201
|
Zhao W, Smith JA, Mao G, Fornage M, Peyser PA, Sun YV, Turner ST, Kardia SLR. The cis and trans effects of the risk variants of coronary artery disease in the Chr9p21 region. BMC Med Genomics 2015; 8:21. [PMID: 25958224 PMCID: PMC4432789 DOI: 10.1186/s12920-015-0094-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/24/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Recent genome-wide association studies (GWAS) have shown that single nucleotide polymorphisms (SNPs) in the Chr9p21 region are associated with coronary artery disease (CAD). Most of the SNPs identified in this region are non-coding SNPs, suggesting that they may influence gene expression by cis or trans mechanisms to affect disease susceptibility. Since all cells from an individual have the same DNA sequence variations, levels of gene expression in immortalized cell lines can reflect the functional effects of DNA sequence variations that influence or regulate gene expression. The objective of this study is to evaluate the functional consequences of the risk variants in the Chr9p21 region on gene expression. METHODS We examined the association between the variants in the Chr9p21 region and the transcript-level mRNA expression of the adjacent genes (cis) as well as all other genes across the whole genome (trans) from transformed beta-lymphocytes in 801 non-Hispanic white participants from The Genetic Epidemiology Network of Arteriopathy (GENOA) study. RESULTS We found that the CAD risk variants in the Chr9p21 region were significantly associated with the mRNA expression of the ANRIL transcript ENST00000428597 (p = 8.58e-06). Importantly, a few distant transcripts were also found to be associated with the variants in this region, including the well-known CAD risk gene ABCA1 (p = 1.01e-05). Gene enrichment testing suggests that retinol metabolism, N-Glycan biosynthesis, and TGF signaling pathways may be involved. CONCLUSION These results suggest that the effect of risk variants in the Chr9p21 region on susceptibility to CAD is likely to be mediated through both cis and trans mechanisms.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Guangmei Mao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Myriam Fornage
- Institute of Molecular Medicine and Human Genetics Center, University of Texas Health Science Center, Houston, TX, USA.
| | - Patricia A Peyser
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
202
|
Amort M, Nachbauer B, Tuzlak S, Kieser A, Schepers A, Villunger A, Polacek N. Expression of the vault RNA protects cells from undergoing apoptosis. Nat Commun 2015; 6:7030. [PMID: 25952297 PMCID: PMC4430821 DOI: 10.1038/ncomms8030] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 03/26/2015] [Indexed: 02/07/2023] Open
Abstract
Non-protein-coding RNAs are a functionally versatile class of transcripts exerting their biological roles on the RNA level. Recently, we demonstrated that the vault complex-associated RNAs (vtRNAs) are significantly upregulated in Epstein-Barr virus (EBV)-infected human B cells. Very little is known about the function(s) of the vtRNAs or the vault complex. Here, we individually express latent EBV-encoded proteins in B cells and identify the latent membrane protein 1 (LMP1) as trigger for vtRNA upregulation. Ectopic expression of vtRNA1-1, but not of the other vtRNA paralogues, results in an improved viral establishment and reduced apoptosis, a function located in the central domain of vtRNA1-1. Knockdown of the major vault protein has no effect on these phenotypes revealing that vtRNA1-1 and not the vault complex contributes to general cell death resistance. This study describes a NF-κB-mediated role of the non-coding vtRNA1-1 in inhibiting both the extrinsic and intrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Melanie Amort
- Division of Genomics and RNomics, Medical University Innsbruck, Innsbruck A-6020, Austria
| | - Birgit Nachbauer
- Division of Genomics and RNomics, Medical University Innsbruck, Innsbruck A-6020, Austria
- Department of Chemistry and Biochemistry, University of Bern, Bern CH-3012, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern Bern 3012, Switzerland
| | - Selma Tuzlak
- Division of Developmental Immunology, Medical University Innsbruck, Innsbruck A-6020, Austria
| | - Arnd Kieser
