151
|
Human Long Noncoding RNA Regulation of Stem Cell Potency and Differentiation. Stem Cells Int 2017; 2017:6374504. [PMID: 28951743 PMCID: PMC5603141 DOI: 10.1155/2017/6374504] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/12/2017] [Accepted: 08/02/2017] [Indexed: 12/15/2022] Open
Abstract
Because of their capability of differentiation into lineage-specific cells, stem cells are an attractive therapeutic modality in regenerative medicine. To develop an effective stem cell-based therapeutic strategy with predictable results, deeper understanding of the underlying molecular mechanisms of stem cell differentiation and/or pluripotency maintenance is required. Thus, reviewing the key factors involved in the transcriptional and epigenetic regulation of stem cell differentiation and maintenance is important. Accumulating data indicate that long noncoding RNAs (lncRNAs) mediate numerous biological processes, including stem cell differentiation and maintenance. Here, we review recent findings on the human lncRNA regulation of stem cell potency and differentiation. Although the clinical implication of these lncRNAs is only beginning to be elucidated, it is anticipated that lncRNAs will become important therapeutic targets in the near future.
Collapse
|
152
|
Leti F, DiStefano JK. Long Noncoding RNAs as Diagnostic and Therapeutic Targets in Type 2 Diabetes and Related Complications. Genes (Basel) 2017; 8:genes8080207. [PMID: 28829354 PMCID: PMC5575670 DOI: 10.3390/genes8080207] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/16/2017] [Accepted: 08/16/2017] [Indexed: 01/20/2023] Open
Abstract
Protein-coding genes represent only a small fraction of the human genome. In the past, the majority of the genomic sequence has been considered transcriptionally silent, but recent large-scale studies have uncovered an array of functionally significant elements, including non-protein-coding transcripts, within these noncoding regions of the human genome. Long noncoding RNAs (lncRNAs), a class of noncoding transcripts with lengths >200 nucleotides, are pervasively transcribed in the genome and function as signals, decoys, guides, or scaffolds to regulate gene expression. More than 200 diseases have been associated with dysregulated or dysfunctional lncRNAs, and new associations continue to accumulate in the literature. The role of lncRNAs in the pathogenesis of type 2 diabetes mellitus and related complications has only recently been recognized, but there is already evidence for their involvement in many of the pathophysiological mechanisms underlying the disease. In this review, we summarize the current knowledge of the functions and underlying mechanisms of lncRNA activity with a focus on type 2 diabetes mellitus and related renal and retinal complications of the disease. We also discuss the potential of lncRNAs to serve as therapeutic targets for drug development and diagnostic markers for clinical applications in the management of diabetes.
Collapse
Affiliation(s)
- Fatjon Leti
- Department of Biomedical Research, National Jewish Health, Denver, CO 80210, USA.
| | - Johanna K DiStefano
- Department of Biomedical Research, National Jewish Health, Denver, CO 80210, USA.
| |
Collapse
|
153
|
Piccoli MT, Gupta SK, Viereck J, Foinquinos A, Samolovac S, Kramer FL, Garg A, Remke J, Zimmer K, Batkai S, Thum T. Inhibition of the Cardiac Fibroblast–Enriched lncRNA
Meg3
Prevents Cardiac Fibrosis and Diastolic Dysfunction. Circ Res 2017. [DOI: 10.1161/circresaha.117.310624] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Rationale:
Cardiac fibroblasts (CFs) drive extracellular matrix remodeling after pressure overload, leading to fibrosis and diastolic dysfunction. Recent studies described the role of long noncoding RNAs (lncRNAs) in cardiac pathologies. Nevertheless, detailed reports on lncRNAs regulating CF biology and describing their implication in cardiac remodeling are still missing.
Objective:
Here, we aimed at characterizing lncRNA expression in murine CFs after chronic pressure overload to identify CF-enriched lncRNAs and investigate their function and contribution to cardiac fibrosis and diastolic dysfunction.
Methods and Results:
Global lncRNA profiling identified several dysregulated transcripts. Among them, the lncRNA maternally expressed gene 3 (
Meg3
) was found to be mostly expressed by CFs and to undergo transcriptional downregulation during late cardiac remodeling. In vitro,
Meg3
regulated the production of matrix metalloproteinase-2 (MMP-2). GapmeR-mediated silencing of
Meg3
in CFs resulted in the downregulation of
Mmp
-2 transcription, which, in turn, was dependent on P53 activity both in the absence and in the presence of transforming growth factor-β I. Chromatin immunoprecipitation showed that further induction of
Mmp
-2 expression by transforming growth factor-β I was blocked by
Meg3
silencing through the inhibition of P53 binding on the
Mmp-2
promoter. Consistently, inhibition of
Meg3
in vivo after transverse aortic constriction prevented cardiac MMP-2 induction, leading to decreased cardiac fibrosis and improved diastolic performance.
Conclusions:
Collectively, our findings uncover a critical role for
Meg3
in the regulation of MMP-2 production by CFs in vitro and in vivo, identifying a new player in the development of cardiac fibrosis and potential new target for the prevention of cardiac remodeling.
Collapse
Affiliation(s)
- Maria-Teresa Piccoli
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Shashi Kumar Gupta
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Janika Viereck
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Ariana Foinquinos
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Sabine Samolovac
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Freya Luise Kramer
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Ankita Garg
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Janet Remke
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Karina Zimmer
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Sandor Batkai
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| | - Thomas Thum
- From the Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-Tx) (M.-T.P., S.K.G., J.V., A.F., S.S., F.L.K., A.G., J.R., K.Z., S.B., T.T.) and Excellence Cluster REBIRTH (M.-T.P., J.V., T.T.), Hannover Medical School, Germany; and National Heart and Lung Institute, Imperial College London, United Kingdom (T.T.)
| |
Collapse
|
154
|
Epigenome Aberrations: Emerging Driving Factors of the Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2017; 18:ijms18081774. [PMID: 28812986 PMCID: PMC5578163 DOI: 10.3390/ijms18081774] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 07/29/2017] [Accepted: 08/12/2017] [Indexed: 12/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC), the most common form of Kidney cancer, is characterized by frequent mutations of the von Hippel-Lindau (VHL) tumor suppressor gene in ~85% of sporadic cases. Loss of pVHL function affects multiple cellular processes, among which the activation of hypoxia inducible factor (HIF) pathway is the best-known function. Constitutive activation of HIF signaling in turn activates hundreds of genes involved in numerous oncogenic pathways, which contribute to the development or progression of ccRCC. Although VHL mutations are considered as drivers of ccRCC, they are not sufficient to cause the disease. Recent genome-wide sequencing studies of ccRCC have revealed that mutations of genes coding for epigenome modifiers and chromatin remodelers, including PBRM1, SETD2 and BAP1, are the most common somatic genetic abnormalities after VHL mutations in these tumors. Moreover, recent research has shed light on the extent of abnormal epigenome alterations in ccRCC tumors, including aberrant DNA methylation patterns, abnormal histone modifications and deregulated expression of non-coding RNAs. In this review, we discuss the epigenetic modifiers that are commonly mutated in ccRCC, and our growing knowledge of the cellular processes that are impacted by them. Furthermore, we explore new avenues for developing therapeutic approaches based on our knowledge of epigenome aberrations of ccRCC.
