673, suggesting that linc00673 could be negatively regulated by miR-150-5p, which was further confirmed by the inverse correlation between linc00673 and miR-150-5p in NSCLC patients' specimen. Furthermore, we proved that miR-150-5p could directly target linc00673 through luciferase assay, so linc00673 could sponge miR-150-5p and modulate the expression of a key EMT regulator ZEB1 indirectly. In addition, miR-150-5p inhibition abrogated linc00673 silence mediated proliferation, migration, invasion and EMT suppressing effect. Moreover, the inhibition of linc00673 significantly attenuated the tumorigenesis ability of A549 cells in vivo. CONCLUSIONS We validated linc00673 as a novel oncogenic lncRNA and demonstrated the molecular mechanism by which it promotes NSCLC, which will advance our understanding of its clinical significance.
Collapse
Affiliation(s)
- Wei Lu
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yuhangtang Road, Hangzhou, People’s Republic of China
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Honghe Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yuequn Niu
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yuhangtang Road, Hangzhou, People’s Republic of China
| | - Yongfeng Wu
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yuhangtang Road, Hangzhou, People’s Republic of China
| | - Wenjie Sun
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Hongyi Li
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yuhangtang Road, Hangzhou, People’s Republic of China
| | - Jianlu Kong
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Kefeng Ding
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Han Wu
- Department of Ophthalmology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Dajing Xia
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yuhangtang Road, Hangzhou, People’s Republic of China
| | - Yihua Wu
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yuhangtang Road, Hangzhou, People’s Republic of China
| |
Collapse
|
361
|
Zhang J, Cao Z, Ding X, Wei X, Zhang X, Hou J, Ouyang J. The lncRNA XIST regulates the tumorigenicity of renal cell carcinoma cells via the miR-302c/SDC1 axis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:7481-7491. [PMID: 31966592 PMCID: PMC6965219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/03/2017] [Indexed: 06/10/2023]
Abstract
Long non-coding RNAs (LncRNAs) are thought to be involved in several biological processes in carcinomas. The aim of this study is to evaluate the roles of lncRNA-XIST in the tumorigenicity of renal cell carcinoma (RCC) cells via the miR-302c/SDC1 axis. In this study, the expression levels of miR-302c and XIST in RCC tissues and cells were analyzed by qRT-PCR. Cell proliferation was measured using MTT and colony formation assays, and cell apoptosis was detected using flow cytometry. The interaction between XIST and miR-302c was analyzed using a luciferase reporter gene assay. RCC tissues and cells exhibited decreased miR-302c expression and increased lncRNA-XIST expression. Furthermore, XIST negatively regulated miR-302c by directly binding regulatory sites in RCC cells. In addition, XIST silencing with siRNAs significantly inhibited the proliferation and promoted the apoptosis of 786-O and Caki-1 cells. Knockdown of Syndecan-1 (SDC1), a miR-302c target gene, yielded similar results as XIST silencing. In summary, XIST regulated the development and progression of RCC by inhibiting the miR302c/SDC1 axis.
Collapse
Affiliation(s)
- Jianglei Zhang
- Department of Urology, The First Affiliated Hospital of Suzhou University Suzhou, China
| | - Zhijun Cao
- Department of Urology, The First Affiliated Hospital of Suzhou University Suzhou, China
| | - Xiang Ding
- Department of Urology, The First Affiliated Hospital of Suzhou University Suzhou, China
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Suzhou University Suzhou, China
| | - Xuefeng Zhang
- Department of Urology, The First Affiliated Hospital of Suzhou University Suzhou, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Suzhou University Suzhou, China
| | - Jun Ouyang
- Department of Urology, The First Affiliated Hospital of Suzhou University Suzhou, China
| |
Collapse
|
362
|
Grelet S, McShane A, Geslain R, Howe PH. Pleiotropic Roles of Non-Coding RNAs in TGF-β-Mediated Epithelial-Mesenchymal Transition and Their Functions in Tumor Progression. Cancers (Basel) 2017; 9:cancers9070075. [PMID: 28671581 PMCID: PMC5532611 DOI: 10.3390/cancers9070075] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/21/2017] [Accepted: 06/30/2017] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a spatially- and temporally-regulated process involved in physiological and pathological transformations, such as embryonic development and tumor progression. While the role of TGF-β as an EMT-inducer has been extensively documented, the molecular mechanisms regulating this transition and their implications in tumor metastasis are still subjects of intensive debates and investigations. TGF-β regulates EMT through both transcriptional and post-transcriptional mechanisms, and recent advances underline the critical roles of non-coding RNAs in these processes. Although microRNAs and lncRNAs have been clearly identified as effectors of TGF-β-mediated EMT, the contributions of other atypical non-coding RNA species, such as piRNAs, snRNAs, snoRNAs, circRNAs, and even housekeeping tRNAs, have only been suggested and remain largely elusive. This review discusses the current literature including the most recent reports emphasizing the regulatory functions of non-coding RNA in TGF-β-mediated EMT, provides original experimental evidence, and advocates in general for a broader approach in the quest of new regulatory RNAs.
Collapse
Affiliation(s)
- Simon Grelet
- Department of Biochemistry and Molecular Biology, MUSC, Charleston, SC 29425, USA.
| | - Ariel McShane
- Laboratory of tRNA Biology, Department of Biology, College of Charleston, Charleston, SC 29424, USA.
| | - Renaud Geslain
- Laboratory of tRNA Biology, Department of Biology, College of Charleston, Charleston, SC 29424, USA.
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, MUSC, Charleston, SC 29425, USA.
| |
Collapse
|
363
|
Liu X, Dai C, Jia G, Xu S, Fu Z, Xu J, Li Q, Ruan H, Xu P. Microarray analysis reveals differentially expressed lncRNAs in benign epithelial ovarian cysts and normal ovaries. Oncol Rep 2017; 38:799-808. [PMID: 28656240 PMCID: PMC5562051 DOI: 10.3892/or.2017.5741] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/09/2017] [Indexed: 12/27/2022] Open
Abstract
Recent studies suggest that long non-coding RNAs (lncRNAs) play crucial roles in many types of human malignant cancers. However, the function of lncRNAs in benign tumors remains poorly understood. In the present study, to explored the potential roles of lncRNAs in benign epithelial ovarian cysts (BEOCs) which commonly occur in young women and possess malignant potential, we described the expression profile of the lncRNAs between BEOC and normal ovarian tissues using lncRNA microarray techniques. The results showed that 1,325 transcripts of lncRNAs (1,014 upregulated and 311 downregulated) were differentially expressed in the BEOCs compared with the normal controls [absolute fold-change ≥2, false discovery rate (FDR) <0.05]. We also conducted quantitative real-time PCR (qPCR) to confirm the microarray data. The results of qPCR revealed that the expression trend of 6 randomly selected lncRNAs was consistent with the microarray data. Furthermore, candidate lncRNAs were characterized by pathway analysis and Gene Ontology (GO). The present study is the first to demonstrate different expression profiles of lncRNAs between BEOCs and normal ovarian tissues. These lncRNAs may play a crucial role in the pathological process of BEOCs.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Nanjing Maternal and Child Health Institute, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Chencheng Dai
- Nanjing Maternal and Child Health Institute, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Genmei Jia
- Nanjing Maternal and Child Health Institute, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Sujuan Xu
- Department of Clinical Laboratory, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Ziyi Fu
- Nanjing Maternal and Child Health Institute, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Juan Xu
- Nanjing Maternal and Child Health Institute, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Qing Li
- Department of Pathology, Shanghai Pudong New Area People's Hospital, Shanghai 201040, P.R. China
| | - Hongjie Ruan
- Department of Gynecology, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Pengfei Xu
- Nanjing Maternal and Child Health Institute, Nanjing Maternal and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| |
Collapse
|
364
|
Wang X, Lv G, Li J, Wang B, Zhang Q, Lu C. LncRNA-RP11-296A18.3/miR-138/HIF1A Pathway Regulates the Proliferation ECM Synthesis of Human Nucleus Pulposus Cells (HNPCs). J Cell Biochem 2017; 118:4862-4871. [PMID: 28543639 DOI: 10.1002/jcb.26166] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/23/2017] [Indexed: 12/18/2022]
Abstract
During the process of Intervertebral disc degeneration (IDD), nucleus pulposus apoptosis increases, extracellular matrix (ECM) alters and/or degrades, abnormal proliferation of cells forms cell clusters, and the expression of various inflammatory factors increases. Thus, regulation of human nucleus pulposus cell (HNPC) proliferation and ECM synthesis present promising strategies for IDD treatment. Accumulating evidence indicates that non-coding RNAs are involved in various cellular processes, including cell proliferation, differentiation, apoptosis, and metastasis. High expression of long non-coding RNA (lncRNA) RP11-296A18.3, as well as a low expression of miR-138 during the IDD process has been reported; yet their functional roles in HNPC proliferation and ECM synthesis still remain unclear. MTT and BrdU assays showed that knockdown of RP11-296A18.3 inhibited the proliferation of HNPC. The ECM marker, MMP-13 and Collagen I expressions were also reduced. Bioinformatics target prediction, qPCR, and luciferase assays identified LncRNA-RP11-296A18.3 interacted with miR-138. Moreover, RP11-296A18.3 regulates HNPC proliferation and ECM synthesis through miR-138. As the target gene of miR-138, hypoxia-inducible factor 1-alpha (HIF1A) was closely associated with cell proliferation which was also regulated by RP11-296A18.3 via miR-138. Immunochemistry and qPCR results showed that miR-138 expression was inversely correlated to RP11-296A18.3 and HIF1A in IDD tissues, respectively; RP11-296A18.3 was positively correlated to HIF1A. We revealed that RP11-296A18.3 promote HIF1A expression through sponging miR-138, thus to promote HNPC proliferation and ECM synthesis. Targeting RP11-296A18.3 to rescue miR-138 expression in HNPCs and IDD tissues presents a promising strategy for IDD improvement. J. Cell. Biochem. 118: 4862-4871, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaobin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Guohua Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Bing Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qianshi Zhang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Chang Lu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
365
|
LncRNA-mediated regulation of cell signaling in cancer. Oncogene 2017; 36:5661-5667. [PMID: 28604750 PMCID: PMC6450570 DOI: 10.1038/onc.2017.184] [Citation(s) in RCA: 1249] [Impact Index Per Article: 156.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/06/2017] [Accepted: 05/08/2017] [Indexed: 12/13/2022]
Abstract
To date, a large number of long non-coding RNAs (lncRNAs) have been recently discovered through functional genomics studies. Importantly, lncRNAs have been shown, in many cases, to function as master regulators for gene expression and thus, they can play a critical role in various biological functions and disease processes including cancer. Although the lncRNA-mediated gene expression involves various mechanisms, such as regulation of transcription, translation, protein modification, and the formation of RNA-protein or protein-protein complexes, in this review we discuss the latest developments primarily in important cell signaling pathways regulated by lncRNAs in cancer.