- Research Unit Gene Vectors, Helmholtz Zentrum München, München D-81377, Germany
- German Center for Infection Research (DZIF), Partner site Munich, München D-81377, Germany
| | - Aloys Schepers
- Research Unit Gene Vectors, Helmholtz Zentrum München, München D-81377, Germany
- German Center for Infection Research (DZIF), Partner site Munich, München D-81377, Germany
| | - Andreas Villunger
- Division of Developmental Immunology, Medical University Innsbruck, Innsbruck A-6020, Austria
| | - Norbert Polacek
- Division of Genomics and RNomics, Medical University Innsbruck, Innsbruck A-6020, Austria
- Department of Chemistry and Biochemistry, University of Bern, Bern CH-3012, Switzerland
| |
Collapse
|
203
|
Hao Z, Fan C, Cheng T, Su Y, Wei Q, Li G. Genome-wide identification, characterization and evolutionary analysis of long intergenic noncoding RNAs in cucumber. PLoS One 2015; 10:e0121800. [PMID: 25799544 PMCID: PMC4370693 DOI: 10.1371/journal.pone.0121800] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 02/04/2015] [Indexed: 12/25/2022] Open
Abstract
Long intergenic noncoding RNAs (lincRNAs) are intergenic transcripts with a length of at least 200 nt that lack coding potential. Emerging evidence suggests that lincRNAs from animals participate in many fundamental biological processes. However, the systemic identification of lincRNAs has been undertaken in only a few plants. We chose to use cucumber (Cucumis sativus) as a model to analyze lincRNAs due to its importance as a model plant for studying sex differentiation and fruit development and the rich genomic and transcriptome data available. The application of a bioinformatics pipeline to multiple types of gene expression data resulted in the identification and characterization of 3,274 lincRNAs. Next, 10 lincRNAs targeted by 17 miRNAs were also explored. Based on co-expression analysis between lincRNAs and mRNAs, 94 lincRNAs were annotated, which may be involved in response to stimuli, multi-organism processes, reproduction, reproductive processes, and growth. Finally, examination of the evolution of lincRNAs showed that most lincRNAs are under purifying selection, while 16 lincRNAs are under natural selection. Our results provide a rich resource for further validation of cucumber lincRNAs and their function. The identification of lincRNAs targeted by miRNAs offers new clues for investigations into the role of lincRNAs in regulating gene expression. Finally, evaluation of the lincRNAs suggested that some lincRNAs are under positive and balancing selection.
Collapse
Affiliation(s)
- Zhiqiang Hao
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
- Co-Innovation Center for Qinba Regions’ Sustainable Development, College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Chunyan Fan
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Tian Cheng
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Ya Su
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Qiang Wei
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Guanglin Li
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
- Co-Innovation Center for Qinba Regions’ Sustainable Development, College of Life Sciences, Shaanxi Normal University, Xi’an, China
- * E-mail:
| |
Collapse
|
204
|
Abstract
Long non-coding RNAs (lncRNAs) are a large and diverse group of RNAs that are often lineage-specific and that regulate multiple biological functions. Many are nuclear and are essential parts of ribonucleoprotein complexes that modify chromatin segments and establish active or repressive chromatin states; others are cytosolic and regulate the stability of mRNA or act as microRNA sponges. This Review summarizes the current knowledge of lncRNAs as regulators of the endocrine system, with a focus on the identification and mode of action of several endocrine-important lncRNAs. We highlight lncRNAs that have a role in the development and function of pancreatic β cells, white and brown adipose tissue, and other endocrine organs, and discuss the involvement of these molecules in endocrine dysfunction (for example, diabetes mellitus). We also address the associations of lncRNAs with nuclear receptors involved in major hormonal signalling pathways, such as estrogen and androgen receptors, and the relevance of these associations in certain endocrine cancers.