Collapse
|
155
|
He Y, Luo Y, Liang B, Ye L, Lu G, He W. Potential applications of MEG3 in cancer diagnosis and prognosis. Oncotarget 2017; 8:73282-73295. [PMID: 29069869 PMCID: PMC5641212 DOI: 10.18632/oncotarget.19931] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022] Open
Abstract
LncRNAs are emerging as integral functional and regulatory components of normal biological activities and are now considered as critically involved in the development of different diseases including cancer. In this review, we summarized recent findings on maternally expressed gene 3 (MEG3), a noncoding lncRNA, locates in the imprinted DLK1–MEG3 locus on human chromosome 14q32.3 region. MEG3 is expressed in normal tissues but is either lost or decreased in many human tumors and tumor derived cell lines. Studies have demonstrated that MEG3 is associated with cancer initiation, progression, metastasis and chemo-resistance. MEG3 may affect the activities of TP53, MDM2, GDF15, RB1 and some other key cell cycle regulators. In addition, the level of MEG3 showed good correlation with cancer clinicopathological grade. In summary, MEGs is an RNA-based tumor suppressor and is involved in the etiology, progression, and chemosensitivity of cancers. The alteration of MEG3 levels in various cancers suggested the possibility of using MEG3 level for cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Yuqing He
- Institute of Medical Systems Biology, Guangdong Medical University, Dongguan 523808, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Guangdong Medical University, Dongguan 523808, China
| | - Yanhong Luo
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Biyu Liang
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Lei Ye
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Guangxing Lu
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Weiming He
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
156
|
He JH, Han ZP, Liu JM, Zhou JB, Zou MX, Lv YB, Li YG, Cao MR. Overexpression of Long Non-Coding RNA MEG3 Inhibits Proliferation of Hepatocellular Carcinoma Huh7 Cells via Negative Modulation of miRNA-664. J Cell Biochem 2017; 118:3713-3721. [PMID: 28374914 DOI: 10.1002/jcb.26018] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/03/2017] [Indexed: 12/30/2022]
Abstract
Evidence is accumulating that long non-coding RNAs (lncRNAs) are involved in human tumorigenesis and dysregulated in many cancers, including hepatocellular carcinoma (HCC). Because lncRNAs can regulate essential pathways that contribute to tumor initiation and progression with their tissue specificity, lncRNAs are valuable biomarkers and therapeutic targets. Maternally expressed gene 3 (MEG3) is a lncRNA overexpressed in HCC cells that inhibits HCC progression, however, the mechanism remains largely unknown. Recently, a novel regulatory mechanism has been proposed in which RNAs can cross-talk with each other via competing for shared microRNAs (miRNAs). The proposed competitive endogenous RNAs could mediate the bioavailability of miRNAs on their targets, thus imposing another level of post-transcriptional regulation. In the current study, we demonstrated that MEG3 is down-regulated in HCC tissues. MEG3 over-expression imposes another level of post-transcriptional regulation, whereas MEG3 overexpression increase the expression of the miR-664 target gene, ADH4, through competitive "sponging" miR-664. In addition, NF-κB may affect transcription of MEG3 by directly binding to the promoter region. Our data revealed that NF-κB may affect the transcript of MEG3. MEG3 overexpression inhibited the proliferation of HCC cells, at least in part by affecting miR-664mediated regulation of ADH4. Together, these results suggest that MEG3 is a suppressor of tumor which acts in part through "sponging" miR-664. J. Cell. Biochem. 118: 3713-3721, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jin-Hua He
- Department of Laboratory, Central Hospital of Panyu District, Guangzhou, Guangdong 511400, China
| | - Ze-Ping Han
- Department of Laboratory, Central Hospital of Panyu District, Guangzhou, Guangdong 511400, China
| | - Ji-Ming Liu
- Department of General Surgery, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Jia-Bin Zhou
- Department of Laboratory, Central Hospital of Panyu District, Guangzhou, Guangdong 511400, China
| | - Mao-Xian Zou
- Department of Laboratory, Central Hospital of Panyu District, Guangzhou, Guangdong 511400, China
| | - Yu-Bing Lv
- Department of Laboratory, Central Hospital of Panyu District, Guangzhou, Guangdong 511400, China
| | - Yu-Guang Li
- Department of Laboratory, Central Hospital of Panyu District, Guangzhou, Guangdong 511400, China
| | - Ming-Rong Cao
- Department of General Surgery, First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| |
Collapse
|
157
|
Sahin Y, Altan Z, Arman K, Bozgeyik E, Koruk Ozer M, Arslan A. Inhibition of miR-664a interferes with the migration of osteosarcoma cells via modulation of MEG3. Biochem Biophys Res Commun 2017; 490:1100-1105. [DOI: 10.1016/j.bbrc.2017.06.174] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 06/28/2017] [Indexed: 12/22/2022]
|
158
|
Enterina JR, Enfield KSS, Anderson C, Marshall EA, Ng KW, Lam WL. DLK1-DIO3 imprinted locus deregulation in development, respiratory disease, and cancer. Expert Rev Respir Med 2017; 11:749-761. [PMID: 28715922 DOI: 10.1080/17476348.2017.1355241] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION The imprinted DLK1-DIO3 locus at 14q32.1-32.31 holds biological significance in fetal development, whereby imprinting errors are causal to developmental disorders. Emerging evidence has implicated this locus in other diseases including cancer, highlighting the biological parallels between fetal organ and tumour development. Areas covered: Controlled regulation of gene expression from the imprinted DLK1-DIO3 locus at 14q32.1-32.31 is crucial for proper fetal development. Deregulation of locus gene expression due to imprinting errors has been mechanistically linked to the developmental disorders Kagami-Ogata Syndrome and Temple Syndrome. In adult tissues, deregulation of locus genes has been associated with multiple malignancies although the causal genetic mechanisms remain largely uncharacterised. Here, we summarize the genetic mechanisms underlying the developmental disorders that arise as a result of improper locus imprinting and the resulting developmental phenotypes, emphasizing both the coding and noncoding components of the locus. We further highlight biological parallels common to both fetal development and disease, with a specific focus on lung development, respiratory disease, and lung cancer. Expert commentary: Many commonalities between respiratory and developmental defects have emerged with respect to the 14q32 locus, emphasizing the importance of studying the effects of imprinting on gene regulation patterns at this locus in both biological settings.
Collapse
Affiliation(s)
- Jhon R Enterina
- a British Columbia Cancer Research Centre , Vancouver , BC , Canada
| | | | | | - Erin A Marshall
- a British Columbia Cancer Research Centre , Vancouver , BC , Canada
| | - Kevin W Ng
- a British Columbia Cancer Research Centre , Vancouver , BC , Canada
| | - Wan L Lam
- a British Columbia Cancer Research Centre , Vancouver , BC , Canada
| |
Collapse
|
159
|
Li L, Shang J, Zhang Y, Liu S, Peng Y, Zhou Z, Pan H, Wang X, Chen L, Zhao Q. MEG3 is a prognostic factor for CRC and promotes chemosensitivity by enhancing oxaliplatin-induced cell apoptosis. Oncol Rep 2017; 38:1383-1392. [PMID: 28731151 PMCID: PMC5549059 DOI: 10.3892/or.2017.5828] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
A major reason for the failure of advanced colorectal cancer (CRC) treatment is the occurrence of chemoresistance to oxaliplatin-based chemotherapy. Recently, studies have shown that long non-coding RNAs (lncRNAs) play an important role in drug resistance. Using HiSeq sequencing methods, we identified that lncRNAs show differential expression levels in oxaliplatin-resistant (OxR) and non-resistant CRC patients. RT-qPCR was then performed in tissues and serum samples, and lncRNA MEG3 was verified to be downregulated in non-responding patients and to have considerable discriminating potential to identify responding patients from non-responding patients. Moreover, decreased serum MEG3 expression was associated with poor chemoresponse and low survival rate in CRC patients receiving oxaliplatin treatment. Subsequently, OxR cell lines were established, and MEG3 was significantly downregulated in HT29 OxR and SW480 OxR cells. In addition, overexpression of MEG3 with pMEG3 reversed oxaliplatin resistance in both CRC cell lines. Flow cytometric apoptosis analysis indicated that MEG3 promoted CRC cell apoptosis. More importantly, MEG3 enhanced oxaliplatin‑induced cell cytotoxicity in CRC. In conclusion, our integrated approach demonstrated that decreased expression of lncRNA MEG3 in CRC confers potent poor therapeutic efficacy, and that MEG3 promotes chemosensitivity by enhancing oxaliplatin-induced cell apoptosis. Thus, overexpression of MEG3 may be a future direction by which to develop a novel therapeutic strategy to overcome oxaliplatin resistance of CRC patients.
Collapse
Affiliation(s)
- Lixia Li
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Jian Shang
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Yupeng Zhang
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Shi Liu
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Yanan Peng
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Zhou Zhou
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Huaqing Pan
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Xiaobing Wang
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Lipng Chen
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| | - Qiu Zhao
- Department of Gastroenterology/Hepatology, Zhongnan Hospital of Wuhan University, The Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
160
|
Bhan A, Soleimani M, Mandal SS. Long Noncoding RNA and Cancer: A New Paradigm. Cancer Res 2017; 77:3965-3981. [PMID: 28701486 DOI: 10.1158/0008-5472.can-16-2634] [Citation(s) in RCA: 2011] [Impact Index Per Article: 287.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 04/05/2017] [Accepted: 05/04/2017] [Indexed: 12/11/2022]
Abstract
In addition to mutations or aberrant expression in the protein-coding genes, mutations and misregulation of noncoding RNAs, in particular long noncoding RNAs (lncRNA), appear to play major roles in cancer. Genome-wide association studies of tumor samples have identified a large number of lncRNAs associated with various types of cancer. Alterations in lncRNA expression and their mutations promote tumorigenesis and metastasis. LncRNAs may exhibit tumor-suppressive and -promoting (oncogenic) functions. Because of their genome-wide expression patterns in a variety of tissues and their tissue-specific expression characteristics, lncRNAs hold strong promise as novel biomarkers and therapeutic targets for cancer. In this article, we have reviewed the emerging functions and association of lncRNAs in different types of cancer and discussed their potential implications in cancer diagnosis and therapy. Cancer Res; 77(15); 3965-81. ©2017 AACR.
Collapse
Affiliation(s)
- Arunoday Bhan
- Gene Regulation and Epigenetics Research Lab, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas
| | - Milad Soleimani
- Gene Regulation and Epigenetics Research Lab, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas
| | - Subhrangsu S Mandal
- Gene Regulation and Epigenetics Research Lab, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas.
| |
Collapse
|
161
|
Kumar MM, Goyal R. LncRNA as a Therapeutic Target for Angiogenesis. Curr Top Med Chem 2017; 17:1750-1757. [PMID: 27848894 DOI: 10.2174/1568026617666161116144744] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/25/2016] [Accepted: 09/21/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Out of 3 billion base pairs in human genome only ~2% code for proteins; and out of 180,000 transcripts in human cells, about 20,000 code for protein, remaining 160,000 are non-coding transcripts. Most of these transcripts are more than 200 base pairs and constitute a group of long non-coding RNA (lncRNA). Many of the lncRNA have its own promoter, and are well conserved in mammals. Accumulating evidence indicates that lncRNAs act as molecular switches in cellular differentiation, movement, apoptosis, and in the reprogramming of cell states by altering gene expression patterns. However, the role of this important group of molecules in angiogenesis is not well understood. Angiogenesis is a complex process and depends on precise regulation of gene expression. CONCLUSION Dysregulation of transcription during this process may lead to several diseases including various cancers. As angiogenesis is an important process in cancer pathogenesis and treatment, lncRNA may be playing an important role in angiogenesis. In support of this, lncRNA microvascular invasion in hepatocellular carcinoma (MVIH) has been shown to activate angiogenesis. Furthermore, lncRNA-Meg3-knockout mouse showed increased expression of vascular endothelial growth factor pathway genes and increased cortical microvessel density. Overall, there is strong evidence that lncRNA is an important class of regulatory molecule, and a number of studies have demonstrated that these can be targeted to change cellular physiology and functions. In this review, we have attempted to summarize these studies and elucidate the potential of this novel regulatory molecule as a therapeutic target.