Collapse
|
366
|
Cui B, Li B, Liu Q, Cui Y. lncRNA CCAT1 Promotes Glioma Tumorigenesis by Sponging miR‐181b. J Cell Biochem 2017; 118:4548-4557. [DOI: 10.1002/jcb.26116] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/04/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Bingzhou Cui
- Department of NeurosurgeryThe People's Hospital of ZhengzhouZhengzhou450002 HenanChina
| | - Baoshan Li
- Department of NeurosurgeryThe Third People's Hospital of QingdaoQingdao266041ShandongChina
| | - Qi Liu
- Department of NeurosurgeryBrain HospitalPeople's Hospital of WeifangWeifang261021ShandongChina
| | - Youqiang Cui
- Department of NeurosurgeryQianfoshan Hospital Affiliated to Shandong UniversityJinan250014ShandongChina
| |
Collapse
|
367
|
EZH2 in Cancer Progression and Potential Application in Cancer Therapy: A Friend or Foe? Int J Mol Sci 2017; 18:ijms18061172. [PMID: 28561778 PMCID: PMC5485996 DOI: 10.3390/ijms18061172] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 05/24/2017] [Accepted: 05/27/2017] [Indexed: 01/26/2023] Open
Abstract
Enhancer of zeste homolog 2 (EZH2), a histone methyltransferase, catalyzes tri-methylation of histone H3 at Lys 27 (H3K27me3) to regulate gene expression through epigenetic machinery. EZH2 functions as a double-facet molecule in regulation of gene expression via repression or activation mechanisms, depending on the different cellular contexts. EZH2 interacts with both histone and non-histone proteins to modulate diverse physiological functions including cancer progression and malignancy. In this review article, we focused on the updated information regarding microRNAs (miRNAs) and long non coding RNAs (lncRNAs) in regulation of EZH2, the oncogenic and tumor suppressive roles of EZH2 in cancer progression and malignancy, as well as current pre-clinical and clinical trials of EZH2 inhibitors.
Collapse
|
368
|
Xu Y, Tong Y, Zhu J, Lei Z, Wan L, Zhu X, Ye F, Xie L. An increase in long non-coding RNA PANDAR is associated with poor prognosis in clear cell renal cell carcinoma. BMC Cancer 2017; 17:373. [PMID: 28545465 PMCID: PMC5445460 DOI: 10.1186/s12885-017-3339-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/10/2017] [Indexed: 12/31/2022] Open
Abstract
Background Nearly 30% of clear cell renal cell carcinoma (ccRCC) patients present with metastasis at the time of diagnosis, and the prognosis for these patients is poor. Therefore, novel potential prognostic biomarkers and therapeutic targets for ccRCC could be helpful. Emerging evidence indicates that lncRNAs play important roles in cancer tumorigenesis and could be used as potential biomarkers or therapeutic targets. PANDAR (promoter of CDKN1A antisense DNA damage activated RNA) is a relatively novel lncRNA that plays an important role in the development of multiple cancers. However, the clinical significance and molecular mechanism of PANDAR in ccRCC are still elusive. In the present study, we attempted to elucidate the role of PANDAR in ccRCC. Methods The relative expression level of lncRNA PANDAR was quantified by real-time qPCR in 62 paired ccRCC tissues and in renal cancer cell lines, and its association with overall survival was assessed by statistical analysis. The biological functions of lncRNA PANDAR on ccRCC cells were determined both in vitro and in vivo. Results PANDAR expression was significantly upregulated in tumor tissues and cell lines compared with normal counterparts. Moreover, PANDAR served as an independent predictor of overall survival, and increased PANDAR expression was positively correlated with an advanced TNM stage. Further experiments demonstrated that PANDAR silencing can significantly inhibit cell proliferation and invasion, induce cell cycle arrest in the G1 phase and significantly promote apoptosis in 7860 and Caki-1 cell lines. In addition, in vivo experiments confirmed that downregulation of PANDAR inhibited the tumorigenic ability of 7860 cells in nude mice. Silencing of PANDAR also inhibited the expression of Bcl-2 and Mcl-1 and upregulated the expression of Bax in vivo. Conclusions Our results suggest that PANDAR is involved in ccRCC progression and may serve as a potential prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yi Xu
- Department of Urology, Quzhou Hospital, Zhejiang University, Quzhou City, 324000, China
| | - Yanyue Tong
- Department of Urology, Quzhou Hospital, Zhejiang University, Quzhou City, 324000, China
| | - Jianyong Zhu
- Department of Urology, Quzhou Hospital, Zhejiang University, Quzhou City, 324000, China
| | - Zhangming Lei
- Department of Urology, Quzhou Hospital, Zhejiang University, Quzhou City, 324000, China
| | - Lijun Wan
- Department of Urology, Quzhou Hospital, Zhejiang University, Quzhou City, 324000, China
| | - Xiuwen Zhu
- Department of Urology, Quzhou Hospital, Zhejiang University, Quzhou City, 324000, China
| | - Feng Ye
- Department of Urology, Quzhou Hospital, Zhejiang University, Quzhou City, 324000, China
| | - Liping Xie
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou City, 310003, China.
| |
Collapse
|
369
|
Hong Q, Li O, Zheng W, Xiao WZ, Zhang L, Wu D, Cai GY, He JC, Chen XM. LncRNA HOTAIR regulates HIF-1α/AXL signaling through inhibition of miR-217 in renal cell carcinoma. Cell Death Dis 2017; 8:e2772. [PMID: 28492542 PMCID: PMC5520706 DOI: 10.1038/cddis.2017.181] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 02/07/2023]
Abstract
Long non-coding RNA HOTAIR was regarded as an oncogene in multiple cancers. Previous studies have shown that HOTAIR is involved in the proliferation and tumorigenesis of renal carcinoma cells, while microRNA (miR)-217 functions as a tumor suppressor in renal cell carcinoma (Rcc). However, the underlying molecular mechanism of HOTAIR in Rcc, especially in association with miR-217, has not been studied. In this study, we first demonstrated that HOTAIR expression was upregulated, which was correlated with tumor progression, and miR-217 downregulated in Rcc tissues and cells. Importantly, HOTAIR expression was negatively correlated with miR-217 expression in Rcc tissues. Gain- and loss-of-function of HOTAIR revealed that HOTAIR functioned as a ceRNA for miR-217 to facilitate HIF-1α expression and then upregulated AXL level promoting Rcc proliferation, migration, and EMT process, and inhibiting apoptosis. Furthermore, HOTAIR knockdown suppressed tumor growth and reduced the expression of proliferation antigen ki-67, HIF-1α, and AXL, but upregulated the expression of miR-217 in vivo. Finally, with AXL inhibitor BGB324, we confirmed that HOTAIR promoted Rcc activity through AXL signaling both in vitro and in vivo. In conclusion, these results suggest that HOTAIR promotes Rcc tumorigenesis via miR-217/HIF-1α/AXL signaling, which may provide a new target for the diagnosis and therapy of Rcc disease.
Collapse
MESH Headings
- Benzocycloheptenes/pharmacology
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Male
- MicroRNAs/biosynthesis
- MicroRNAs/genetics
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Signal Transduction
- Triazoles/pharmacology
- Axl Receptor Tyrosine Kinase
Collapse
Affiliation(s)
- Quan Hong
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Haidian District, Beijing 100853, China
| | - Ou Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Haidian District, Beijing 100853, China
| | - Wei Zheng
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Haidian District, Beijing 100853, China
| | - Wen-zhen Xiao
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lu Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Di Wu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Haidian District, Beijing 100853, China
| | - Guang-yan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Haidian District, Beijing 100853, China
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiang-mei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Fuxing Road 28, Haidian District, Beijing 100853, China
| |
Collapse
|
370
|
Huang FT, Chen WY, Gu ZQ, Zhuang YY, Li CQ, Wang LY, Peng JF, Zhu Z, Luo X, Li YH, Yao HR, Zhang SN. The novel long intergenic noncoding RNA UCC promotes colorectal cancer progression by sponging miR-143. Cell Death Dis 2017; 8:e2778. [PMID: 28492554 PMCID: PMC5520712 DOI: 10.1038/cddis.2017.191] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 03/21/2017] [Accepted: 03/28/2017] [Indexed: 01/12/2023]
Abstract
The human genome contains thousands of long intergenic noncoding RNAs (lincRNAs). However, the functional roles of these transcripts and the mechanisms responsible for their deregulation in colorectal cancer (CRC) remain elusive. A novel lincRNA termed upregulated in CRC (UCC) was found to be highly expressed in human CRC tissues and cell lines. UCC levels correlated with lymph node metastasis, Dukes' stage, and patient outcomes. In SW480 and SW620 cells, knockdown of UCC inhibited proliferation, invasion, and cell cycle progression and induced apoptosis in vitro. Xenograft tumors grown from UCC-silenced SW620 cells had smaller mean volumes and formed more slowly than xenograft tumors grown from control cells. Inversely, overexpression of UCC in HCT116 promoted cell growth and invasion in vitro. Bioinformatics analysis, dual-luciferase reporter assays, and RNA immunoprecipitation assays showed that miR-143 can interact with UCC, and we found that UCC expression inversely correlates with miR-143 expression in CRC specimens. Moreover, mechanistic investigations showed that UCC may act as an endogenous sponge by competing for miR-143, thereby regulating the targets of this miRNA. Our results suggest that UCC and miR-143 may be promising molecular targets for CRC therapy.