Collapse
Affiliation(s)
- Marko Knoll
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, MA 02142, USA
| | - Harvey F Lodish
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, MA 02142, USA
| | - Lei Sun
- Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Graduate Medical School, 8 College Road, 169857, Singapore
| |
Collapse
|
205
|
Cui M, Xiao Z, Wang Y, Zheng M, Song T, Cai X, Sun B, Ye L, Zhang X. Long noncoding RNA HULC modulates abnormal lipid metabolism in hepatoma cells through an miR-9-mediated RXRA signaling pathway. Cancer Res 2015; 75:846-57. [PMID: 25592151 DOI: 10.1158/0008-5472.can-14-1192] [Citation(s) in RCA: 306] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
HULC is a long noncoding RNA overexpressed in hepatocellular carcinoma (HCC), but its functional contributions in this setting have not been determined. In this study, we explored the hypothesis that HULC contributes to malignant development by supporting abnormal lipid metabolism in hepatoma cells. HULC modulated the deregulation of lipid metabolism in HCC by activating the acyl-CoA synthetase subunit ACSL1. Immunohistochemical analysis of tissue microarrays revealed that approximately 77% (180/233) of HCC tissues were positive for ACSL1. Moreover, HULC mRNA levels correlated positively with ACSL1 levels in 60 HCC cases according to real-time PCR analysis. Mechanistic investigations showed that HULC upregulated the transcriptional factor PPARA, which activated the ACSL1 promoter in hepatoma cells. HULC also suppressed miR-9 targeting of PPARA mRNA by eliciting methylation of CpG islands in the miR-9 promoter. We documented the ability of HULC to promote lipogenesis, thereby stimulating accumulation of intracellular triglycerides and cholesterol in vitro and in vivo. Strikingly, ACSL1 overexpression that generates cholesterol was sufficient to enhance the proliferation of hepatoma cells. Further, cholesterol addition was sufficient to upregulate HULC expression through a positive feedback loop involving the retinoid receptor RXRA, which activated the HULC promoter. Overall, we concluded that HULC functions as an oncogene in hepatoma cells, acting mechanistically by deregulating lipid metabolism through a signaling pathway involving miR-9, PPARA, and ACSL1 that is reinforced by a feed-forward pathway involving cholesterol and RXRA to drive HULC signaling.
Collapse
Affiliation(s)
- Ming Cui
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin, China
| | - Zelin Xiao
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin, China
| | - Yue Wang
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, China
| | - Minying Zheng
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin, China
| | - Tianqiang Song
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China. Key Laboratory of Cancer Prevention and Therapy, Tianjin Department of Hepatobiliary Tumor, Tianjin, China
| | - Xiaoli Cai
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, China
| | - Baodi Sun
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin, China
| | - Lihong Ye
- State Key Laboratory of Medicinal Chemical Biology, Department of Biochemistry, College of Life Sciences, Nankai University, Tianjin, China.
| | - Xiaodong Zhang
- State Key Laboratory of Medicinal Chemical Biology, Department of Cancer Research, College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
206
|
Peng L, Paulson A, Li H, Piekos S, He X, Li L, Zhong XB. Developmental programming of long non-coding RNAs during postnatal liver maturation in mice. PLoS One 2014; 9:e114917. [PMID: 25503252 PMCID: PMC4263710 DOI: 10.1371/journal.pone.0114917] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/15/2014] [Indexed: 11/19/2022] Open
Abstract
The liver is a vital organ with critical functions in metabolism, protein synthesis, and immune defense. Most of the liver functions are not mature at birth and many changes happen during postnatal liver development. However, it is unclear what changes occur in liver after birth, at what developmental stages they occur, and how the developmental processes are regulated. Long non-coding RNAs (lncRNAs) are involved in organ development and cell differentiation. Here, we analyzed the transcriptome of lncRNAs in mouse liver from perinatal (day −2) to adult (day 60) by RNA-Sequencing, with an attempt to understand the role of lncRNAs in liver maturation. We found around 15,000 genes expressed, including about 2,000 lncRNAs. Most lncRNAs were expressed at a lower level than coding RNAs. Both coding RNAs and lncRNAs displayed three major ontogenic patterns: enriched at neonatal, adolescent, or adult stages. Neighboring coding and non-coding RNAs showed the trend to exhibit highly correlated ontogenic expression patterns. Gene ontology (GO) analysis revealed that some lncRNAs enriched at neonatal ages have their neighbor protein coding genes also enriched at neonatal ages and associated with cell proliferation, immune activation related processes, tissue organization pathways, and hematopoiesis; other lncRNAs enriched at adolescent ages have their neighbor protein coding genes associated with different metabolic processes. These data reveal significant functional transition during postnatal liver development and imply the potential importance of lncRNAs in liver maturation.