Collapse
Affiliation(s)
- Mohan M Kumar
- Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, United States
| | - Ravi Goyal
- Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University, School of Medicine, Loma Linda, CA 92350, United States
| |
Collapse
|
162
|
Zhou C, Huang C, Wang J, Huang H, Li J, Xie Q, Liu Y, Zhu J, Li Y, Zhang D, Zhu Q, Huang C. LncRNA MEG3 downregulation mediated by DNMT3b contributes to nickel malignant transformation of human bronchial epithelial cells via modulating PHLPP1 transcription and HIF-1α translation. Oncogene 2017; 36:3878-3889. [PMID: 28263966 PMCID: PMC5525547 DOI: 10.1038/onc.2017.14] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/11/2016] [Accepted: 12/27/2016] [Indexed: 02/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) are emerging as key factors in various fundamental cellular biological processes, and many of them are likely to have functional roles in tumorigenesis. Maternally expressed gene 3 (MEG3) is an imprinted gene located at 14q32 that encodes a lncRNA, and the decreased MEG3 expression has been reported in multiple cancer tissues. However, nothing is known about the alteration and role of MEG3 in environmental carcinogen-induced lung tumorigenesis. Our present study, for the first time to the best of our knowledge, discovered that environmental carcinogen nickel exposure led to MEG3 downregulation, consequently initiating c-Jun-mediated PHLPP1 transcriptional inhibition and hypoxia-inducible factor-1α (HIF-1α) protein translation upregulation, in turn resulting in malignant transformation of human bronchial epithelial cells. Mechanistically, MEG3 downregulation was attributed to nickel-induced promoter hypermethylation via elevating DNMT3b expression, whereas PHLPP1 transcriptional inhibition was due to the decreasing interaction of MEG3 with its inhibitory transcription factor c-Jun. Moreover, HIF-1α protein translation was upregulated via activating the Akt/p70S6K/S6 axis resultant from PHLPP1 inhibition in nickel responses. Collectively, we uncover that nickel exposure results in DNMT3b induction and MEG3 promoter hypermethylation and expression inhibition, further reduces its binding to c-Jun and in turn increasing c-Jun inhibition of PHLPP1 transcription, leading to the Akt/p70S6K/S6 axis activation, and HIF-1α protein translation, as well as malignant transformation of human bronchial epithelial cells. Our studies provide a significant insight into understanding the alteration and role of MEG3 in nickel-induced lung tumorigenesis.
Collapse
MESH Headings
- Adenocarcinoma/enzymology
- Adenocarcinoma/pathology
- Adenocarcinoma of Lung
- Bronchi/pathology
- Carcinogens/toxicity
- Carcinoma, Squamous Cell/enzymology
- Carcinoma, Squamous Cell/pathology
- Cell Line
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/metabolism
- DNA (Cytosine-5-)-Methyltransferases/physiology
- Down-Regulation
- Epithelial Cells/enzymology
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lung Neoplasms/enzymology
- Lung Neoplasms/pathology
- Nickel/toxicity
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phosphoprotein Phosphatases/genetics
- Phosphoprotein Phosphatases/metabolism
- Promoter Regions, Genetic
- Protein Biosynthesis
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Transcription, Genetic
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- Chengfan Zhou
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Chao Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Jingjing Wang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Qipeng Xie
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yu Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Junlan Zhu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Yang Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Dongyun Zhang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Qixing Zhu
- Department of Occupational and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Chuanshu Huang
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| |
Collapse
|
163
|
Ma B, Gao Z, Lou J, Zhang H, Yuan Z, Wu Q, Li X, Zhang B. Long non-coding RNA MEG3 contributes to cisplatin-induced apoptosis via inhibition of autophagy in human glioma cells. Mol Med Rep 2017; 16:2946-2952. [DOI: 10.3892/mmr.2017.6897] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 04/06/2017] [Indexed: 11/05/2022] Open
|
164
|
Ding Y, Yang DZ, Zhai YN, Xue K, Xu F, Gu XY, Wang SM. Microarray expression profiling of long non-coding RNAs in epithelial ovarian cancer. Oncol Lett 2017; 14:2523-2530. [PMID: 28781691 DOI: 10.3892/ol.2017.6448] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/13/2017] [Indexed: 12/17/2022] Open
Abstract
Although numerous long non-coding RNAs (lncRNAs) have been identified to be important in human cancer, their potential regulatory roles in epithelial tumorigenesis and tumor progression in ovarian cancer remain unclear. The purpose of the present study was to investigate lncRNAs that were differentially expressed (DE) in epithelial ovarian cancer and to explore their potential functions. The lncRNA profiles in five pairs of human epithelial ovarian cancer tissues and their adjacent normal tissues were described using microarrays. The results of the microarray analysis revealed that 672 upregulated and 549 downregulated (fold-change ≥2.0) lncRNAs were DE between the cancerous and normal tissues. Reverse transcription-quantitative polymerase chain reaction was used to validate the microarray results using four upregulated (RP11-1C1.7, XLOC_003286, growth arrest-specific 5 and ZNF295-AS1) and four downregulated (protein tyrosine kinase 7, maternally expressed gene 3, AC079776.2 and ribosomal protein lateral stalk subunit P0 pseudogene 2) lncRNAs. Furthermore, gene ontology and pathway analyses were used to carry out functional analyses of the candidate genes of DE lncRNAs. The results identified lncRNAs with significantly altered expression profiles in human epithelial ovarian cancer cells compared with those in adjacent normal cells. These data offer new insights into the occurrence and development of epithelial ovarian cancer, and these lncRNAs may provide novel molecular biomarkers for further research on epithelial ovarian cancer.
Collapse
Affiliation(s)
- Ye Ding
- State Key Laboratory of Reproductive Medicine, Department of Endoscopic Diagnostic and Treatment Center, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Da-Zheng Yang
- State Key Laboratory of Reproductive Medicine, Department of Endoscopic Diagnostic and Treatment Center, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Yong-Ning Zhai
- State Key Laboratory of Reproductive Medicine, Department of Endoscopic Diagnostic and Treatment Center, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Kai Xue
- State Key Laboratory of Reproductive Medicine, Department of Endoscopic Diagnostic and Treatment Center, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Feng Xu
- State Key Laboratory of Reproductive Medicine, Department of Endoscopic Diagnostic and Treatment Center, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Xiao-Yan Gu
- State Key Laboratory of Reproductive Medicine, Department of Endoscopic Diagnostic and Treatment Center, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Su-Min Wang
- State Key Laboratory of Reproductive Medicine, Department of Endoscopic Diagnostic and Treatment Center, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| |
Collapse
|
165
|
Intersecting transcriptomic profiling technologies and long non-coding RNA function in lung adenocarcinoma: discovery, mechanisms, and therapeutic applications. Oncotarget 2017; 8:81538-81557. [PMID: 29113413 PMCID: PMC5655308 DOI: 10.18632/oncotarget.18432] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/13/2017] [Indexed: 02/07/2023] Open
Abstract
Previously thought of as junk transcripts and pseudogene remnants, long non-coding RNAs (lncRNAs) have come into their own over the last decade as an essential component of cellular activity, regulating a plethora of functions within multicellular organisms. lncRNAs are now known to participate in development, cellular homeostasis, immunological processes, and the development of disease. With the advent of next generation sequencing technology, hundreds of thousands of lncRNAs have been identified. However, movement beyond mere discovery to the understanding of molecular processes has been stymied by the complicated genomic structure, tissue-restricted expression, and diverse regulatory roles lncRNAs play. In this review, we will focus on lncRNAs involved in lung cancer, the most common cause of cancer-related death in the United States and worldwide. We will summarize their various methods of discovery, provide consensus rankings of deregulated lncRNAs in lung cancer, and describe in detail the limited functional analysis that has been undertaken so far.
Collapse
|
166
|
He C, Yang W, Yang J, Ding J, Li S, Wu H, Zhou F, Jiang Y, Teng L, Yang J. Long Noncoding RNAMEG3Negatively Regulates Proliferation and Angiogenesis in Vascular Endothelial Cells. DNA Cell Biol 2017; 36:475-481. [PMID: 28418724 DOI: 10.1089/dna.2017.3682] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Chao He
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Wei Yang
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Jun Yang
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Jiawang Ding
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Song Li
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Hui Wu
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Fei Zhou
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Yurong Jiang
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Lin Teng
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| | - Jian Yang
- Institute of Cardiology, China Three Gorges University, Hubei Province, People's Republic of China
| |
Collapse
|
167
|
Yao H, Sun P, Duan M, Lin L, Pan Y, Wu C, Fu X, Wang H, Guo L, Jin T, Ding Y. microRNA-22 can regulate expression of the long non-coding RNA MEG3 in acute myeloid leukemia. Oncotarget 2017; 8:65211-65217. [PMID: 29029424 PMCID: PMC5630324 DOI: 10.18632/oncotarget.18059] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/19/2017] [Indexed: 11/25/2022] Open
Abstract
Aim Acute myeloid leukemia (AML) is the most common blood tumor with poor prognosis. At present, the research found that the pathogenesis of AML is related to many factors, such as recurrent somatic mutations and gene expression and epigenetic changes, however, the molecular mechanism of AML is still unclear. Long non-coding RNA MEG3 is a newly found tumor suppressor and plays a very important role in the regulation of a variety of tumor formation and progression. Studies found that the MEG3 expression was significantly decreased in AML. However, to date, it is not clear the cause of its abnormal expression. Therefore, the molecular mechanism of AML is urgently needed to study the molecular mechanism of AML. Methods The different expression level of MEG3, TET2, miR-22-3p, miR-22-5p in AML was detected by real-time quantification PCR. MEG3, TET2, miR-22-3p, miR-22-3p expression cell pools in K562 cells was used to interfering and TET2, MEG3 TET2, relations with miR-22-3p, miR-22-5p. The effect of AML cell on proliferation was evaluated by TET2 lower expression. Results 1. The lower expression of MEG3 and TET2 in AML cell lines was detected by RT-qPCR. 2. The stable MEG3, TET2 overexpression cell pools in K562 cells was successful established. 3. After transfection, MTT assay revealed that cell growth was significantly increased in AML cell lines transfected with TET2 compared with controls. Conclusions Our findings suggested that MEG3 is significantly down regulated in AML cell lines.