Collapse
Affiliation(s)
- Feng-Ting Huang
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wen-Ying Chen
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhi-Qiang Gu
- Department of Gastroenterology, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Yan-Yan Zhuang
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Chu-Qiang Li
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ling-Yun Wang
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Juan-Fei Peng
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhe Zhu
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, USA
| | - Xin Luo
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuan-Hua Li
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - He-Rui Yao
- Breast Tumor Center and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shi-Neng Zhang
- Department of Gastroenterology and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
371
|
LncRNA MALAT1 sponges miR-204 to promote osteoblast differentiation of human aortic valve interstitial cells through up-regulating Smad4. Int J Cardiol 2017; 243:404-412. [PMID: 28522163 DOI: 10.1016/j.ijcard.2017.05.037] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/08/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Emerging evidences have indicated that long non-coding RNAs (lncRNAs) play vital roles in cardiovascular physiology and pathology. The lncRNA MALAT1, a highly abundant and conserved imprinted gene, has been implicated in many cardiovascular diseases. However, the function of MALAT1 in calcific aortic valve disease (CAVD) remains unknown. This study sought to document the function and underlying mechanism of MALAT1 in regulating CAVD. METHODS Protein level was determined by immunoblotting and immunofluorescence staining. MALAT1, miR-204 and mRNA expressions were detected by qRT-PCR. Mineralized bone matrix formation was assessed by Alizarin Red staining. The interaction between MALAT1 and miR-204 was studied using luciferase reporter assay, RNA pull-down assay and RNA-binding protein immunoprecipitation assay. RESULTS Ectopic expression of MALAT1 was observed in calcific valves and after osteogenic induction in human aortic valve interstitial cells (VICs). In vitro experiments revealed that MALAT1 acted as a positive regulator of osteogenic differentiation by repressing miR-204 expression and activity and thereby promoting expression of osteoblast-specific markers, including alkaline phosphatase, mineralized bone matrix formation and osteocalcin. Mechanistically, we identified Smad4 as a direct target of miR-204. Importantly, MALAT1 could directly interact with miR-204 and overexpression of miR-204 efficiently reversed the upregulation of Smad4 induced by MALAT1. Thus, MALAT1 positively regulated the expression of Smad4 through sponging miR-204, and promoted osteogenic differentiation of VICs. CONCLUSIONS Our study provides novel mechanistic insights into a critical role for lncRNA MALAT1 as a miRNA sponge in CAVD and sheds new light on lncRNA-directed diagnostics and therapeutics in CAVD.
Collapse
|
372
|
Wei W, Liu Y, Lu Y, Yang B, Tang L. LncRNA XIST Promotes Pancreatic Cancer Proliferation Through miR-133a/EGFR. J Cell Biochem 2017; 118:3349-3358. [PMID: 28295543 DOI: 10.1002/jcb.25988] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 03/09/2017] [Indexed: 12/17/2022]
Abstract
According to recent studies, long non-coding RNA X-inactive specific transcript (XIST) is involved in the development and progression of many malignant tumors including pancreatic cancer. We validated the detailed role of XIST in human pancreatic cancer (PC) cell lines and PC tissues so as to determine its exact function and the mechanism by which it affected PC proliferation. In our research, lncRNA-XIST was specifically upregulated in PC tissues and cell lines, and high XIST expression in PC was related to poorer prognosis (larger tumor size, perineural invasion, lymph node micrometastases, and shorter overall survival). XIST augmented PC cell proliferation. Recently, the interaction between lncRNA and miRNA has been frequently reported to play major role in several biological processes. In the present study, XIST and miR-133a reciprocally inhibited each other in PC cells. Exogenous miR-133a expression significantly inhibited PC cell proliferation. Moreover, as exhibited by luciferase reporter gene assays, miR-133a bound to XIST and the 3'UTR of EGFR by direct targeting. In PC tissues, miR-133a expression was down-regulated and EGFR expression was up-regulated; miR-133a was inversely correlated with EGFR and XIST, respectively; XIST was positively correlated with EGFR. Taken together, these findings will shed light on the role and mechanism of XIST/miR-133a/EGFR in regulating PC cells proliferation. XIST may serve as a potential therapeutic target in PC in the future. J. Cell. Biochem. 118: 3349-3358, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wei Wei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu Liu
- Department of Pathology, Hunan Provincial People's Hospital, Changsha, 410005, China
| | - Yebin Lu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Bo Yang
- Department of General Surgery, The Fourth Hospital of Changde City, Changde, 415000, China
| | - Ling Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| |
Collapse
|
373
|
Zhang H, Wang G, Yin R, Qiu M, Xu L. [Comprehensive Identification of MicroRNAs Regulated by Long Non-coding RNA MALAT1]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 19:247-51. [PMID: 27215451 PMCID: PMC5973056 DOI: 10.3779/j.issn.1009-3419.2016.05.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Long non-coding RNA (lncRNA) plays important regulatory roles in the development and invasion of various cancers. The aim of current study is to comprehensively identify microRNAs (miRNA) regulated by lncRNA MALAT1 via experimental and bioinformatics methods. METHODS Antisense oligonucleotides (ASO) specifically targeting MALAT1 were designed and synthesized. After knockdown of MALAT1 by ASO in A549 cells, miRNA expression changes were profiled by TqaMan Low Density Array (TLDA). Gene set enrichment analysis (GSEA) was used to search enriched miRNAs among differentially expressed genes after knockdown of MALAT1. RESULTS After efficient knockdown of MALAT1 by ASO, 153 miRNAs were differentially expressed, 131 up-regulated and 22 down-regulated. Among the 458 differentially expressed genes after MALAT1 silence, GSEA results revealed lots of enriched miRNAs. There were 28 overlapped miRNAs between TLDA and GSEA results, suggesting these 28 miRNAs are regulated by MALAT1. CONCLUSIONS This study comprehensively identified MALAT1 regulated miRNAs, providing resources for further research.
Collapse
Affiliation(s)
- Haitian Zhang
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou 221004, China
| | - Guoxiang Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Rong Yin
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing 210009, China
| | - Mantang Qiu
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing 210009, China
| | - Lin Xu
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing 210009, China
| |
Collapse
|
374
|
Nadal C, Winder T, Gerger A, Tougeron D. Future perspectives of circulating tumor DNA in colorectal cancer. Tumour Biol 2017; 39:1010428317705749. [PMID: 28488528 DOI: 10.1177/1010428317701632] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Tumor biopsy is currently the gold standard for diagnosis and in determining cell signaling pathways involved in the development of treatment resistance. However, there are major challenges with this technique, including the need for serial sampling to monitor treatment resistance, which is invasive and also has the potential for selection bias due to intra-tumoral and inter-tumoral heterogeneity. These challenges highlight the need for more effective methods for obtaining Tumor samples. Liquid biopsy analyzes genetic material or tumor cells shed into the blood from the primary tumor and metastatic sites and consequently provides a comprehensive, real-time picture of the tumor burden in an individual patient. Indeed, liquid biopsy has the potential to revolutionize cancer management. Here, we review recent studies on the potential clinical applications of liquid biopsy using circulating tumor DNA in colorectal cancer, including screening, diagnosis, detection of minimal residual disease after surgery, detection of recurrence, prognosis, predicting treatment response, monitoring tumor burden or response during treatment, and tracking resistance. We also discuss recent data demonstrating the utility of detecting KRAS-mutated circulating tumor DNA, both at diagnosis to determine an appropriate treatment strategy and during anti-epidermal growth factor receptor therapy to predict treatment resistance. The future integration of liquid biopsy into clinical practice is discussed, together with alternative approaches and key questions that need to be answered in future clinical studies before this technology can be implemented and used routinely.
Collapse
Affiliation(s)
- C Nadal
- 1 Department of Medical Oncology, Institut Clínic de Malalties Hemato-Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain
- 2 Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd), Madrid, Spain
- 3 August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - T Winder
- 4 Department of Oncology, University Hospital Zurich, Zurich, Switzerland
| | - A Gerger
- 5 Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- 6 Center for Biomarker Research in Medicine, Graz, Austria
| | - David Tougeron
- 7 Laboratoire Inflammation, Tissus Epithéliaux et Cytokines (LITEC)-EA 4331, Université de Poitiers, Poitiers, France
- 8 Gastroenterology Department, Poitiers University Hospital, Poitiers, France
- 9 Department of Oncology, CHU de Poitiers, Poitiers, France
| |
Collapse
|
375
|
Shi D, Qu Q, Chang Q, Wang Y, Gui Y, Dong D. A five-long non-coding RNA signature to improve prognosis prediction of clear cell renal cell carcinoma. Oncotarget 2017; 8:58699-58708. [PMID: 28938589 PMCID: PMC5601685 DOI: 10.18632/oncotarget.17506] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/22/2017] [Indexed: 12/17/2022] Open
Abstract
Recent works have reported that long non-coding RNAs (lncRNAs) play critical roles in tumorigenesis and prognosis of cancers, suggesting the potential utility of lncRNAs as cancer prognostic markers. However, lncRNA signatures in predicting the survival of patients with clear cell renal cell carcinoma (ccRCC) remain unknown. In this study, we attempted to identify lncRNA signatures and their prognostic values in ccRCC. Using lncRNA expression profiling data in 440 ccRCC tumors from The Cancer Genome Atlas (TCGA) data, a five-lncRNA signature (AC069513.4, AC003092.1, CTC-205M6.2, RP11-507K2.3, U91328.21) has been identified to be significantly associated with ccRCC patients’ overall survival in both training set and testing set. Based on the lncRNA signature, ccRCC patients could be divided into high-risk and low-risk group with significantly different survival rate. Further multivariable Cox regression analysis suggested that the prognostic value of this signature was independent of clinical factors. Functional enrichment analyses showed the potential functional roles of the five prognostic lncRNAs in ccRCC oncogenesis. These results indicated that this five-lncRNA signature could be used as an independent prognostic biomarker in the prediction of ccRCC patients’ survival.