Collapse
Affiliation(s)
- Lai Peng
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
| | - Ariel Paulson
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Hua Li
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Stephanie Piekos
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
| | - Xi He
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Linheng Li
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Xiao-bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
207
|
Witzany G. RNA sociology: group behavioral motifs of RNA consortia. Life (Basel) 2014; 4:800-18. [PMID: 25426799 PMCID: PMC4284468 DOI: 10.3390/life4040800] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023] Open
Abstract
RNA sociology investigates the behavioral motifs of RNA consortia from the social science perspective. Besides the self-folding of RNAs into single stem loop structures, group building of such stem loops results in a variety of essential agents that are highly active in regulatory processes in cellular and non-cellular life. RNA stem loop self-folding and group building do not depend solely on sequence syntax; more important are their contextual (functional) needs. Also, evolutionary processes seem to occur through RNA stem loop consortia that may act as a complement. This means the whole entity functions only if all participating parts are coordinated, although the complementary building parts originally evolved for different functions. If complementary groups, such as rRNAs and tRNAs, are placed together in selective pressure contexts, new evolutionary features may emerge. Evolution initiated by competent agents in natural genome editing clearly contrasts with statistical error replication narratives.
Collapse
Affiliation(s)
- Guenther Witzany
- Telos-Philosophische Praxis, Vogelsangstraße 18c, 5111-Buermoos, Austria.
| |
Collapse
|
208
|
Ultsch A, Lötsch J. What do all the (human) micro-RNAs do? BMC Genomics 2014; 15:976. [PMID: 25404408 PMCID: PMC4289375 DOI: 10.1186/1471-2164-15-976] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 10/13/2014] [Indexed: 12/23/2022] Open
Abstract
Background Micro-RNAs (miRNA) are attributed to the systems biological role of a regulatory mechanism of the expression of protein coding genes. Research has identified miRNAs dysregulations in several but distinct pathophysiological processes, which hints at distinct systems-biology functions of miRNAs. The present analysis approached the role of miRNAs from a genomics perspective and assessed the biological roles of 2954 genes and 788 human miRNAs, which can be considered to interact, based on empirical evidence and computational predictions of miRNA versus gene interactions. Results From a genomics perspective, the biological processes in which the genes that are influenced by miRNAs are involved comprise of six major topics comprising biological regulation, cellular metabolism, information processing, development, gene expression and tissue homeostasis. The usage of this knowledge as a guidance for further research is sketched for two genetically defined functional areas: cell death and gene expression. Results suggest that the latter points to a fundamental role of miRNAs consisting of hyper-regulation of gene expression, i.e., the control of the expression of such genes which control specifically the expression of genes. Conclusions Laboratory research identified contributions of miRNA regulation to several distinct biological processes. The present analysis transferred this knowledge to a systems-biology level. A comprehensible and precise description of the biological processes in which the genes that are influenced by miRNAs are notably involved could be made. This knowledge can be employed to guide future research concerning the biological role of miRNA (dys-) regulations. The analysis also suggests that miRNAs especially control the expression of genes that control the expression of genes. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-976) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe - University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
209
|
Tian S, Cordero P, Kladwang W, Das R. High-throughput mutate-map-rescue evaluates SHAPE-directed RNA structure and uncovers excited states. RNA (NEW YORK, N.Y.) 2014; 20:1815-26. [PMID: 25183835 PMCID: PMC4201832 DOI: 10.1261/rna.044321.114] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The three-dimensional conformations of noncoding RNAs underpin their biochemical functions but have largely eluded experimental characterization. Here, we report that integrating a classic mutation/rescue strategy with high-throughput chemical mapping enables rapid RNA structure inference with unusually strong validation. We revisit a 16S rRNA domain for which SHAPE (selective 2'-hydroxyl acylation with primer extension) and limited mutational analysis suggested a conformational change between apo- and holo-ribosome conformations. Computational support estimates, data from alternative chemical probes, and mutate-and-map (M(2)) experiments highlight issues of prior methodology and instead give a near-crystallographic secondary structure. Systematic interrogation of single base pairs via a high-throughput mutation/rescue approach then permits incisive validation and refinement of the M(2)-based secondary structure. The data further uncover the functional conformation as an excited state (20 ± 10% population) accessible via a single-nucleotide register shift. These results correct an erroneous SHAPE inference of a ribosomal conformational change, expose critical limitations of conventional structure mapping methods, and illustrate practical steps for more incisively dissecting RNA dynamic structure landscapes.