Collapse
Affiliation(s)
- Hongxia Yao
- Department of Hematology, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Pei Sun
- Department of Hematology, Hunan Yiyang Central Hospital, Yiyang, Hunan 413000, P.R. China
| | - Mengling Duan
- Department of Hematology, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Lie Lin
- Department of Hematology, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Yanping Pan
- Department of Hematology, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Congming Wu
- Department of Hematology, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Xiangjun Fu
- Department of Hematology, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Hua Wang
- Department of Hematology, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Li Guo
- Department of Hematology, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Tianbo Jin
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, Shaanxi 710069, P.R. China
| | - Yipeng Ding
- Department of Emergency, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
168
|
Kaneda M, Takahashi M, Yamanaka KI, Saito K, Taniguchi M, Akagi S, Watanabe S, Nagai T. Epigenetic analysis of bovine parthenogenetic embryonic fibroblasts. J Reprod Dev 2017; 63:365-375. [PMID: 28484201 PMCID: PMC5593088 DOI: 10.1262/jrd.2017-040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Although more than 100 imprinted genes have already been identified in the mouse and human genomes, little is known about genomic imprinting in cattle. For a better understanding of these genes in cattle, parthenogenetically activated bovine blastocysts were transferred to recipient cows to obtain parthenotes, and fibroblasts derived from a Day 40 (Day 0 being the day of parthenogenetic activation) parthenogenetic embryo (BpEFs) were successfully obtained. Bovine embryonic fibroblasts (BEFs) were also isolated from a normal fertilized embryo obtained from an artificially inseminated cow. The expression of imprinted genes was analyzed by RT-PCR. Paternally expressed genes (PEGs) in mouse (viz., IGF2, PEG3, ZAC1, NDN, DLK1, SGCE, and PEG10) were expressed in BEFs, but not in BpEFs, suggesting that these genes are also imprinted in cattle. However, other PEGs in mouse (viz., IMPACT, MAGEL2, SNRPN, and PEG1/MEST) were expressed in both BEFs and BpEFs. These genes may not be imprinted in BEFs. The expression of seven maternally expressed genes in mouse was also analyzed, and only CDKN1C was not expressed in BpEFs. The DNA methylation patterns of repetitive elements (Satellite I, Satellite II, alpha-satellite, and Art2) were not different between the BEFs and BpEFs; however, the differentially methylated region (DMR) of paternally methylated H19 was hypomethylated, whereas those of maternally methylated PEG3 and PEG10 were hypermethylated in BpEFs, as expected. The methylation of the SNRPN DMR was not different between the BEFs and BpEFs, in accordance with the SNRPN expression levels in both cell types. The XIST gene, which is essential for X chromosome inactivation in females, was expressed in BpEFs, whereas its DMR was half-methylated, suggesting that X chromosome inactivation is normal in these cells. Microarray analysis was also applied to identify novel PEGs that should be expressed only in BEFs but not in BpEFs. More than 300 PEG candidate genes, including IGF2, PEG3, and PEG10, were obtained. These results illustrate the epigenetic characteristic of bovine parthenogenetic embryos and contribute to the identification of novel imprinted genes in cattle.
Collapse
Affiliation(s)
- Masahiro Kaneda
- Division of Animal Life Science, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Masashi Takahashi
- Department of Animal Science, Graduate School of Agriculture, Hokkaido University, Hokkaido 060-8589, Japan
| | | | - Koji Saito
- Kumamoto Prefectural Agriculture Research Center, Kumamoto 861-1113, Japan
| | - Masanori Taniguchi
- Kumamoto Prefectural Agriculture Research Center, Kumamoto 861-1113, Japan
| | - Satoshi Akagi
- Animal Breeding and Reproduction Research Division, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Ibaraki 305-0901, Japan
| | - Shinya Watanabe
- Animal Breeding and Reproduction Research Division, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Ibaraki 305-0901, Japan
| | - Takashi Nagai
- Headquarters, National Agriculture and Food Research Organization, Ibaraki 305-8517, Japan
| |
Collapse
|
169
|
Huang ZL, Chen RP, Zhou XT, Zhan HL, Hu MM, Liu B, Wu GD, Wu LF. Long non-coding RNA MEG3 induces cell apoptosis in esophageal cancer through endoplasmic reticulum stress. Oncol Rep 2017; 37:3093-3099. [PMID: 28405686 DOI: 10.3892/or.2017.5568] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/27/2017] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play important roles in diverse biological processes, such as cell growth, apoptosis and migration. Although downregulation of lncRNA MEG3 has been identified in several cancers, little is known about its role in esophageal squamous cell carcinoma (ESCC). The aim of the present study was to detect MEG3 expression in clinical ESCC tissues, investigate its biological functions and the endoplasmic reticulum (ER) stress-relative mechanism. MEG3 expression levels were detected by qRT-PCR in both tumor tissues and adjacent non-tumor tissues from 28 ESCC patients. PcDNA3.1-MEG3 recombinant plasmids were constructed and transfected to EC109 cells. Cell growth was analyzed by CCK-8 assay. Cell apoptosis was analyzed by fluorescence microscope and Annexin V/PI assay. The protein expression was determined by western blot analysis. The results showed that MEG3 decreased significantly in ESCC tissues relative to adjacent normal tissues. PcDNA3.1-MEG3 plasmids were successfully constructed and the expression level of MEG3 significantly increased after MEG3 transfection to EC109 cells. Ectopic expression of MEG3 inhibited EC109 cell proliferation and induced apoptosis in vitro. MEG3 overexpression increased the expression of ER stress‑related proteins (GRP78, IRE1, PERK, ATF6, CHOP and cleaved‑caspase-3). Our results first demonstrate that MEG3 is downregulated in ESCC tissues. MEG3 was able to inhibit cell growth and induced apoptosis in EC109 cells, most probably via activation of the ER stress pathway.
Collapse
Affiliation(s)
- Zhen-Lun Huang
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Rui-Pei Chen
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xiao-Tao Zhou
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Hao-Lian Zhan
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Min-Min Hu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Bin Liu
- Department of Neurosurgery, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guan-Di Wu
- First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ling-Fei Wu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
170
|
Chen Y, Zhou J. LncRNAs: macromolecules with big roles in neurobiology and neurological diseases. Metab Brain Dis 2017; 32:281-291. [PMID: 28161776 DOI: 10.1007/s11011-017-9965-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 01/31/2017] [Indexed: 01/05/2023]
Abstract
Long noncoding RNAs (lncRNAs) are recently defined as thousands of RNA molecules longer than 200 nucleotides and lacking an appreciable open reading frame in mammals. Although lncRNAs lack protein-coding function, they play critical roles in the regulation of almost all the protein-coding genes in a cell at various stages including chromatin modification, transcription and post-transcriptional processing. It is thus not surprising that lncRNAs may be the crucial regulators in the normal development, physiology and pathology. LncRNAs in neuroscience is a novel research field. Interestingly, recent studies have demonstrated that many lncRNAs are highly expressed in brain and their dysregulations occur in neurological disorders. In this review, we describe the current understanding of lncRNAs in neurobiology and neurological diseases including cerebral injury. LncRNAs could be novel biomarkers and could be potential new targets for new drugs for many neurological diseases in the future, although the related studies are still at in the early stages.
Collapse
Affiliation(s)
- Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, No.25 Taiping Street, Luzhou, Sichuan Province, 646000, People's Republic of China.
| |
Collapse
|
171
|
Abstract
Huntington's disease (HD) is a genetic, fatal autosomal dominant neurodegenerative disorder typically occurring in midlife with symptoms ranging from chorea, to dementia, to personality disturbances (Philos Trans R Soc Lond Ser B Biol Sci 354:957-961, 1999). HD is inherited in a dominant fashion, and the underlying mutation in all cases is a CAG trinucleotide repeat expansion within exon 1 of the HD gene (Cell 72:971-983, 1993). The expanded CAG repeat, translated into a lengthened glutamine tract at the amino terminus of the huntingtin protein, affects its structural properties and functional activities. The effects are pleiotropic, as huntingtin is broadly expressed in different cellular compartments (i.e., cytosol, nucleus, mitochondria) as well as in all cell types of the body at all developmental stages, such that HD pathogenesis likely starts at conception and is a lifelong process (Front Neurosci 9:509, 2015). The rate-limiting mechanism(s) of neurodegeneration in HD still remains elusive: many different processes are commonly disrupted in HD cell lines and animal models, as well as in HD patient cells (Eur J Neurosci 27:2803-2820, 2008); however, epigenetic-chromatin deregulation, as determined by the analysis of DNA methylation, histone modifications, and noncoding RNAs, has now become a prevailing feature. Thus, the overarching goal of this chapter is to discuss the current status of the literature, reviewing how an aberrant epigenetic landscape can contribute to altered gene expression and neuronal dysfunction in HD.
Collapse
|
172
|
Fan L, Hu Z. [Progress of Long Non-coding RNAs in Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 19:108-12. [PMID: 26903166 PMCID: PMC6015147 DOI: 10.3779/j.issn.1009-3419.2016.02.10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Lung cancer related death is the most common cause of cancer death worldwide. Non-coding RNA has no protein coding capacity, but can play roles in a variety of biology. Long non-coding RNA (lncRNA) is at least 200 nt long. A variety of lncRNAs could promote or inhibit tumor development and progression in many tumors, including non-small cell lung cancer (NSCLC). Due to their fundamental role in regulating gene expression, they involve in biological mechanism of the tumor, and so they are expected to be new cancer biomarkers. In this review, we emphasized the roles of lncRNAs in NSCLC, and discuss them as diagnostic and prognostic markers and as therapeutic targets.
Collapse
Affiliation(s)
- Liming Fan
- Department of Medical Clinical Laboratory, Tianjin Medical University General Hospital, 300052 Tianjin, China
| | - Zhidong Hu
- Department of Medical Clinical Laboratory, Tianjin Medical University General Hospital, 300052 Tianjin, China
| |
Collapse
|
173
|
Bian EB, Wang YY, Yang Y, Wu BM, Xu T, Meng XM, Huang C, Zhang L, Lv XW, Xiong ZG, Li J. Hotair facilitates hepatic stellate cells activation and fibrogenesis in the liver. Biochim Biophys Acta Mol Basis Dis 2016; 1863:674-686. [PMID: 27979710 DOI: 10.1016/j.bbadis.2016.12.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/25/2016] [Accepted: 12/12/2016] [Indexed: 12/18/2022]
Abstract
Long non-coding RNAs (lncRNAs) are increasingly recognized as major players in regulating various biological processes. LncRNA HOX transcript antisense RNA (Hotair) has been extensively studied in cancer. However, the role of Hotair in liver fibrosis remains unknown. Here we observed that Hotair expression was significantly increased in CCl4-induced mouse liver fibrosis models, human fibrotic livers and activated hepatic stellate cells (HSCs) by TGF-β1 stimulation. Enforced expression of Hotair in LX-2 cells promoted cell proliferation and activation while inhibition of its expression had an opposite effect. Furthermore, we found that Hotair may act as an endogenous 'sponge' of miR-148b, which regulates expression of the DNMT1/MEG3/p53 pathways in HSCs. Intriguingly, Hotair enhanced polycomb repressive complex 2 (PRC2) occupancy and histone H3K27me3 repressive marks, specifically at the MEG3 promoter region. Finally, we found that Hotair forms an RNA/DNA hybrid and recruits PRC2 to MEG3 promoter. These data suggest that Hotair inhibition may represent a promising therapeutic option for suppressing liver fibrosis.