Collapse
Affiliation(s)
- Da Shi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Qinghua Qu
- Department of Urology, Pudong People's Hospital, Shanghai, China
| | - Qimeng Chang
- Department of General Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Yilin Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yaping Gui
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong Dong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
376
|
Heery R, Finn SP, Cuffe S, Gray SG. Long Non-Coding RNAs: Key Regulators of Epithelial-Mesenchymal Transition, Tumour Drug Resistance and Cancer Stem Cells. Cancers (Basel) 2017; 9:cancers9040038. [PMID: 28430163 PMCID: PMC5406713 DOI: 10.3390/cancers9040038] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/14/2017] [Accepted: 04/18/2017] [Indexed: 02/07/2023] Open
Abstract
Epithelial mesenchymal transition (EMT), the adoption by epithelial cells of a mesenchymal-like phenotype, is a process co-opted by carcinoma cells in order to initiate invasion and metastasis. In addition, it is becoming clear that is instrumental to both the development of drug resistance by tumour cells and in the generation and maintenance of cancer stem cells. EMT is thus a pivotal process during tumour progression and poses a major barrier to the successful treatment of cancer. Non-coding RNAs (ncRNA) often utilize epigenetic programs to regulate both gene expression and chromatin structure. One type of ncRNA, called long non-coding RNAs (lncRNAs), has become increasingly recognized as being both highly dysregulated in cancer and to play a variety of different roles in tumourigenesis. Indeed, over the last few years, lncRNAs have rapidly emerged as key regulators of EMT in cancer. In this review, we discuss the lncRNAs that have been associated with the EMT process in cancer and the variety of molecular mechanisms and signalling pathways through which they regulate EMT, and finally discuss how these EMT-regulating lncRNAs impact on both anti-cancer drug resistance and the cancer stem cell phenotype.
Collapse
Affiliation(s)
- Richard Heery
- Thoracic Oncology Research Group, Rm 2.09, Trinity Translational Medical Institute, St. James's Hospital, Dublin D08 W9RT, Ireland.
- Masters in Translational Oncology Program, Department of Surgery, Trinity College Dublin, Trinity Translational Medical Institute, St. James's Hospital, Dublin D08 W9RT, Ireland.
| | - Stephen P Finn
- Department of Histopathology & Morbid Anatomy, Trinity College Dublin, Dublin D08 RX0X, Ireland.
| | - Sinead Cuffe
- HOPE Directorate, St. James's Hospital, Dublin D08 RT2X, Ireland.
| | - Steven G Gray
- Thoracic Oncology Research Group, Rm 2.09, Trinity Translational Medical Institute, St. James's Hospital, Dublin D08 W9RT, Ireland.
- HOPE Directorate, St. James's Hospital, Dublin D08 RT2X, Ireland.
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin D02 R590, Ireland.
- Labmed Directorate, St. James's Hospital, Dublin D08 K0Y5, Ireland.
| |
Collapse
|
377
|
Liu W, Zhang Q, Zhang J, Pan W, Zhao J, Xu Y. Long non-coding RNA MALAT1 contributes to cell apoptosis by sponging miR-124 in Parkinson disease. Cell Biosci 2017; 7:19. [PMID: 28439401 PMCID: PMC5401610 DOI: 10.1186/s13578-017-0147-5] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/19/2017] [Indexed: 12/31/2022] Open
Abstract
Background Parkinson disease (PD) is the most common movement disturbance characterized by the loss of dopaminergic (DA) neurons in midbrain. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is aberrantly expressed in neurons and is involved in the dendritic and synapse development. However, the role of MALAT1 and its underlying mechanism in PD remain to be defined. Methods The expressions of MALAT1 and miR-124 were evaluated by qRT-PCR. N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice and SH-SY5Y cells subjected to N-methyl-4-phenylpyridinium (MPP+) were utilized to investigate the effect of MALAT1 on PD. TUNEL assay was performed to detect apoptosis of DA neurons in PD mice. Flow cytometry analysis was carried out to measure apoptosis of SH-SY5Y cells. Caspase3 activity and Cleaved Caspase3 expression were tested by caspase3 assay kit and western blot, respectively. TargetScan software and luciferase reporter assay were used to explore the relationship between MALAT1 and miR-124. Results MALAT1 was up-regulated and miR-124 was down-regulated in MPTP-induced PD mice and MPP+-treated SH-SY5Y cells. MALAT1 knockdown attenuated MPTP-induced apoptosis of DA neurons in MPTP-induced PD mouse model. MALAT1 interacted with miR-124 to negatively regulate its expression. MALAT1 knockdown suppressed MPP+-induced apoptosis in SH-SY5Y cells, while miR-124 downregulation abrogated this effect. Moreover, MALAT1 knockdown improved miR-124 expression in MPTP/MPP+ induced models of PD. Conclusions MALAT1 promotes the apoptosis by sponging miR-124 in mouse models of PD and in vitro model of PD, providing a potential theoretical foundation for the clinical application of MALAT1 against PD.
Collapse
Affiliation(s)
- Wei Liu
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, 475000 China
| | - Qishun Zhang
- Department of Rehabilitation, Huaihe Hospital of Henan University, Kaifeng, 475000 China
| | - Jianlei Zhang
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, 475000 China
| | - Wujun Pan
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, 475000 China
| | - Jingya Zhao
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, 475000 China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052 China
| |
Collapse
|
378
|
Zhang Y, Zhang Q, Zhang M, Yuan M, Wang Z, Zhang J, Zhou X, Zhang Y, Lin F, Na H, Ren S, Zuo Y. DC - SIGNR by influencing the lncRNA HNRNPKP2 upregulates the expression of CXCR4 in gastric cancer liver metastasis. Mol Cancer 2017; 16:78. [PMID: 28403883 PMCID: PMC5390362 DOI: 10.1186/s12943-017-0639-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 03/16/2017] [Indexed: 12/25/2022] Open
Abstract
Background Profiling evidences of selectin demonstrate that they play an crucial role in cancer progression and metastasis. However, DC-SIGNR as a family member of selectin participates in gastric cancer liver metastasis remains unknown. Methods The serum level of DC-SIGNR was evaluated in gastric cancer patients by ELISA. Manipulation DC-SIGNR expression in BGC823 and SGC7901 cell lines was mediated by lentivirus. Investigation the biological effects of DC-SIGNR were verified by MTT, wounding and transwell in vitro and experiments on animals to confirm gastric cancer liver metastasis by IVIS. Insights of the mechanism were employed microarray and bioinformatic analysis. Further to confirm the results were conducted by qRT-PCR, western blot and by flow cytometry. Results DC-SIGNR serum level was significantly increased in gastric cancer patients compared with healthy group. Additionally, DC-SIGNR level was associated with an advanced pathological stage in gastric cancer patients. DC-SIGNR knockdown inhibited the proliferation, migration and invasion of gastric cancer cells in vitro and suppressed the liver metastasis in vivo. While, DC-SIGNR overexpression promoted cell proliferation, migration and invasion. In mechanism, HNRNPKP2 as a lncRNA was upregulated after DC-SIGNR knockdown. Importantly, STAT5A promoted HNRNPKP2 expression after knockdown DC-SIGNR. Furthermore after HNRNPKP2 depletion, the downstream target gene CXCR4 was downregulated. Conclusions DC-SIGNR promoted gastric cancer liver metastasis mediated with HNRNPKP2 which expression was regulated by STAT5A. And HNRNPKP2 decreased the expression of downstream target gene CXCR4. These findings indicated potential therapeutic candidates for gastric cancer liver metastasis. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0639-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, lvshun South Road West 9, Dalian, 116044, China
| | - Qianshi Zhang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Shahekou District Zhongshan Road no. 467, 116023, Dalian, China
| | - Mengyang Zhang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Shahekou District Zhongshan Road no. 467, 116023, Dalian, China
| | - Menglang Yuan
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Shahekou District Zhongshan Road no. 467, 116023, Dalian, China
| | - Zhaohui Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Jingbo Zhang
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, lvshun South Road West 9, Dalian, 116044, China
| | - Xu Zhou
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, lvshun South Road West 9, Dalian, 116044, China
| | - Yinan Zhang
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, lvshun South Road West 9, Dalian, 116044, China
| | - Fang Lin
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, lvshun South Road West 9, Dalian, 116044, China
| | - Heya Na
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, lvshun South Road West 9, Dalian, 116044, China
| | - Shuangyi Ren
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Shahekou District Zhongshan Road no. 467, 116023, Dalian, China.
| | - Yunfei Zuo
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, lvshun South Road West 9, Dalian, 116044, China.
| |
Collapse
|
379
|
Wu L, Wang X, Guo Y. Long non-coding RNA MALAT1 is upregulated and involved in cell proliferation, migration and apoptosis in ovarian cancer. Exp Ther Med 2017; 13:3055-3060. [PMID: 28587379 DOI: 10.3892/etm.2017.4304] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/09/2016] [Indexed: 01/22/2023] Open
Abstract
Ovarian cancer (OC) is the leading cause of mortality among gynecological malignancies. Although microRNAs are known to have a key regulatory role in OC, the involvement of long non-coding RNAs in the disease is less established. Previous studies have demonstrated that metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a tumor oncogene in many cancers, though its role in OC remains unclear. The present study reported that MALAT1 expression was markedly upregulated in OC, by knockdown of MALAT1 expression in vivo, using RNA interference (RNAi) with small-interfering RNA (siRNA). It was found that MALAT1 expression was positively correlated with the International Federation of Gynecology and Obstetrics stages of OC, tumor histological grade and lymph node metastasis. In addition, the differential MALAT1 levels between a human ovarian epithelial cell line (HOSE) and OC cell lines (ES-2, OVCAR3, SKOV3 and HO8910) were compared in vitro. Notably, MALAT1 was expressed to a high level in OC cells. Furthermore, exogenous knockdown of MALAT1 significantly repressed growth and migration of OC cells, and promoted their apoptosis. Collectively, the current findings suggest that upregulation of MALAT1 in OC may facilitate tumorigenesis and metastasis. Knockdown of MALAT1 expression has potential as a novel target for the diagnosis and therapy of OC.