Collapse
Affiliation(s)
- Siqi Tian
- Department of Biochemistry, Stanford University, Stanford, California 94305, USA
| | - Pablo Cordero
- Biomedical Informatics Program, Stanford University, Stanford, California 94305, USA
| | - Wipapat Kladwang
- Department of Physics, Stanford University, Stanford, California 94305, USA
| | - Rhiju Das
- Department of Biochemistry, Stanford University, Stanford, California 94305, USA Biomedical Informatics Program, Stanford University, Stanford, California 94305, USA Department of Physics, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
210
|
Zuo L, Lu L, Tan Y, Pan X, Cai Y, Wang X, Hong J, Zhong C, Wang F, Zhang XY, Vanderlinden LA, Tabakoff B, Luo X. Genome-wide association discoveries of alcohol dependence. Am J Addict 2014; 23:526-39. [PMID: 25278008 PMCID: PMC4187224 DOI: 10.1111/j.1521-0391.2014.12147.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 04/17/2014] [Accepted: 05/12/2014] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE To report the genome-wide significant and/or replicable risk variants for alcohol dependence and explore their potential biological functions. METHODS We searched in PubMed for all genome-wide association studies (GWASs) of alcohol dependence. The following three types of the results were extracted: genome-wide significant associations in an individual sample, the combined samples, or the meta-analysis (p < 5 × 10(-8) ); top-ranked associations in an individual sample (p < 10(-5) ) that were nominally replicated in other samples (p < .05); and nominally replicable associations across at least three independent GWAS samples (p < .05). These results were meta-analyzed. cis-eQTLs in human, RNA expression in rat and mouse brains and bioinformatics properties of all of these risk variants were analyzed. RESULTS The variants located within the alcohol dehydrogenase (ADH) cluster were significantly associated with alcohol dependence at the genome-wide level (p < 5 × 10(-8) ) in at least one sample. Some associations with the ADH cluster were replicable across six independent GWAS samples. The variants located within or near SERINC2, KIAA0040, MREG-PECR or PKNOX2 were significantly associated with alcohol dependence at the genome-wide level (p < 5 × 10(-8) ) in meta-analysis or combined samples, and these associations were replicable across at least one sample. The associations with the variants within NRD1, GPD1L-CMTM8 or MAP3K9-PCNX were suggestive (5 × 10(-8) < p < 10(-5) ) in some samples, and nominally replicable in other samples. The associations with the variants at HTR7 and OPA3 were nominally replicable across at least three independent GWAS samples (10(-5) < p < .05). Some risk variants at the ADH cluster, SERINC2, KIAA0040, NRD1, and HTR7 had potential biological functions. CONCLUSION The most robust risk locus was the ADH cluster. SERINC2, KIAA0040, NRD1, and HTR7 were also likely to play important roles in alcohol dependence. PKNOX2, MREG, PECR, GPD1L, CMTM8, MAP3K9, PCNX, and OPA3 might play less important roles in risk for alcohol dependence based on the function analysis. This conclusion will significantly contribute to the post-GWAS follow-up studies on alcohol dependence.
Collapse
Affiliation(s)
- Lingjun Zuo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Yunlong Tan
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China
| | - Xinghua Pan
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Yiqiang Cai
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaoping Wang
- Department of Neurology, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jiang Hong
- Department of Internal Medicine, First People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Chunlong Zhong
- Department of Neurosurgery, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Fei Wang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Xiang-yang Zhang
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | | | - Boris Tabakoff
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China
| |
Collapse
|
211
|
Hansji H, Leung EY, Baguley BC, Finlay GJ, Askarian-Amiri ME. Keeping abreast with long non-coding RNAs in mammary gland development and breast cancer. Front Genet 2014; 5:379. [PMID: 25400658 PMCID: PMC4215690 DOI: 10.3389/fgene.2014.00379] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/13/2014] [Indexed: 12/18/2022] Open
Abstract
The majority of the human genome is transcribed, even though only 2% of transcripts encode proteins. Non-coding transcripts were originally dismissed as evolutionary junk or transcriptional noise, but with the development of whole genome technologies, these non-coding RNAs (ncRNAs) are emerging as molecules with vital roles in regulating gene expression. While shorter ncRNAs have been extensively studied, the functional roles of long ncRNAs (lncRNAs) are still being elucidated. Studies over the last decade show that lncRNAs are emerging as new players in a number of diseases including cancer. Potential roles in both oncogenic and tumor suppressive pathways in cancer have been elucidated, but the biological functions of the majority of lncRNAs remain to be identified. Accumulated data are identifying the molecular mechanisms by which lncRNA mediates both structural and functional roles. LncRNA can regulate gene expression at both transcriptional and post-transcriptional levels, including splicing and regulating mRNA processing, transport, and translation. Much current research is aimed at elucidating the function of lncRNAs in breast cancer and mammary gland development, and at identifying the cellular processes influenced by lncRNAs. In this paper we review current knowledge of lncRNAs contributing to these processes and present lncRNA as a new paradigm in breast cancer development.