Collapse
Affiliation(s)
- Er-Bao Bian
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Yuan-Yuan Wang
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Yang Yang
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Bao-Ming Wu
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Tao Xu
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Xiao-Ming Meng
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Cheng Huang
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Lei Zhang
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Xiong-Wen Lv
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China
| | - Zhi-Gang Xiong
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA 30310-1945, USA.
| | - Jun Li
- School of pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University (AMU), Hefei 230032, China.
| |
Collapse
|
174
|
Smekalova EM, Kotelevtsev YV, Leboeuf D, Shcherbinina EY, Fefilova AS, Zatsepin TS, Koteliansky V. lncRNA in the liver: Prospects for fundamental research and therapy by RNA interference. Biochimie 2016; 131:159-172. [DOI: 10.1016/j.biochi.2016.06.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 06/14/2016] [Indexed: 12/19/2022]
|
175
|
Yan H, Yuan J, Gao L, Rao J, Hu J. Long noncoding RNA MEG3 activation of p53 mediates ischemic neuronal death in stroke. Neuroscience 2016; 337:191-199. [DOI: 10.1016/j.neuroscience.2016.09.017] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/04/2016] [Accepted: 09/09/2016] [Indexed: 12/23/2022]
|
176
|
Chak WP, Lung RWM, Tong JHM, Chan SYY, Lun SWM, Tsao SW, Lo KW, To KF. Downregulation of long non-coding RNA MEG3 in nasopharyngeal carcinoma. Mol Carcinog 2016; 56:1041-1054. [PMID: 27597634 DOI: 10.1002/mc.22569] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/12/2016] [Accepted: 09/04/2016] [Indexed: 12/13/2022]
Abstract
In our previous whole-transcriptome sequencing analysis, downregulation of a long non-coding RNA, maternally expressed gene 3 (MEG3), was identified in NPC samples. This finding suggests the possible role of MEG3 as a tumor suppressor in this distinctive disease. In the present study, two MEG3 variants, AF119863 (MEG3-AF) and BX247998 (MEG3-BX), were found abundantly expressed in a normal nasopharyngeal epithelial cell line, NP69. Significant downregulation of MEG3-AF was further verified in a panel of NPC samples including xenografts and primary biopsies. MEG3 is an imprinted gene located within chromosome 14q32, a common deleted region in NPC. Both DNA copy number loss and aberrant promoter methylation contributed to MEG3 inactivation. Interestingly, MEG3 expression could successfully be rescued by the treatment of a demethylation agent. Besides, ectopic expression of MEG3 in NPC cell lines resulted in considerable repression of in vitro anchorage-independent growth and in vivo tumorigenicity, in addition to significant inhibition in cell proliferation, colony formation, and induction of cell cycle arrest. Finally, we revealed the association between MEG3 activity and the p53 signaling cascade. Our findings characterize MEG3 as a tumor suppressive long non-coding RNA in NPC and encourage the development of precise long non-coding RNA-targeted epigenetic therapy against this malignancy. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wing-Po Chak
- State Key Laboratory of Oncology in South China, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, HKSAR, China.,Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, HKSAR, China
| | - Raymond Wai-Ming Lung
- State Key Laboratory of Oncology in South China, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, HKSAR, China.,Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, HKSAR, China
| | - Joanna Hung-Man Tong
- State Key Laboratory of Oncology in South China, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, HKSAR, China.,Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, HKSAR, China
| | - Sylvia Yat-Yee Chan
- State Key Laboratory of Oncology in South China, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, HKSAR, China.,Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, HKSAR, China
| | - Samantha Wei-Man Lun
- State Key Laboratory of Oncology in South China, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, HKSAR, China.,Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, HKSAR, China
| | - Sai-Wah Tsao
- Li Ka Shing Faculty of Medicine, School of Biomedical Science and Center for Cancer Research, The University of Hong Kong, HKSAR, China
| | - Kwok-Wai Lo
- State Key Laboratory of Oncology in South China, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, HKSAR, China.,Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, HKSAR, China
| | - Ka-Fai To
- State Key Laboratory of Oncology in South China, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, HKSAR, China.,Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, HKSAR, China.,Partner State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, HKSAR, China
| |
Collapse
|
177
|
Gao YF, Wang ZB, Zhu T, Mao CX, Mao XY, Li L, Yin JY, Zhou HH, Liu ZQ. A critical overview of long non-coding RNA in glioma etiology 2016: an update. Tumour Biol 2016; 37:14403-14413. [DOI: 10.1007/s13277-016-5307-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/05/2016] [Indexed: 12/31/2022] Open
|
178
|
Chen RP, Huang ZL, Liu LX, Xiang MQ, Li GP, Feng JL, Liu B, Wu LF. Involvement of endoplasmic reticulum stress and p53 in lncRNA MEG3-induced human hepatoma HepG2 cell apoptosis. Oncol Rep 2016; 36:1649-57. [PMID: 27432655 DOI: 10.3892/or.2016.4919] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 05/27/2016] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play important roles in diverse biological processes. Although downregulation of lncRNA maternally expressed gene 3 (MEG3) has been identified in several types of cancers, little is known concerning its biological role and regulatory mechanism in hepatoma. Our previous studies demonstrated that MEG3 induces apoptosis in a p53-dependent manner. The aim of the present study was to determine whether endoplasmic reticulum (ER) stress is involved in MEG3‑induced apoptosis. Recombinant lentiviral vectors containing MEG3 (Lv‑MEG3) were constructed and transfected into HepG2 cells. A 3‑(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, RT‑PCR, flow cytometry, western blot analysis, immunofluorescence and immunohistochemistry were applied. Transfected HepG2 cells were also transplanted into nude mice, and the tumor growth curves were determined. The results showed that the recombinant lentivirus of MEG3 was transfected successfully into the HepG2 cells and the expression level of MEG3 was significantly increased. Ectopic expression of MEG3 inhibited HepG2 cell proliferation in vitro and in vivo, and also induced apoptosis. Ectopic expression of MEG3 increased ER stress‑related proteins 78‑kDa glucose‑regulated protein (GRP78), inositol‑requiring enzyme 1 (IRE1), RNA‑dependent protein kinase‑like ER kinase (PERK), activating transcription factor 6 (ATF6), C/EBP homologous protein (CHOP), caspase‑3, as well as p53 and NF‑κB expression accompanied by NF‑κB translocation from the cytoplasm to the nucleus. Furthermore, inhibition of NF‑κB with Bay11‑7082 decreased p53 expression in the MEG3‑transfected cells. These results indicate that MEG3 inhibits cell proliferation and induces apoptosis, partially via the activation of the ER stress and p53 pathway, in which NF‑κB signaling is required for p53 activation in ER stress.
Collapse
Affiliation(s)
- Rui-Pei Chen
- Department of Gastroenterology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Zhen-Lun Huang
- Department of Gastroenterology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Li-Xuan Liu
- Department of Gastroenterology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Meng-Qi Xiang
- Department of Gastroenterology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guo-Ping Li
- Department of Gastroenterology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Jia-Lin Feng
- Department of Information, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Bin Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Ling-Fei Wu
- Department of Gastroenterology, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
179
|
Downregulated long non-coding RNA MEG3 in breast cancer regulates proliferation, migration and invasion by depending on p53’s transcriptional activity. Biochem Biophys Res Commun 2016; 478:323-329. [DOI: 10.1016/j.bbrc.2016.05.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 05/06/2016] [Indexed: 11/23/2022]
|
180
|
Wang T, Ma S, Qi X, Tang X, Cui D, Wang Z, Chi J, Li P, Zhai B. Long noncoding RNA ZNFX1-AS1 suppresses growth of hepatocellular carcinoma cells by regulating the methylation of miR-9. Onco Targets Ther 2016; 9:5005-14. [PMID: 27574442 PMCID: PMC4990377 DOI: 10.2147/ott.s103329] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Many long noncoding RNAs have been reported to play pivotal roles in cancer biology. Among them, the long noncoding RNA ZNFX1-AS1 has been confirmed to function in breast cancer progression, but the role of ZNFX1-AS1 in hepatocellular carcinoma (HCC) growth and the related molecular mechanisms still remains unknown. In the present study, we first identified the expression of ZNFX1-AS1 in HCC patients' specimens and HCC cell lines through quantitative reverse transcription polymerase chain reaction. Next, the effects of ZNFX1-AS1 on HCC cell growth and apoptosis were analyzed. MTT assay was used to measure the cell numbers, and fluorescence-activated cell sorting analysis was performed to evaluate cell apoptosis. Finally, the relationship between ZNFX1-AS1 and miR-9 in HCC was studied. Our results suggest that ZNFX1-AS1 was markedly downregulated in HCC samples and cell lines. Overexpression of ZNFX1-AS1 inhibited the cell proliferation and colony formation in HCC cell lines and also induced HCC cell apoptosis. Additionally, miR-9 was lowly expressed in HCC tissues and positively correlated with ZNFX1-AS1 expression. Meanwhile, significant upregulation of miR-9 and downregulation of the methylation of miR-9 promoter CpG island were observed when ZNFX1-AS1 was overexpressed. In summary, our results indicate that ZNFX1-AS1 plays a vital role in HCC progression via regulating the methylation of miR-9 and may be a potential tumor suppressor.