Collapse
Affiliation(s)
- Liqin Wu
- Department of Obstetrics and Gynecology, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163001, P.R. China
| | - Xiaoyu Wang
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Yuna Guo
- Department of Obstetrics, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, P.R. China
| |
Collapse
|
380
|
Chen W, Xu XK, Li JL, Kong KK, Li H, Chen C, He J, Wang F, Li P, Ge XS, Li FC. MALAT1 is a prognostic factor in glioblastoma multiforme and induces chemoresistance to temozolomide through suppressing miR-203 and promoting thymidylate synthase expression. Oncotarget 2017; 8:22783-22799. [PMID: 28187000 PMCID: PMC5410262 DOI: 10.18632/oncotarget.15199] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/22/2017] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor with limited therapeutic options. Temozolomide (TMZ) is a novel cytotoxic agent used as first-line chemotherapy for GBM, however, some individual cells can't be isolated for surgical resection and show treatment-resistance, thus inducing poor prognosis. By using the HiSeq sequencing and bioinformatics methods, we identified lncRNAs showing different expression levels in TMZ-resistant and non-resistant patients. RT-qPCR was then performed in tissues and serum samples, and lncRNA MALAT1 was finally identified to show considerable discriminating potential to identify responding patients from non-responding patients. Moreover, high serum MALAT1 expression was associated with poor chemoresponse and survival in GBM patients receiving TMZ treatment. Subsequently, the TMZ resistant cell lines were established, and the CCK8 assay showed that lncRNA MALAT1 knockdown significantly reversed TMZ resistance in GBM cells. The gain and loss-function experiments revealed that miR-203 was down-regulated by MALAT1 and this interaction has reciprocal effects. Besides, thymidylate synthase (TS) mRNA was identified as a direct target of miR-203. LncRNA MALAT1 inhibition re-sensitized TMZ resistant cells through up-regulating miR-203 and down-regulating TS expression. On the other hand, MALAT1 overexpression promoted resistance by suppressing miR-203 and promoting TS expression. In conclusion, our integrated approach demonstrates that enhanced expression of lncRNA MALAT1 confers a potent poor therapeutic efficacy and inhibition of MALAT1 levels could be a future direction to develop a novel therapeutic strategy to overcome TMZ resistance in GBM patients.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xin-Ke Xu
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Jun-Liang Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Kuan-Kei Kong
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Hui Li
- Department of Respiratory, The First People's Hospital of Foshan, Sun Yat-Sen University, Guangdong Foshan 528000, China
| | - Cheng Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Jing He
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Fangyu Wang
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ping Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xiao-Song Ge
- Department of Oncology, The Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu, China
| | - Fang-Cheng Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| |
Collapse
|
381
|
Sun H, Lin DC, Cao Q, Pang B, Gae DD, Lee VKM, Lim HJ, Doan N, Said JW, Gery S, Chow M, Mayakonda A, Forscher C, Tyner JW, Koeffler HP. Identification of a Novel SYK/c-MYC/MALAT1 Signaling Pathway and Its Potential Therapeutic Value in Ewing Sarcoma. Clin Cancer Res 2017; 23:4376-4387. [DOI: 10.1158/1078-0432.ccr-16-2185] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/05/2016] [Accepted: 03/21/2017] [Indexed: 11/16/2022]
|
382
|
Chung IH, Lu PH, Lin YH, Tsai MM, Lin YW, Yeh CT, Lin KH. The long non-coding RNA LINC01013 enhances invasion of human anaplastic large-cell lymphoma. Sci Rep 2017; 7:295. [PMID: 28331184 PMCID: PMC5428265 DOI: 10.1038/s41598-017-00382-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 02/22/2017] [Indexed: 12/28/2022] Open
Abstract
Anaplastic large-cell lymphoma (ALCL) is a rare type of highly malignant, non-Hodgkin lymphoma (NHL). Currently, only studies on the chimeric oncogene NPM-ALK have reported a link to ALCL progression. However, the specific molecular mechanisms underlying the invasion of ALCL are still unclear. Here, we sought to investigate differentially expressed, long non-coding RNAs (lncRNAs) in ALCL and their potential biological function. Our microarray analyses revealed that LINC01013, a novel non-coding RNA gene, was highly expressed in clinical specimens of ALCL and was significantly upregulated in invasive ALCL cell lines. Knockdown of LINC01013 suppressed tumor cell invasion; conversely, its overexpression enhanced tumor cell invasion. LINC01013-induced invasion was mediated by activation of the epithelial-to-mesenchymal transition (EMT)-associated proteins, snail and fibronectin. Specifically, LINC01013 induced snail, resulting in activation of fibronectin and enhanced ALCL cell invasion. Collectively, these findings support a potential role for LINC01013 in cancer cell invasion through the snail-fibronectin activation cascade and suggest that LINC01013 could potentially be utilized as a metastasis marker in ALCL.
Collapse
Affiliation(s)
- I-Hsiao Chung
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan (R.O.C.)
| | - Pei-Hsuan Lu
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan (R.O.C.).,Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan (R.O.C.)
| | - Yang-Hsiang Lin
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan (R.O.C.)
| | - Ming-Ming Tsai
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan (R.O.C.).,Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan (R.O.C.)
| | - Yun-Wen Lin
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan (R.O.C.)
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan (R.O.C.)
| | - Kwang-Huei Lin
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan (R.O.C.). .,Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan (R.O.C.). .,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan (R.O.C.).
| |
Collapse
|
383
|
Wan L, Zhang L, Fan K, Cheng ZX, Sun QC, Wang JJ. Knockdown of Long Noncoding RNA PCAT6 Inhibits Proliferation and Invasion in Lung Cancer Cells. Oncol Res 2017; 24:161-70. [PMID: 27458097 PMCID: PMC7838661 DOI: 10.3727/096504016x14618564639178] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As a newly identified oncogenic long noncoding RNA (lncRNA), prostate cancer-associated transcript 6 (PCAT6) promoted cellular proliferation and colony formation of prostate cancer. However, the biological function of PCAT6 in lung cancer is still largely unknown. In this study, we found that PCAT6 is significantly increased in cancer tissues compared to normal tissues and positively correlates with metastasis of lung cancer in patients. We then examined PCAT6 expression in lung cancer cell lines and identified that PCAT6 expression was significantly elevated in lung cancer cells compared to normal human bronchial epithelial (NHBE) cells, especially in CL1-5 and H446 cells. PCAT6 knockdown significantly inhibited cellular proliferation and metastasis, as well as induced early apoptosis of lung cancer cells. Molecular analysis revealed that PCAT6 regulated the expression of two pivotal cancer-related proteins, c-Myc and p53, in lung cancer cells. However, PCAT6 was not directly combined with c-Myc and p53 as confirmed by RNA immunoprecipitation. Finally, a retrospective study further revealed that PCAT6 negatively correlates with overall survival of lung cancer patients. In conclusion, these results suggest that PCAT6 could play an oncogenic role in lung cancer progression and may serve as a biomarker for prognosis of lung cancer patients.
Collapse
Affiliation(s)
- Li Wan
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
384
|
Malat1 regulates myogenic differentiation and muscle regeneration through modulating MyoD transcriptional activity. Cell Discov 2017; 3:17002. [PMID: 28326190 PMCID: PMC5348715 DOI: 10.1038/celldisc.2017.2] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/29/2016] [Indexed: 12/21/2022] Open
Abstract
Malat1 is one of the most abundant long non-coding RNAs in various cell types; its exact cellular function is still a matter of intense investigation. In this study we characterized the function of Malat1 in skeletal muscle cells and muscle regeneration. Utilizing both in vitro and in vivo assays, we demonstrate that Malat1 has a role in regulating gene expression during myogenic differentiation of myoblast cells. Specifically, we found that knockdown of Malat1 accelerates the myogenic differentiation in cultured cells. Consistently, Malat1 knockout mice display enhanced muscle regeneration after injury and deletion of Malat1 in dystrophic mdx mice also improves the muscle regeneration. Mechanistically, in the proliferating myoblasts, Malat1 recruits Suv39h1 to MyoD-binding loci, causing trimethylation of histone 3 lysine 9 (H3K9me3), which suppresses the target gene expression. Upon differentiation, the pro-myogenic miR-181a is increased and targets the nuclear Malat1 transcripts for degradation through Ago2-dependent nuclear RNA-induced silencing complex machinery; the Malat1 decrease subsequently leads to the destabilization of Suv39h1/HP1β/HDAC1-repressive complex and displacement by a Set7-containing activating complex, which allows MyoD trans-activation to occur. Together, our findings identify a regulatory axis of miR-181a-Malat1-MyoD/Suv39h1 in myogenesis and uncover a previously unknown molecular mechanism of Malat1 action in gene regulation.
Collapse
|
385
|
Tang PMK, Tang PCT, Chung JYF, Lan HY. TGF-β1 signaling in kidney disease: From Smads to long non-coding RNAs. Noncoding RNA Res 2017; 2:68-73. [PMID: 30159422 PMCID: PMC6096420 DOI: 10.1016/j.ncrna.2017.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/14/2017] [Accepted: 04/06/2017] [Indexed: 01/07/2023] Open
Abstract
Transforming growth factor-β1 (TGF-β1) has an essential role in the development of kidney diseases. However, targeting TGF-β1 is not a good strategy for fibrotic diseases due to its multifunctional characteristic in physiology. A precise therapeutic target maybe identified by further resolving the underlying TGF-β1 driven mechanisms in renal inflammation and fibrosis. Smad signaling is uncovered as a key pathway of TGF-β1-mediated renal injury, where Smad3 is hyper-activated but Smad7 is suppressed. Mechanistic studies revealed that TGF-β1/Smad3 is capable of promoting renal inflammation and fibrosis via regulating non-coding RNAs. More importantly, involvement of disease- and tissue-specific TGF-β1-dependent long non-coding RNAs (lncRNA) have been recently recognized in a number of kidney diseases. In this review, current understanding of TGF-β1 driven lncRNAs in the pathogenesis of kidney injury, diabetic nephropathy and renal cell carcinoma will be intensively discussed.