Collapse
Affiliation(s)
- Herah Hansji
- Auckland Cancer Society Research Centre, University of Auckland Auckland, New Zealand
| | - Euphemia Y Leung
- Auckland Cancer Society Research Centre, University of Auckland Auckland, New Zealand
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, University of Auckland Auckland, New Zealand
| | - Graeme J Finlay
- Auckland Cancer Society Research Centre, University of Auckland Auckland, New Zealand ; Department of Molecular Medicine and Pathology, University of Auckland Auckland, New Zealand
| | | |
Collapse
|
212
|
Quek XC, Thomson DW, Maag JLV, Bartonicek N, Signal B, Clark MB, Gloss BS, Dinger ME. lncRNAdb v2.0: expanding the reference database for functional long noncoding RNAs. Nucleic Acids Res 2014; 43:D168-73. [PMID: 25332394 PMCID: PMC4384040 DOI: 10.1093/nar/gku988] [Citation(s) in RCA: 396] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Despite the prevalence of long noncoding RNA (lncRNA) genes in eukaryotic genomes, only a small proportion have been examined for biological function. lncRNAdb, available at http://lncrnadb.org, provides users with a comprehensive, manually curated reference database of 287 eukaryotic lncRNAs that have been described independently in the scientific literature. In addition to capturing a great proportion of the recent literature describing functions for individual lncRNAs, lncRNAdb now offers an improved user interface enabling greater accessibility to sequence information, expression data and the literature. The new features in lncRNAdb include the integration of Illumina Body Atlas expression profiles, nucleotide sequence information, a BLAST search tool and easy export of content via direct download or a REST API. lncRNAdb is now endorsed by RNAcentral and is in compliance with the International Nucleotide Sequence Database Collaboration.
Collapse
Affiliation(s)
- Xiu Cheng Quek
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Daniel W Thomson
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia
| | - Jesper L V Maag
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Nenad Bartonicek
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia
| | - Bethany Signal
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia
| | - Michael B Clark
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia MRC Functional Genomics Unit, Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Brian S Gloss
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Marcel E Dinger
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, NSW 2010, Australia St Vincent's Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
213
|
Yin D, He X, Zhang E, Kong R, De W, Zhang Z. Long noncoding RNA GAS5 affects cell proliferation and predicts a poor prognosis in patients with colorectal cancer. Med Oncol 2014; 31:253. [PMID: 25326054 DOI: 10.1007/s12032-014-0253-8] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/16/2014] [Indexed: 12/28/2022]
Abstract
Colorectal cancer (CRC) is the third most common type of cancer worldwide. Recent studies have shown that lncRNAs play important roles in carcinogenesis. The aim of this study was to explore the role of lncRNA GAS5 in CRC. Real-time PCR was performed to investigate the expression of GAS5 in tumor tissues and corresponding non-tumor colorectal tissues from 66 patients with CRC. The lower expression of GAS5 was significantly correlated with large tumor size, low histological grade and advanced TNM stage. Multivariate analyses revealed that GAS5 expression served as an independent predictor for overall survival (P = 0.034). Further experiments revealed that overexpressed GAS5 significantly repressed the proliferation both in vitro and in vivo. In conclusion, our results suggest that GAS5, as a growth regulator, may serve as a candidate prognostic biomarker in human colorectal cancer.