Collapse
Affiliation(s)
- Tao Wang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Sicong Ma
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xingxing Qi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xiaoyin Tang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Zhi Wang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jiachang Chi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Ping Li
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Bo Zhai
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
181
|
Abstract
Despite great progress in research and treatment options, lung cancer remains the leading cause of cancer-related deaths worldwide. Oncogenic driver mutations in protein-encoding genes were defined and allow for personalized therapies based on genetic diagnoses. Nonetheless, diagnosis of lung cancer mostly occurs at late stages, and chronic treatment is followed by a fast onset of chemoresistance. Hence, there is an urgent need for reliable biomarkers and alternative treatment options. With the era of whole genome and transcriptome sequencing technologies, long noncoding RNAs emerged as a novel class of versatile, functional RNA molecules. Although for most of them the mechanism of action remains to be defined, accumulating evidence confirms their involvement in various aspects of lung tumorigenesis. They are functional on the epigenetic, transcriptional, and posttranscriptional level and are regulators of pathophysiological key pathways including cell growth, apoptosis, and metastasis. Long noncoding RNAs are gaining increasing attention as potential biomarkers and a novel class of druggable molecules. It has become clear that we are only beginning to understand the complexity of tumorigenic processes. The clinical integration of long noncoding RNAs in terms of prognostic and predictive biomarker signatures and additional cancer targets could provide a chance to increase the therapeutic benefit. Here, we review the current knowledge about the expression, regulation, biological function, and clinical relevance of long noncoding RNAs in lung cancer.
Collapse
Affiliation(s)
- Anna Roth
- Division of RNA Biology and Cancer, German Cancer Research Center (DKFZ) and Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 280 (B150), 69120, Heidelberg, Germany
| | - Sven Diederichs
- Division of RNA Biology and Cancer, German Cancer Research Center (DKFZ) and Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 280 (B150), 69120, Heidelberg, Germany.
| |
Collapse
|
182
|
Seles M, Hutterer GC, Kiesslich T, Pummer K, Berindan-Neagoe I, Perakis S, Schwarzenbacher D, Stotz M, Gerger A, Pichler M. Current Insights into Long Non-Coding RNAs in Renal Cell Carcinoma. Int J Mol Sci 2016; 17:573. [PMID: 27092491 PMCID: PMC4849029 DOI: 10.3390/ijms17040573] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/08/2016] [Accepted: 04/12/2016] [Indexed: 02/07/2023] Open
Abstract
Renal cell carcinoma (RCC) represents a deadly disease with rising mortality despite intensive therapeutic efforts. It comprises several subtypes in terms of distinct histopathological features and different clinical presentations. Long non-coding RNAs (lncRNAs) are non-protein-coding transcripts in the genome which vary in expression levels and length and perform diverse functions. They are involved in the inititation, evolution and progression of primary cancer, as well as in the development and spread of metastases. Recently, several lncRNAs were described in RCC. This review emphasises the rising importance of lncRNAs in RCC. Moreover, it provides an outlook on their therapeutic potential in the future.
Collapse
Affiliation(s)
- Maximilian Seles
- Department of Urology, Medical University of Graz, A-8036 Graz, Austria.
| | - Georg C Hutterer
- Department of Urology, Medical University of Graz, A-8036 Graz, Austria.
| | - Tobias Kiesslich
- Department of Internal Medicine I, Salzburger Landeskliniken (SALK), Paracelsus Medical University, A-5020 Salzburg, Austria.
- Laboratory for Tumour Biology and Experimental Therapies, Institute of Physiology and Pathophysiology, Paracelsus Medical University, A-5020 Salzburg, Austria.
| | - Karl Pummer
- Department of Urology, Medical University of Graz, A-8036 Graz, Austria.
| | - Ioana Berindan-Neagoe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
- Research Center of Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.
- Department of Experimental Pathology, The Oncology Institute Ion Chiricuta, 400015 Cluj-Napoca, Romania.
| | - Samantha Perakis
- Institute of Human Genetics, Medical University of Graz, A-8036 Graz, Austria.
| | - Daniela Schwarzenbacher
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria.
| | - Michael Stotz
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria.
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria.
- Center for Biomarker Research in Medicine, Medical University of Graz, A-8036 Graz, Austria.
| | - Martin Pichler
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria.
| |
Collapse
|
183
|
Liu SJ, Nowakowski TJ, Pollen AA, Lui JH, Horlbeck MA, Attenello FJ, He D, Weissman JS, Kriegstein AR, Diaz AA, Lim DA. Single-cell analysis of long non-coding RNAs in the developing human neocortex. Genome Biol 2016; 17:67. [PMID: 27081004 PMCID: PMC4831157 DOI: 10.1186/s13059-016-0932-1] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/01/2016] [Indexed: 12/22/2022] Open
Abstract
Background Long non-coding RNAs (lncRNAs) comprise a diverse class of transcripts that can regulate molecular and cellular processes in brain development and disease. LncRNAs exhibit cell type- and tissue-specific expression, but little is known about the expression and function of lncRNAs in the developing human brain. Furthermore, it has been unclear whether lncRNAs are highly expressed in subsets of cells within tissues, despite appearing lowly expressed in bulk populations. Results We use strand-specific RNA-seq to deeply profile lncRNAs from polyadenylated and total RNA obtained from human neocortex at different stages of development, and we apply this reference to analyze the transcriptomes of single cells. While lncRNAs are generally detected at low levels in bulk tissues, single-cell transcriptomics of hundreds of neocortex cells reveal that many lncRNAs are abundantly expressed in individual cells and are cell type-specific. Notably, LOC646329 is a lncRNA enriched in single radial glia cells but is detected at low abundance in tissues. CRISPRi knockdown of LOC646329 indicates that this lncRNA regulates cell proliferation. Conclusion The discrete and abundant expression of lncRNAs among individual cells has important implications for both their biological function and utility for distinguishing neural cell types. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-0932-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Siyuan John Liu
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Tomasz J Nowakowski
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,Department of Neurology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Alex A Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,Department of Neurology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Jan H Lui
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,Department of Neurology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA.,Present Address: Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA
| | - Max A Horlbeck
- Department of Cellular and Molecular Pharmacology, San Francisco, CA, 94143, USA.,Howard Hughes Medical Institute, San Francisco, CA, 94143, USA.,California Institute for Quantitative Biomedical Research, San Francisco, CA, 94143, USA.,Center for RNA Systems Biology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Frank J Attenello
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Daniel He
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology, San Francisco, CA, 94143, USA.,Howard Hughes Medical Institute, San Francisco, CA, 94143, USA.,California Institute for Quantitative Biomedical Research, San Francisco, CA, 94143, USA.,Center for RNA Systems Biology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Arnold R Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA.,Department of Neurology, San Francisco, CA, 94143, USA.,University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Aaron A Diaz
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA. .,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA. .,University of California, San Francisco, San Francisco, CA, 94143, USA.
| | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, Ray and Dagmar Dolby Regeneration Medicine Building, 35 Medical Center Way, RMB 1037, San Francisco, CA, 94143, USA. .,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, 94143, USA. .,University of California, San Francisco, San Francisco, CA, 94143, USA. .,San Francisco Veterans Affairs Medical Center, San Francisco, CA, 94121, USA.
| |
Collapse
|
184
|
Li F, Wen X, Zhang H, Fan X. Novel Insights into the Role of Long Noncoding RNA in Ocular Diseases. Int J Mol Sci 2016; 17:478. [PMID: 27043545 PMCID: PMC4848934 DOI: 10.3390/ijms17040478] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 12/19/2022] Open
Abstract
Recent advances have suggested that long noncoding RNAs (lncRNAs) are differentially expressed in ocular tissues and play a critical role in the pathogenesis of different types of eye diseases. Here, we summarize the functions and mechanisms of known aberrantly-expressed lncRNAs and present a brief overview of relevant reports about lncRNAs in such ocular diseases as glaucoma, proliferative vitreoretinopathy (PVR), diabeticretinopathy (DR), and ocular tumors. We intend to highlight comprehensive studies that provide detailed data about the mechanisms of lncRNAs, their applications as diagnostic or prognostic biomarkers, and their potential therapeutic targets. Although our understanding of lncRNAs is still in its infancy, these examples may provide helpful insights into the methods by which lncRNAs interfere with ocular diseases.
Collapse
Affiliation(s)
- Fang Li
- Department of Ophthalmology, Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200025, China.
| | - Xuyang Wen
- Department of Ophthalmology, Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200025, China.
| | - He Zhang
- Department of Ophthalmology, Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200025, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200025, China.
| |
Collapse
|
185
|
Kour S, Rath PC. Long noncoding RNAs in aging and age-related diseases. Ageing Res Rev 2016; 26:1-21. [PMID: 26655093 DOI: 10.1016/j.arr.2015.12.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/08/2015] [Accepted: 12/01/2015] [Indexed: 12/14/2022]
Abstract
Aging is the universal, intrinsic, genetically-controlled, evolutionarily-conserved and time-dependent intricate biological process characterised by the cumulative decline in the physiological functions and their coordination in an organism after the attainment of adulthood resulting in the imbalance of neurological, immunological and metabolic functions of the body. Various biological processes and mechanisms along with altered levels of mRNAs and proteins have been reported to be involved in the progression of aging. It is one of the major risk factors in the patho-physiology of various diseases and disorders. Recently, the discovery of pervasive transcription of a vast pool of heterogeneous regulatory noncoding RNAs (ncRNAs), including small ncRNAs (sncRNAs) and long ncRNAs (lncRNAs), in the mammalian genome have provided an alternative way to study and explore the missing links in the aging process, its mechanism(s) and related diseases in a whole new dimension. The involvement of small noncoding RNAs in aging and age-related diseases have been extensively studied and recently reviewed. However, lncRNAs, whose function is far less explored in relation to aging, have emerged as a class of major regulators of genomic functions. Here, we have described some examples of known as well as novel lncRNAs that have been implicated in the progression of the aging process and age-related diseases. This may further stimulate research on noncoding RNAs and the aging process.