Collapse
Affiliation(s)
- Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Philip Chiu-Tsun Tang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jeff Yat-Fai Chung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hui-Yao Lan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
386
|
Li S, Shuch BM, Gerstein MB. Whole-genome analysis of papillary kidney cancer finds significant noncoding alterations. PLoS Genet 2017; 13:e1006685. [PMID: 28358873 PMCID: PMC5391127 DOI: 10.1371/journal.pgen.1006685] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 04/13/2017] [Accepted: 03/13/2017] [Indexed: 01/30/2023] Open
Abstract
To date, studies on papillary renal-cell carcinoma (pRCC) have largely focused on coding alterations in traditional drivers, particularly the tyrosine-kinase, Met. However, for a significant fraction of tumors, researchers have been unable to determine a clear molecular etiology. To address this, we perform the first whole-genome analysis of pRCC. Elaborating on previous results on MET, we find a germline SNP (rs11762213) in this gene predicting prognosis. Surprisingly, we detect no enrichment for small structural variants disrupting MET. Next, we scrutinize noncoding mutations, discovering potentially impactful ones associated with MET. Many of these are in an intron connected to a known, oncogenic alternative-splicing event; moreover, we find methylation dysregulation nearby, leading to a cryptic promoter activation. We also notice an elevation of mutations in the long noncoding RNA NEAT1, and these mutations are associated with increased expression and unfavorable outcome. Finally, to address the origin of pRCC heterogeneity, we carry out whole-genome analyses of mutational processes. First, we investigate genome-wide mutational patterns, finding they are governed mostly by methylation-associated C-to-T transitions. We also observe significantly more mutations in open chromatin and early-replicating regions in tumors with chromatin-modifier alterations. Finally, we reconstruct cancer-evolutionary trees, which have markedly different topologies and suggested evolutionary trajectories for the different subtypes of pRCC.
Collapse
Affiliation(s)
- Shantao Li
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Brian M. Shuch
- Department of Urology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Mark B. Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, United States of America
- Department of Computer Science, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
387
|
Zhang R, Hardin H, Huang W, Chen J, Asioli S, Righi A, Maletta F, Sapino A, Lloyd RV. MALAT1 Long Non-coding RNA Expression in Thyroid Tissues: Analysis by In Situ Hybridization and Real-Time PCR. Endocr Pathol 2017; 28:7-12. [PMID: 27696303 PMCID: PMC5313332 DOI: 10.1007/s12022-016-9453-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Long non-coding RNAs (lncRNAs) are important for transcription and for epigenetic or posttranscriptional regulation of gene expression and may contribute to carcinogenesis. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), an lncRNA involved in the regulation of the cell cycle, cell proliferation, and cell migration, is known to be deregulated in multiple cancers. Here, we analyzed the expression of MALAT1 on 195 cases of benign and malignant thyroid neoplasms by using tissue microarrays for RNA in situ hybridization (ISH) and real-time PCR. MALAT1 is highly expressed in normal thyroid (NT) tissues and thyroid tumors, with increased expression during progression from NT to papillary thyroid carcinomas (PTCs) but is downregulated in poorly differentiated thyroid cancers (PDCs) and anaplastic thyroid carcinomas (ATCs) compared to NT. Induction of epithelial to mesenchymal transition (EMT) by transforming growth factor (TGF)-beta in a PTC cell line (TPC1) led to increased MALAT1 expression, supporting a role for MALAT1 in EMT in thyroid tumors. This is the first ISH study of MALAT1 expression in thyroid tissues. It also provides the first piece of evidence suggesting MALAT1 downregulation in certain thyroid malignancies. Our findings support the notion that ATCs may be molecularly distinct from low-grade thyroid malignancies and suggest that MALAT1 may function both as an oncogene and as a tumor suppressor in different types of thyroid tumors.
Collapse
Affiliation(s)
- Ranran Zhang
- Department of Laboratory Medicine and Pathology K4/436, University of Wisconsin School of Medicine and Public Health, Box 8550, Madison, WI, 53792, USA
| | - Heather Hardin
- Department of Laboratory Medicine and Pathology K4/436, University of Wisconsin School of Medicine and Public Health, Box 8550, Madison, WI, 53792, USA
| | - Wei Huang
- Department of Laboratory Medicine and Pathology K4/436, University of Wisconsin School of Medicine and Public Health, Box 8550, Madison, WI, 53792, USA
| | - Jidong Chen
- Department of Laboratory Medicine and Pathology K4/436, University of Wisconsin School of Medicine and Public Health, Box 8550, Madison, WI, 53792, USA
| | | | | | | | | | - Ricardo V Lloyd
- Department of Laboratory Medicine and Pathology K4/436, University of Wisconsin School of Medicine and Public Health, Box 8550, Madison, WI, 53792, USA.
| |
Collapse
|
388
|
Malakar P, Shilo A, Mogilevsky A, Stein I, Pikarsky E, Nevo Y, Benyamini H, Elgavish S, Zong X, Prasanth KV, Karni R. Long Noncoding RNA MALAT1 Promotes Hepatocellular Carcinoma Development by SRSF1 Upregulation and mTOR Activation. Cancer Res 2017; 77:1155-1167. [PMID: 27993818 PMCID: PMC5334181 DOI: 10.1158/0008-5472.can-16-1508] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/26/2016] [Accepted: 12/06/2016] [Indexed: 12/30/2022]
Abstract
Several long noncoding RNAs (lncRNA) are abrogated in cancer but their precise contributions to oncogenesis are still emerging. Here we report that the lncRNA MALAT1 is upregulated in hepatocellular carcinoma and acts as a proto-oncogene through Wnt pathway activation and induction of the oncogenic splicing factor SRSF1. Induction of SRSF1 by MALAT1 modulates SRSF1 splicing targets, enhancing the production of antiapoptotic splicing isoforms and activating the mTOR pathway by modulating the alternative splicing of S6K1. Inhibition of SRSF1 expression or mTOR activity abolishes the oncogenic properties of MALAT1, suggesting that SRSF1 induction and mTOR activation are essential for MALAT1-induced transformation. Our results reveal a mechanism by which lncRNA MALAT1 acts as a proto-oncogene in hepatocellular carcinoma, modulating oncogenic alternative splicing through SRSF1 upregulation. Cancer Res; 77(5); 1155-67. ©2016 AACR.
Collapse
Affiliation(s)
- Pushkar Malakar
- Department of Biochemistry and Molecular Biology, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Asaf Shilo
- Department of Biochemistry and Molecular Biology, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Adi Mogilevsky
- Department of Biochemistry and Molecular Biology, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Ilan Stein
- Department of Immunology and Cancer Research, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Eli Pikarsky
- Department of Immunology and Cancer Research, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Yuval Nevo
- Bioinformatics unit, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Hadar Benyamini
- Bioinformatics unit, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Sharona Elgavish
- Bioinformatics unit, the Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel
| | - Xinying Zong
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Rotem Karni
- Department of Biochemistry and Molecular Biology, Hebrew University-Hadassah Medical School, Ein Karem, Jerusalem, Israel.
| |
Collapse
|
389
|
Wu T, Du Y. LncRNAs: From Basic Research to Medical Application. Int J Biol Sci 2017; 13:295-307. [PMID: 28367094 PMCID: PMC5370437 DOI: 10.7150/ijbs.16968] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 11/02/2016] [Indexed: 01/17/2023] Open
Abstract
This review aimed to summarize the current research contents about long noncoding RNAs (lncRNAs) and some related lncRNAs as molecular biomarkers or therapy strategies in human cancer and cardiovascular diseases. Following the development of various kinds of sequencing technologies, lncRNAs have become one of the most unknown areas that need to be explored. First, the definition and classification of lncRNAs were constantly amended and supplemented because of their complexity and diversity. Second, several methods and strategies have been developed to study the characteristic of lncRNAs, including new species identifications, subcellular localization, gain or loss of function, molecular interaction, and bioinformatics analysis. Third, based on the present results from basic researches, the working mechanisms of lncRNAs were proved to be different forms of interactions involving DNAs, RNAs, and proteins. Fourth, lncRNA can play different important roles during the embryogenesis and organ differentiations. Finally, because of the tissue-specific expression of lncRNAs, they could be used as biomarkers or therapy targets and effectively applied in different kinds of diseases, such as human cancer and cardiovascular diseases.
Collapse
Affiliation(s)
- Tao Wu
- Cardiovascular Department, The Affiliated Hospital of Medical College, Ningbo University, No.247, Renmin Road, Jiangbei District, Ningbo, China
| | - Yantao Du
- Ningbo Institute of Medical Science, No.42-46, Yangshan Road, Jiangbei District, Ningbo, China
| |
Collapse
|
390
|
Zhang TH, Liang LZ, Liu XL, Wu JN, Su K, Chen JY, Zheng QY, Huang HZ, Liao GQ. Long non-coding RNA MALAT1 interacts with miR-124 and modulates tongue cancer growth by targeting JAG1. Oncol Rep 2017; 37:2087-2094. [DOI: 10.3892/or.2017.5445] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 10/10/2016] [Indexed: 11/06/2022] Open
|
391
|
Liu K, Huang J, Ni J, Song D, Ding M, Wang J, Huang X, Li W. MALAT1 promotes osteosarcoma development by regulation of HMGB1 via miR-142-3p and miR-129-5p. Cell Cycle 2017; 16:578-587. [PMID: 28346809 DOI: 10.1080/15384101.2017.1288324] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Recently, emerging evidence has demonstrated that metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long non-coding RNAs (lncRNAs), contributes to the initiation and development of tumors, including osteosarcoma (OS). Multiple studies have suggested an oncogenic role of MALAT1 and high-mobility group protein B1 (HMGB1) in OS tumorigenesis and metastasis, but the effects and mechanisms are not unanimous. Here, we showed that MALAT1 and HMGB1 were significantly increased in human OS cell lines and knockdown of MALAT1 reduced HMGB1 expression. By using online tools, we screen out 2 candidate miRNAs, miR-142-3p and miR-129-5p which may be associated with both MALAT1 and HMGB1. Luciferase reporter assay revealed a direct interaction between the 2 miRNAs and MALAT1, respectively, via a putative binding site within MALAT1. Meanwhile, both the 2 miRNAs could bind to HMGB1 3'-untranslated region (3'-UTR) and regulate HMGB1 expression. Moreover, knockdown of MALAT1 decreased HMGB1 expression, inhibited OS cell growth and promoted apoptosis, while miR-142-3p and miR-129-5p inhibitor partly restored the inhibitory effect of MALAT1 knockdown on HMGB1 expression, OS cell growth and the promotion of apoptosis. In OS tissues, the expression of MALAT1 and HMGB1 was upregulated while the expression of miR-142-3p and miR-129-5p was downregulated. Together, our results support a MALAT1/miR-142-3p/miR-129-5p/HMGB1 axis in OS cell proliferation and tumor progression. MALAT1 promoted OS cell growth through inhibition of miR-142-3p or miR-129-5p and by targeting HMGB1.