Collapse
Affiliation(s)
- Sukhleen Kour
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
186
|
Qiao S, Nordström K, Muijs L, Gasparoni G, Tierling S, Krause E, Walter J, Boehm U. Molecular Plasticity of Male and Female Murine Gonadotropes Revealed by mRNA Sequencing. Endocrinology 2016; 157:1082-93. [PMID: 26677881 DOI: 10.1210/en.2015-1836] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Gonadotropes in the anterior pituitary gland are of particular importance within the hypothalamic-pituitary-gonadal axis because they provide a means of communication and thus a functional link between the brain and the gonads. Recent results indicate that female gonadotropes may be organized in the form of a network that shows plasticity and adapts to the altered endocrine conditions of different physiological states. However, little is known about functional changes on the molecular level within gonadotropes during these different conditions. In this study we capitalize on a binary genetic strategy in order to fluorescently label murine gonadotrope cells. Using this mouse model allows to produce an enriched gonadotrope population using fluorescence activated cell sorting to perform mRNA sequencing. By using this strategy, we analyze and compare the expression profile of murine gonadotropes in different genders and developmental and hormonal stages. We find that gonadotropes taken from juvenile males and females, from cycling females at diestrus and at proestrus, from lactating females, and from adult males each have unique gene expression patterns with approximately 100 to approximately 500 genes expressed only in one particular stage. We also demonstrate extensive gene-expression profile changes with up to approximately 2200 differentially expressed genes when comparing female and male development, juveniles and adults, and cycling females. Differentially expressed genes were significantly enriched in the GnRH signaling, calcium signaling, and MAPK signaling pathways by Kyoto Encyclopedia of Genes and Genomes analysis. Our data provide an unprecedented molecular view of the primary gonadotropes and reveal a high degree of molecular plasticity within the gonadotrope population.
Collapse
Affiliation(s)
- Sen Qiao
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Karl Nordström
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Leon Muijs
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Gilles Gasparoni
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Sascha Tierling
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Elmar Krause
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Jörn Walter
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| |
Collapse
|
187
|
McFadden EJ, Hargrove AE. Biochemical Methods To Investigate lncRNA and the Influence of lncRNA:Protein Complexes on Chromatin. Biochemistry 2016; 55:1615-30. [PMID: 26859437 DOI: 10.1021/acs.biochem.5b01141] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Long noncoding RNAs (lncRNAs), defined as nontranslated transcripts greater than 200 nucleotides in length, are often differentially expressed throughout developmental stages, tissue types, and disease states. The identification, visualization, and suppression/overexpression of these sequences have revealed impacts on a wide range of biological processes, including epigenetic regulation. Biochemical investigations on select systems have revealed striking insight into the biological roles of lncRNAs and lncRNA:protein complexes, which in turn prompt even more unanswered questions. To begin, multiple protein- and RNA-centric technologies have been employed to isolate lncRNA:protein and lncRNA:chromatin complexes. LncRNA interactions with the multi-subunit protein complex PRC2, which acts as a transcriptional silencer, represent some of the few cases where the binding affinity, selectivity, and activity of a lncRNA:protein complex have been investigated. At the same time, recent reports of full-length lncRNA secondary structures suggest the formation of complex structures with multiple independent folding domains and pave the way for more detailed structural investigations and predictions of lncRNA three-dimensional structure. This review will provide an overview of the methods and progress made to date as well as highlight new methods that promise to further inform the molecular recognition, specificity, and function of lncRNAs.
Collapse
Affiliation(s)
- Emily J McFadden
- Department of Biochemistry, Duke University Medical Center , Durham, North Carolina 27710, United States
| | - Amanda E Hargrove
- Department of Biochemistry, Duke University Medical Center , Durham, North Carolina 27710, United States.,Department of Chemistry, Duke University , 124 Science Drive, Durham, North Carolina 27708, United States
| |
Collapse
|
188
|
Expression and mechanisms of long non-coding RNA genes MEG3 and ANRIL in gallbladder cancer. Tumour Biol 2016; 37:9875-86. [DOI: 10.1007/s13277-016-4863-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/13/2016] [Indexed: 01/17/2023] Open
|
189
|
Age-Related Expression of a Repeat-Rich Intergenic Long Noncoding RNA in the Rat Brain. Mol Neurobiol 2016; 54:639-660. [DOI: 10.1007/s12035-015-9634-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 12/15/2015] [Indexed: 12/14/2022]
|
190
|
Liu LX, Deng W, Zhou XT, Chen RP, Xiang MQ, Guo YT, Pu ZJ, Li R, Wang GF, Wu LF. The mechanism of adenosine-mediated activation of lncRNA MEG3 and its antitumor effects in human hepatoma cells. Int J Oncol 2016; 48:421-9. [PMID: 26647875 DOI: 10.3892/ijo.2015.3248] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/30/2015] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNA MEG3 is suggested to function as a tumor suppressor. However, the activation mechanism of MEG3 is still not well understood and data are not available on its role under adenosine-induced apoptosis. In this study, HepG2 cells were treated with adenosine or 5-Aza‑cdR. Methylation status of MEG3 promoter was detected by methylation specific PCR (MSP) and MEG3 expression was determined by qRT-PCR. PcDNA3.1-MEG3 recombinant plasmid was constructed and transfected to hepatoma HepG2 and Huh7 cells. Cell growth, morphological changes, cell-cycle distribution and apoptosis were analyzed by MTT assay, fluorescence microscopy and flow cytometry. The mRNA and protein expression levels were detected by qRT-PCR and western blot analysis. MEG3 binding proteins were screened by the improved MS2 biotin tagged RNA affinity purification method. The co-expression network of MEG3 was generated by GO analysis and ILF3 was identified as MEG3 binding protein by RNA pulldown and western blot analysis. Both adenosine and 5-Aza-CdR increased MEG3 mRNA expression and the CpG island of MEG3 gene in HepG2 cells was typical hypermethylation. Ectopic expression of MEG3 inhibited hepatoma cell growth in a time-dependent manner, resulted in cell cycle arrest and induced apoptosis. Ectopic expression of MEG3 increased p53, caspase-3 mRNA and protein levels, decreased MDM2 and cyclin D1 mRNA and protein levels, as well as ILF3 protein expression in HepG2 cells. These findings are the first to identify that adenosine increases MEG3 expression by inhibition of DNA methylation and its antitumor effects is involved in MEG3 activation. ILF3 may participate in the anticancer regulation of MEG3 by interacting with MEG3.
Collapse
Affiliation(s)
- Li-Xuan Liu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Wei Deng
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xiao-Tao Zhou
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Rui-Pei Chen
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Meng-Qi Xiang
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yi-Tian Guo
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Ze-Jin Pu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Rui Li
- Key Laboratory of Infectious Diseases and Immunopathology of Guangdong Province, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Ge-Fei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Ling-Fei Wu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
191
|
Li Y, Wang X. Role of long noncoding RNAs in malignant disease (Review). Mol Med Rep 2015; 13:1463-9. [PMID: 26708950 DOI: 10.3892/mmr.2015.4711] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 11/24/2015] [Indexed: 11/06/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are endogenous transcribed RNA molecules without protein-coding potential, ranging between 200 and 100,000 nt in length. LncRNAs regulate the expression of specific genes in several ways, including guiding chromatin-remodeling, and affecting splicing, transcription or translation. The mutations and dysregulation of lncRNAs have been found to be important in various human diseases, but particularly in human cancer. Previous studies have demonstrated that changes to lncRNAs are closely associated with tumorigenesis, metastasis, prognosis and diagnosis. The current review aims to present a brief overview of the associated reports of lncRNAs in malignant neoplasms, including breast cancer, prostate cancer and hematological malignancies. LncRNAs may be evaluated as novel markers in disease diagnosis, and as prospective therapeutic targets for the prevention and treatment of human diseases.
Collapse
Affiliation(s)
- Ying Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
192
|
Chunharojrith P, Nakayama Y, Jiang X, Kery RE, Ma J, De La Hoz Ulloa CS, Zhang X, Zhou Y, Klibanski A. Tumor suppression by MEG3 lncRNA in a human pituitary tumor derived cell line. Mol Cell Endocrinol 2015; 416:27-35. [PMID: 26284494 PMCID: PMC4605874 DOI: 10.1016/j.mce.2015.08.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023]
Abstract
Human clinically non-functioning pituitary adenomas (NFAs) account for approximately 40% of diagnosed pituitary tumors. Epigenetic mutations in tumor suppressive genes play an important role in NFA development. Maternally expressed gene 3 (MEG3) is a long non-coding RNA (lncRNA) and we hypothesized that it is a candidate tumor suppressor whose epigenetic silencing is specifically linked to NFA development. In this study, we introduced MEG3 expression into PDFS cells, derived from a human NFA, using both inducible and constitutively active expression systems. MEG3 expression significantly suppressed xenograft tumor growth in vivo in nude mice. When induced in culture, MEG3 caused cell cycle arrest at the G1 phase. In addition, inactivation of p53 completely abolished tumor suppression by MEG3, indicating that MEG3 tumor suppression is mediated by p53. In conclusion, our data support the hypothesis that MEG3 is a lncRNA tumor suppressor in the pituitary and its inactivation contributes to NFA development.
Collapse
Affiliation(s)
- Paweena Chunharojrith
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yuki Nakayama
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Xiaobing Jiang
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Rachel E Kery
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jun Ma
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Xun Zhang
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yunli Zhou
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Anne Klibanski
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
193
|
Long Noncoding RNA MEG3 Interacts with p53 Protein and Regulates Partial p53 Target Genes in Hepatoma Cells. PLoS One 2015; 10:e0139790. [PMID: 26444285 PMCID: PMC4596861 DOI: 10.1371/journal.pone.0139790] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/17/2015] [Indexed: 12/13/2022] Open
Abstract
Maternally Expressed Gene 3 (MEG3) encodes a lncRNA which is suggested to function as a tumor suppressor. Previous studies suggested that MEG3 functioned through activation of p53, however, the functional properties of MEG3 remain obscure and their relevance to human diseases is under continuous investigation. Here, we try to illuminate the relationship of MEG3 and p53, and the consequence in hepatoma cells. We find that transfection of expression construct of MEG3 enhances stability and transcriptional activity of p53. Deletion analysis of MEG3 confirms that full length and intact structure of MEG3 are critical for it to activate p53-mediated transactivation. Interestingly, our results demonstrate for the first time that MEG3 can interact with p53 DNA binding domain and various p53 target genes are deregulated after overexpression of MEG3 in hepatoma cells. Furthermore, results of qRT-PCR have shown that MEG3 RNA is lost or reduced in the majority of HCC samples compared with adjacent non-tumorous samples. Ectopic expression of MEG3 in hepatoma cells significantly inhibits proliferation and induces apoptosis. In conclusion, our data demonstrates that MEG3 functions as a tumor suppressor in hepatoma cells through interacting with p53 protein to activate p53-mediated transcriptional activity and influence the expression of partial p53 target genes.
Collapse
|
194
|
Expression of the long non-coding RNAs MEG3, HOTAIR, and MALAT-1 in non-functioning pituitary adenomas and their relationship to tumor behavior. Pituitary 2015. [PMID: 24469926 DOI: 10.1007/s11033-013-2852-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE It is becoming evident that long non-coding RNAs (lncRNAs) participate in diverse biological processes via distinct mechanisms. Many lncRNAs have altered expression and likely to have functional roles in tumorigenesis. Although loss of maternally-expressed gene 3 (MEG3) expression has been detected in non-functioning pituitary adenomas (NFPAs), there are no published reports regarding the association between MEG3 expression and the invasive ability of NFPAs. Moreover, the roles of Hox transcript antisense intergenic RNA (HOTAIR) and metastasis-associated lung adenocarcinoma transcript 1 (MALAT-1) in NFPAs have not been examined. To investigate the role of MEG3, HOTAIR, and MALAT-1 in NFPA development and invasion. METHODS MEG3, HOTAIR, MALAT-1 and proliferating cell nuclear antigen (PCNA) were detected in 52 NFPA samples and seven normal human anterior pituitaries using real-time quantitative reverse transcription polymerase chain reaction. RESULTS MEG3 lncRNA levels gradually decreased whereas HOTAIR lncRNA levels gradually increased from normal anterior pituitaries to non-invasive NFPAs to invasive NFPAs. There was a significant association between MEG3 (P < 0.01) and HOTAIR (P < 0.05) expression and the biological behavior of the tumor. Furthermore, PCNA mRNA levels markedly increased in invasive NFPAs compared to non-invasive ones (P < 0.01). In addition, PCNA mRNA negatively correlated with MEG3 lncRNA levels (P < 0.05). CONCLUSIONS MEG3 and HOTAIR expression may correlate with NFPA development and invasion.
Collapse
|
195
|
Huang Y, Liu T. Amyloid Beta Peptide 1-42 Induces SH-SY5Y Cell Apoptosis via the Promotion of Meg3 Long Noncoding RNA Expression. ACTA ACUST UNITED AC 2015. [DOI: 10.1159/000438702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
196
|
You L, Wang N, Yin D, Wang L, Jin F, Zhu Y, Yuan Q, De W. Downregulation of Long Noncoding RNA Meg3 Affects Insulin Synthesis and Secretion in Mouse Pancreatic Beta Cells. J Cell Physiol 2015; 231:852-62. [PMID: 26313443 DOI: 10.1002/jcp.25175] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 08/25/2015] [Indexed: 12/15/2022]
Affiliation(s)
- LiangHui You
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing China
- Nanjing Maternity and Child Health Care Institute; Nanjing Maternity and Child Health Care Hospital Affiliated with Nanjing Medical University; Nanjing China
| | - Ning Wang
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing China
| | - DanDan Yin
- Department of Central Laboratory; The Second Affiliated Hospital of Southeast University; Nanjing China
| | - LinTao Wang
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing China
| | - FeiYan Jin
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing China
| | - YaNan Zhu
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing China
| | - QingXin Yuan
- Department of Endocrinology; First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Wei De
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing China
| |
Collapse
|
197
|
Corsello G, Salzano E, Vecchio D, Antona V, Grasso M, Malacarne M, Carella M, Palumbo P, Piro E, Giuffrè M. Paternal uniparental disomy chromosome 14-like syndrome due a maternal de novo 160 kb deletion at the 14q32.2 region not encompassing the IG- and the MEG3-DMRs: Patient report and genotype-phenotype correlation. Am J Med Genet A 2015; 167A:3130-8. [PMID: 26333487 DOI: 10.1002/ajmg.a.37293] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 07/17/2015] [Indexed: 11/12/2022]
Abstract
The human chromosome 14q32 carries a cluster of imprinted genes which include the paternally expressed genes (PEGs) DLK1 and RTL1, as well as the maternally expressed genes (MEGs) MEG3, RTL1as, and MEG8. PEGs and MEGs expression at the 14q32.2-imprinted region are regulated by two differentially methylated regions (DMRs): the IG-DMR and the MEG3-DMR, which are respectively methylated on the paternal and unmethylated on the maternal chromosome 14 in most cells. Genetic and epigenetic abnormalities affecting these imprinted gene clusters result in two different phenotypes currently known as maternal upd(14) syndrome and paternal upd(14) syndrome. However, only few patients carrying a maternal deletion at the 14q32.2-imprinted critical region have been reported so far. Here we report on the first patient with a maternal de novo deletion of 160 kb at the 14q32.2 chromosome that does not involves the IG-DMR or the MEG3-DMR but elicits a full upd(14)pat syndrome's phenotype encompassing the three mentioned MEGs. By the analysis of this unique genotype-phenotype correlation, we further widen the spectrum of the congenital anomalies associated to this rare disorder and we propose that the paternally expressed imprinted RTL1 gene, as well as its maternally expressed RTL1as antisense transcript, may play a prominent causative role.
Collapse
Affiliation(s)
- Giovanni Corsello
- Department of Sciences for Health Promotion and Mother and Child Care, Università di Palermo, Palermo, Italy
| | - Emanuela Salzano
- Department of Sciences for Health Promotion and Mother and Child Care, Università di Palermo, Palermo, Italy
| | - Davide Vecchio
- Department of Sciences for Health Promotion and Mother and Child Care, Università di Palermo, Palermo, Italy
| | - Vincenzo Antona
- Department of Sciences for Health Promotion and Mother and Child Care, Università di Palermo, Palermo, Italy
| | - Marina Grasso
- Laboratory of Human Genetics, Galliera Hospital, Genoa, Italy
| | | | - Massimo Carella
- Medical Genetics Service, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Pietro Palumbo
- Medical Genetics Service, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Ettore Piro
- Department of Sciences for Health Promotion and Mother and Child Care, Università di Palermo, Palermo, Italy
| | - Mario Giuffrè
- Department of Sciences for Health Promotion and Mother and Child Care, Università di Palermo, Palermo, Italy
| |
Collapse
|
198
|
Wang M, Huang T, Luo G, Huang C, Xiao XY, Wang L, Jiang GS, Zeng FQ. Long non-coding RNA MEG3 induces renal cell carcinoma cells apoptosis by activating the mitochondrial pathway. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s11596-015-1467-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
199
|
Identification of lncRNA MEG3 Binding Protein Using MS2-Tagged RNA Affinity Purification and Mass Spectrometry. Appl Biochem Biotechnol 2015; 176:1834-45. [PMID: 26155902 DOI: 10.1007/s12010-015-1680-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 05/25/2015] [Indexed: 12/12/2022]
Abstract
Long noncoding RNAs (lncRNAs) are nonprotein coding transcripts longer than 200 nucleotides. Recently in mammals, thousands of long noncoding RNAs have been identified and studied as key molecular players in different biological processes with protein complexes. As a long noncoding RNA, maternally expressed gene 3 (MEG3) plays an important role in many cellular processes. However, the mechanism underlying MEG3 regulatory effects remains enigmatic. By using the specific interaction between MS2 coat protein and MS2 RNA hairpin, we developed a method (MS2-tagged RNA affinity purification and mass spectrometry (MTRAP-MS)) to identify proteins that interact with MEG3. Mass spectrometry and gene ontology (GO) analysis showed that MEG3 binding proteins possess nucleotide binding properties and take part in transport, translation, and other biological processes. In addition, interleukin enhancer binding factor 3 (ILF3) and poly(A) binding protein, cytoplasmic 3 (PABPC3) were validated for their interaction with MEG3. These findings indicate that the newly developed method can effectively enrich lncRNA binding proteins and provides a strong basis for studying MEG3 functions.
Collapse
|
200
|
Liu J, Wan L, Lu K, Sun M, Pan X, Zhang P, Lu B, Liu G, Wang Z. The Long Noncoding RNA MEG3 Contributes to Cisplatin Resistance of Human Lung Adenocarcinoma. PLoS One 2015; 10:e0114586. [PMID: 25992654 PMCID: PMC4439130 DOI: 10.1371/journal.pone.0114586] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/11/2014] [Indexed: 01/01/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have been identified as oncogenes or tumor suppressors that are involved in tumorigenesis and chemotherapy drug resistance. Maternally expressed gene 3 (MEG3) is an imprinted gene located at 14q32 that encodes an lncRNA, and decreased MEG3 expression plays an important role in multiple cancers. However, its biological role in the development of the chemoresistance phenotype of human lung adenocarcinoma (LAD) is unknown. This study aimed to observe the expression of MEG3 in LAD and to evaluate its biological role and clinical significance in the resistance of LAD cells to cisplatin. MEG3 expression was markedly decreased in cisplatin-resistant A549/DDP cells compared with parental A549 cells as shown by an lncRNA microarray. MEG3 overexpression in A549/DDP cells increased their chemosensitivity to cisplatin both in vitro and in vivo by inhibiting cell proliferation and inducing apoptosis. By contrast, MEG3 knockdown in A549 cells decreased the chemosensitivity. Moreover, MEG3 was decreased in cisplatin-insensitive LAD tissues while p53 protein levels were decreased and Bcl-xl protein levels increased. Furthermore, patients with lower levels of MEG3 expression showed worse responses to cisplatin-based chemotherapy. These findings demonstrate that MEG3 is significantly downregulated in LAD and partially regulates the cisplatin resistance of LAD cells through the control of p53 and Bcl-xl expression. Thus, MEG3 may represent a new marker of poor response to cisplatin and could be a potential therapeutic target for LAD chemotherapy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Li Wan
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Kaihua Lu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Ming Sun
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Xuan Pan
- Department of Medical Oncology, Nanjing Medical University Affiliated Cancer Hospital of Jiangsu Province, Cancer Institution of Jiangsu Province, Nanjing,P.R. China
| | - Ping Zhang
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Binbin Lu
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Guojian Liu
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Zhaoxia Wang
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
- * E-mail:
| |
Collapse
|