Collapse
Affiliation(s)
- Ke Liu
- a Department of Ophthalmology , The Second Xiangya Hospital, Central South University , Changsha , Hunan , P.R. China
| | - Jun Huang
- b Department of Orthopaedics , The Second Xiangya Hospital, Central South University , Changsha , Hunan , P.R. China
| | - Jiangdong Ni
- b Department of Orthopaedics , The Second Xiangya Hospital, Central South University , Changsha , Hunan , P.R. China
| | - Deye Song
- b Department of Orthopaedics , The Second Xiangya Hospital, Central South University , Changsha , Hunan , P.R. China
| | - Muliang Ding
- b Department of Orthopaedics , The Second Xiangya Hospital, Central South University , Changsha , Hunan , P.R. China
| | - Junjie Wang
- b Department of Orthopaedics , The Second Xiangya Hospital, Central South University , Changsha , Hunan , P.R. China
| | - Xianzhe Huang
- b Department of Orthopaedics , The Second Xiangya Hospital, Central South University , Changsha , Hunan , P.R. China
| | - Wenzhao Li
- b Department of Orthopaedics , The Second Xiangya Hospital, Central South University , Changsha , Hunan , P.R. China
| |
Collapse
|
392
|
Wang J, Sun G. FOXO1-MALAT1-miR-26a-5p Feedback Loop Mediates Proliferation and Migration in Osteosarcoma Cells. Oncol Res 2017; 25:1517-1527. [PMID: 28160461 PMCID: PMC7841132 DOI: 10.3727/096504017x14859934460780] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
miR-26a has been found to be downregulated in osteosarcoma (OS) when compared with normal control tissues and has been shown to suppress the malignant behaviors of OS cells. The underlying mechanism, nevertheless, remains unknown. In our study, the long noncoding RNA MALAT1, confirmed to be significantly upregulated in OS, is first shown to be capable of promoting proliferation and migration by directly suppressing miR-26a-5p in OS cells. In addition, we have identified forkhead box O1 (FOXO1) as a transcriptional factor of MALAT1 that can negatively regulate MALAT1. We have shown that MALAT1 promoted growth and migration through inhibiting miR-26a-5p in OS cells. Suppression of FOXO1, identified as a regulatory transcriptional factor of MALAT1, was shown to be able to slow down both proliferation and metastases in OS cells, suggesting that targeting FOXO1 can be useful in the therapy of patients with OS.
Collapse
|
393
|
Wang Y, Liu J, Bai H, Dang Y, Lv P, Wu S. Long intergenic non-coding RNA 00152 promotes renal cell carcinoma progression by epigenetically suppressing P16 and negatively regulates miR-205. Am J Cancer Res 2017; 7:312-322. [PMID: 28337379 PMCID: PMC5336504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 11/02/2016] [Indexed: 06/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been reported to play important roles in the tumorigenesis and development of several human cancers. Long intergenic non-coding RNA 152 (LINC00152) is significantly up-regulated in some solid tumors. However, the role of LINC00152 in the pathogenesis and development of renal cell carcinoma (RCC) remains largely unclear. In the study, we showed that LINC00152 expression was up-regulated in RCC tissues compared with adjacent normal tissues and revealed that LINC00152 expression was positively correlated with lymph node metastasis, higher TNM stage, and poor over survival (OS) time in RCC patients. Furthermore, knockdown of LINC00152 inhibited RCC cell proliferation and S phase cell proportion in vitro. Mechanistically, RNA immunoprecipitation (RIP) and Chromatin immunoprecipitation (ChIP) verified that LINC00152 bound to Enhancer of zeste homolog 2 (EZH2), LSD1 and histone H3 at lysine 27 (H3K27me3) and epigenetically suppressing P16 expression. In addition, LINC00152 expression was negatively correlated with miR-205 in RCC and luciferase reporter assays demonstrated that miR-205 was a target of LINC00152. These findings suggested that LINC00152 may contribute to RCC progression by epigenetically repressing P16 expression and interacted with miR-205. Thus, LINC00152 acted as a novel prognostic marker and a potential therapeutic target for RCC.
Collapse
Affiliation(s)
- Yongjun Wang
- Department of Cardiovascular, Hebei Chest HospitalShijiazhuang, Hebei, China
| | - Jianzhen Liu
- Department of Urology, Hebei Chest HospitalShijiazhuang, Hebei, China
| | - Hongzhong Bai
- Department of Imaging, Hebei Chest HospitalShijiazhuang, Hebei, China
| | - Yi Dang
- Department of Cardiovascular, Hebei general HospitalShijiazhuang, Hebei, China
| | - Pei Lv
- Department of Nephrology, Hebei Chest HospitalShijiazhuang, Hebei, China
| | - Shucai Wu
- Department of Respiratory, Hebei Chest HospitalShijiazhuang, Hebei, China
| |
Collapse
|
394
|
Wang Y, Cheng N, Luo J. Downregulation of lncRNA ANRIL represses tumorigenicity and enhances cisplatin-induced cytotoxicity via regulating microRNA let-7a in nasopharyngeal carcinoma. J Biochem Mol Toxicol 2017; 31. [PMID: 28117929 DOI: 10.1002/jbt.21904] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/26/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Yandan Wang
- Department of Otorhinolaryngology; Huaihe Hospital of Henan University; Kaifeng 475000 People's Republic of China
| | - Nan Cheng
- Department of Otorhinolaryngology; Huaihe Hospital of Henan University; Kaifeng 475000 People's Republic of China
| | - Junpeng Luo
- Department of Oncology; Huaihe Hospital of Henan University; Kaifeng 475000 People's Republic of China
| |
Collapse
|
395
|
Long Noncoding RNA Malat1 Regulates Cerebrovascular Pathologies in Ischemic Stroke. J Neurosci 2017; 37:1797-1806. [PMID: 28093478 DOI: 10.1523/jneurosci.3389-16.2017] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/04/2017] [Accepted: 01/10/2017] [Indexed: 11/21/2022] Open
Abstract
The study was designed to determine the role of long noncoding RNA (lncRNA), metastasis-associated lung adenocarcinoma transcript 1 (Malat1), in ischemic stroke outcome. Primary mouse brain microvascular endothelial cells (BMECs) were cultured and treated with Malat1 GapmeR before 16 h oxygen and glucose depravation (OGD). Cell death was assayed by LDH and MTT methods. Malat1 knock-out and wild-type mice were subjected to 1 h of middle cerebral artery occlusion (MCAO) and 24-72 h of reperfusion. To explore the underlying mechanism, apoptotic and inflammatory factors were measured by qPCR, ELISA, and Western blotting. The physical interaction between Malat1 and apoptotic or inflammatory factors was measured by RNA immunoprecipitation. Increased Malat1 levels were found in cultured mouse BMECs after OGD as well as in isolated cerebral microvessels in mice after MCAO. Silencing of Malat1 by Malat1 GapmeR significantly increased OGD-induced cell death and Caspase 3 activity in BMECs. Silencing of Malat1 also significantly aggravated OGD-induced expression of the proapoptotic factor Bim and proinflammatory cytokines MCP-1, IL-6, and E-selectin. Moreover, Malat1 KO mice presented larger brain infarct size, worsened neurological scores, and reduced sensorimotor functions. Consistent with in vitro findings, significantly increased expression of proapoptotic and proinflammatory factors was also found in the cerebral cortex of Malat1 KO mice after ischemic stroke compared with WT controls. Finally, we demonstrated that Malat1 binds to Bim and E-selectin both in vitro and in vivo Our study suggests that Malat1 plays critical protective roles in ischemic stroke.SIGNIFICANCE STATEMENT Accumulative studies have demonstrated the important regulatory roles of microRNAs in vascular and neural damage after ischemic stroke. However, the functional significance and mechanisms of other classes of noncoding RNAs in cerebrovascular pathophysiology after stroke are less studied. Here we demonstrate a novel role of Malat1, a long noncoding RNA that has been originally identified as a prognostic marker for non-small cell lung cancer, in cerebrovascular pathogenesis of ischemic stroke. Our experiments have provided the first evidence that Malat1 plays anti-apoptotic and anti-inflammatory roles in brain microvasculature to reduce ischemic cerebral vascular and parenchymal damages. Our studies also suggest that lncRNAs can be therapeutically targeted to minimize poststroke brain damage.
Collapse
|
396
|
Li P, Zhang X, Wang H, Wang L, Liu T, Du L, Yang Y, Wang C. MALAT1 Is Associated with Poor Response to Oxaliplatin-Based Chemotherapy in Colorectal Cancer Patients and Promotes Chemoresistance through EZH2. Mol Cancer Ther 2017; 16:739-751. [PMID: 28069878 DOI: 10.1158/1535-7163.mct-16-0591] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/03/2016] [Accepted: 12/23/2016] [Indexed: 12/21/2022]
Abstract
A major reason for oxaliplatin chemoresistance in colorectal cancer is the acquisition of epithelial-mesenchymal transition (EMT) in cancer cells. The long noncoding RNA (lncRNA), MALAT1, is a highly conserved nuclear ncRNA and a key regulator of metastasis development in several cancers. However, its role in oxaliplatin-induced metastasis and chemoresistance is not well known. In this study, we aim to investigate the prognostic and therapeutic role of lncRNA MALAT1 in colorectal cancer patients receiving oxaliplatin-based therapy and further explore the potential transcriptional regulation through interaction with EZH2 based on the established HT29 oxaliplatin-resistant cells. Our results showed that high MALAT1 expression was associated with reduced patient survival and poor response to oxaliplatin-based chemotherapy in advanced colorectal cancer patients. Oxaliplatin-resistant colorectal cancer cells exhibited high MALAT1 expression and EMT. LncRNA MALAT1 knockdown enhances E-cadherin expression and inhibits oxaliplatin-induced EMT in colorectal cancer cells. EZH2 is highly expressed and associated with the 3' end region of lncRNA MALAT1 in colorectal cancer, and this association suppressed the expression of E-cadherin. Furthermore, targeted inhibition of MALAT1 or EZH2 reversed EMT and chemoresistance induced by oxaliplatin. Finally, the interaction between lncRNA MALAT1 and miR-218 was observed, which further indicated its prognostic value in patients who received standard FOLFOX (oxaliplatin combine with 5-fluorouracil and leucovorin) treatment. In conclusion, this study illuminates the prognostic role of lncRNA MALAT1 in colorectal cancer patients receiving oxaliplatin-based treatment and further demonstrates how lncRNA MALAT1 confers a chemoresistant function in colorectal cancer. Thus, lncRNA MALAT1 may serve as a promising prognostic and therapeutic target for colorectal cancer patients. Mol Cancer Ther; 16(4); 739-51. ©2017 AACR.
Collapse
Affiliation(s)
- Peilong Li
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Haiyan Wang
- Department of Clinical Laboratory, Women & Children's Hospital of Linyi, Linyi, Shandong Province, China
| | - Lili Wang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Tong Liu
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Lutao Du
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Yongmei Yang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
397
|
Brock M, Schuoler C, Leuenberger C, Bühlmann C, Haider TJ, Vogel J, Ulrich S, Gassmann M, Kohler M, Huber LC. Analysis of hypoxia-induced noncoding RNAs reveals metastasis-associated lung adenocarcinoma transcript 1 as an important regulator of vascular smooth muscle cell proliferation. Exp Biol Med (Maywood) 2017; 242:487-496. [PMID: 28056547 DOI: 10.1177/1535370216685434] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Vascular remodeling, a pathogenic hallmark in pulmonary hypertension, is mainly driven by a dysbalance between proliferation and apoptosis of human pulmonary artery smooth muscle cells. It has previously been shown that microRNAs are involved in the pathogenesis of pulmonary hypertension. However, the role of long noncoding RNAs has not been evaluated. long noncoding RNA expression was quantified in human pulmonary artery smooth muscle cells using PCR arrays and quantitative PCR. Knockdown of genes was performed by transfection of siRNA or GapmeR. Proliferation and migration were measured using BrdU incorporation and wound healing assays. The mouse model of hypoxia-induced PH was used to determine the physiological meaning of identified long noncoding RNAs. The expression of 84 selected long noncoding RNAs was assessed in hypoxic human pulmonary artery smooth muscle cells and the levels of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) were significantly increased. Depletion of hypoxia-inducible factor 1α abolished the hypoxia-induced upregulation of metastasis-associated lung adenocarcinoma transcript 1 expression. Silencing of MALAT1 significantly decreased proliferation and migration of human pulmonary artery smooth muscle cells. In vivo, MALAT1 expression was significantly increased in lungs of hypoxic mice. Of note, targeting of MALAT1 by GapmeR ameliorated heart hypertrophy in mice with pulmonary hypertension. This is the first report on functional characterization of MALAT1 in the pulmonary vasculature. Our data provide evidence that MALAT1 expression is significantly increased by hypoxia, probably by hypoxia-inducible factor 1α. Intervention experiments confirmed that MALAT1 regulates the proliferative phenotype of smooth muscle cells and silencing of MALAT1 reduced heart hypertrophy in mice with pulmonary hypertension. These data indicate a potential role of MALAT1 in the pathogenesis of pulmonary hypertension. Impact statement Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long noncoding RNA that mediates several biological processes. In the context of vascular biology, MALAT1 has been shown to be inducible by hypoxia and to control cell proliferation. These processes are of major importance for the pathophysiology of hypoxia-induced pulmonary hypertension (PH). Until now, the physiological role of MALAT1 in PH remains unclear. By using smooth muscle cells and by employing an established PH mouse model, we provide evidence that hypoxia induces MALAT1 expression. Moreover, depletion of MALAT1 inhibited migration and proliferation of smooth muscle cells, probably by the induction of cyclin-dependent kinase inhibitors. Of note, MALAT1 was significantly increased in mice exposed to hypoxia and silencing of MALAT1 ameliorated heart hypertrophy in mice with hypoxia-induced PH. Since vascular remodeling and right heart failure as a consequence of pulmonary pressure overload is a major problem in PH, these data have implications for our pathogenetic understanding.
Collapse
Affiliation(s)
- Matthias Brock
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Claudio Schuoler
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland.,2 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich CH-8057, Switzerland
| | - Caroline Leuenberger
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Carlo Bühlmann
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Thomas J Haider
- 2 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich CH-8057, Switzerland.,3 Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich CH-8057, Switzerland
| | - Johannes Vogel
- 2 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich CH-8057, Switzerland.,3 Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich CH-8057, Switzerland
| | - Silvia Ulrich
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Max Gassmann
- 2 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich CH-8057, Switzerland.,3 Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich CH-8057, Switzerland
| | - Malcolm Kohler
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| | - Lars C Huber
- 1 Division of Pulmonology, University Hospital Zurich, University of Zurich, Zurich CH-8091, Switzerland
| |
Collapse
|
398
|
Taheri M, Omrani M, Ghafouri-Fard S. Long Non-coding RNAs Expression in Renal Cell Carcinoma. ACTA ACUST UNITED AC 2017. [DOI: 10.15412/j.jbtw.01061201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
399
|
Chen WM, Huang MD, Sun DP, Kong R, Xu TP, Xia R, Zhang EB, Shu YQ. Long intergenic non-coding RNA 00152 promotes tumor cell cycle progression by binding to EZH2 and repressing p15 and p21 in gastric cancer. Oncotarget 2016; 7:9773-87. [PMID: 26799422 PMCID: PMC4891083 DOI: 10.18632/oncotarget.6949] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 12/22/2015] [Indexed: 12/28/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) play important regulatory roles in several human cancers. Integrated analysis revealed that expression of long intergenic non-coding RNA 152 (LINC00152) was significantly upregulated in gastric cancer (GC). Further analysis in a cohort of 97 GC patients revealed that LINC00152 expression was positively correlated with tumor invasion depth, lymph node metastasis, higher TNM stage, and poor survival. Gene set enrichment analysis revealed that cell proliferation and cell cycle progression were increased in patients with high LINC00152 expression. In both GC cell lines and xenograft systems, LINC00152 overexpression facilitated GC cell proliferation by accelerating the cell cycle, whereas LINC00152 knockdown had the opposite effect. Moreover, by binding to enhancer of zeste homolog 2 (EZH2), LINC00152 promotes GC tumor cell cycle progression by silencing the expression of p15 and p21. These findings suggest that LINC00152 may play contribute to the progression of GC and may be an effective therapeutic target.
Collapse
Affiliation(s)
- Wen-ming Chen
- Department of Oncology, Jining NO.1 People's Hospital, Jining City, Shandong Province 272011, China
| | - Ming-de Huang
- Department of Medical Oncology, Huai'an First People's Hospital, Nanjing Medical University, Jiangsu Province 223300, China
| | - Dao-ping Sun
- Department of Oncology, Jining NO.1 People's Hospital, Jining City, Shandong Province 272011, China
| | - Rong Kong
- Central Laboratory, Subei People's Hospital of Jiangsu province, Yangzhou, Jiangsu Province 225001, China
| | - Tong-peng Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu Province 210029, China
| | - Rui Xia
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Er-bao Zhang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Yong-qian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, Jiangsu Province 210029, China
| |
Collapse
|
400
|
Wang X, Sehgal L, Jain N, Khashab T, Mathur R, Samaniego F. LncRNA MALAT1 promotes development of mantle cell lymphoma by associating with EZH2. J Transl Med 2016; 14:346. [PMID: 27998273 PMCID: PMC5175387 DOI: 10.1186/s12967-016-1100-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/29/2016] [Indexed: 02/07/2023] Open
Abstract
Background Mantle cell lymphoma (MCL) is considered an aggressive subtype of non-Hodgkin’s lymphoma with variable treatment responses. There is an urgent need to identify novel markers with prognostic and therapeutic value for MCL. Long non-coding RNAs (lncRNAs) have emerged as key regulators in cancers, including MCL. Metastasis-associated lung adenocarcinoma transcript 1(MALAT1), a lncRNA located at pathognomonic translocation site of t (11; 14) of MCL. MALAT1 is known to be overexpressed in solid tumors and hematologic malignancies. However, the pathological role and clinical relevance of MALAT1 in MCL are not completely understood. Methods We quantified MALAT1 in MCL samples (40) and CD19+ B cells by quantitative real time polymerase chain reaction (qRT-PCR) and correlated levels with clinical outcome. We silenced MALAT1 in MCL cell lines and analyzed cells in tumorigenic assays and formation of transcription complexes. Results We found that the expression of MALAT1 was elevated in human MCL tumors and cell lines as compared to normal controls, and the elevated levels of MALAT1 correlated with higher MCL international prognostic index (MIPI) and reduced overall survival. MCL with knockdown of MALAT1 showed impaired cell proliferation, facilitated apoptosis and produced fewer clonogenic foci. The increased expression of p21 and p27 upon MALAT1 knockdown was regulated by enhancer of zeste homolog 2 (EZH2). Moreover, decreased phosphorylation of EZH2 at T350 attenuated the binding to MALAT1. Conclusions Our findings illuminate the oncogenic role of MALAT1, which may serve as a novel biomarker and as a therapeutic target in MCL. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1100-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xin Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lalit Sehgal
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| | - Neeraj Jain
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Tamer Khashab
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.,Department of Internal Medicine, Lankenau Medical Center, Wynnewood, PA, USA
| | - Rohit Mathur
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Felipe Samaniego
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|