Collapse
Affiliation(s)
- Dandan Yin
- Cancer Research and Therapy Center, The Second Affiliated Hospital of Southeast University, Nanjing, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
214
|
Hu YW, Zhao JY, Li SF, Wang Q, Zheng L. Genome-wide profiling to analyze the effects of Ox-LDL induced THP-1 macrophage-derived foam cells on gene expression. GENOMICS DATA 2014; 2:328-31. [PMID: 26484122 PMCID: PMC4535961 DOI: 10.1016/j.gdata.2014.09.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 09/29/2014] [Indexed: 01/07/2023]
Abstract
Atherosclerosis has a high incidence and is harmful to human health. An elevated level of oxidized low-density lipoprotein (Ox-LDL) is one of the major risk factors for atherosclerosis. During atherogenesis progression, circulating monocytes adhere to the intima and differentiate into macrophages. After differentiation, intimal macrophages intake Ox-LDL via scavenger receptors, thereby transforming into foam cells. Foam cell formation due to excessive accumulation of cholesterol by macrophages is a pathological hallmark of atherosclerosis. To gain a molecular understanding of the effect of Ox-LDL in atherosclerosis development, we conducted a genome-wide analysis of the Ox-LDL-induced macrophage transformation by microarray gene expression profiling. Here we describe in details the contents and quality controls for the gene expression and related results associated with the data uploaded to Gene Expression Omnibus (accession number GSE54039).
Collapse
Affiliation(s)
- Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jia-Yi Zhao
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shu-Fen Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
215
|
Role of epigenetics in expression of recombinant proteins from mammalian cells. ACTA ACUST UNITED AC 2014. [DOI: 10.4155/pbp.14.47] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
216
|
Koppers-Lalic D, Hackenberg M, Bijnsdorp IV, van Eijndhoven MAJ, Sadek P, Sie D, Zini N, Middeldorp JM, Ylstra B, de Menezes RX, Würdinger T, Meijer GA, Pegtel DM. Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes. Cell Rep 2014; 8:1649-1658. [PMID: 25242326 DOI: 10.1016/j.celrep.2014.08.027] [Citation(s) in RCA: 444] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 03/25/2014] [Accepted: 08/13/2014] [Indexed: 01/11/2023] Open
Abstract
Functional biomolecules, including small noncoding RNAs (ncRNAs), are released and transmitted between mammalian cells via extracellular vesicles (EVs), including endosome-derived exosomes. The small RNA composition in cells differs from exosomes, but underlying mechanisms have not been established. We generated small RNA profiles by RNA sequencing (RNA-seq) from a panel of human B cells and their secreted exosomes. A comprehensive bioinformatics and statistical analysis revealed nonrandomly distributed subsets of microRNA (miRNA) species between B cells and exosomes. Unexpectedly, 3' end adenylated miRNAs are relatively enriched in cells, whereas 3' end uridylated isoforms appear overrepresented in exosomes, as validated in naturally occurring EVs isolated from human urine samples. Collectively, our findings suggest that posttranscriptional modifications, notably 3' end adenylation and uridylation, exert opposing effects that may contribute, at least in part, to direct ncRNA sorting into EVs.
Collapse
Affiliation(s)
- Danijela Koppers-Lalic
- Department of Pathology, VU University Medical Center, 1007MB Amsterdam, the Netherlands; Exosomes Research Group, VU University Medical Center, 1007MB Amsterdam, the Netherlands.
| | - Michael Hackenberg
- Department of Genetics, Computational Genomics and Bioinformatics Group, University of Granada, Granada 18071, Spain
| | - Irene V Bijnsdorp
- Department of Urology, VU University Medical Center, 1007MB Amsterdam, the Netherlands
| | - Monique A J van Eijndhoven
- Department of Pathology, VU University Medical Center, 1007MB Amsterdam, the Netherlands; Exosomes Research Group, VU University Medical Center, 1007MB Amsterdam, the Netherlands
| | - Payman Sadek
- Department of Pathology, VU University Medical Center, 1007MB Amsterdam, the Netherlands; Exosomes Research Group, VU University Medical Center, 1007MB Amsterdam, the Netherlands
| | - Daud Sie
- Department of Pathology, VU University Medical Center, 1007MB Amsterdam, the Netherlands
| | - Nicoletta Zini
- CNR-National Research Council of Italy, IGM, and SC Laboratory of Musculoskeletal Cell Biology, IOR, 40136 Bologna, Italy
| | - Jaap M Middeldorp
- Department of Pathology, VU University Medical Center, 1007MB Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, 1007MB Amsterdam, the Netherlands
| | - Renee X de Menezes
- Department of Epidemiology and Biostatistics, VU University Medical Center, 1007MB Amsterdam, the Netherlands
| | - Thomas Würdinger
- Department of Neurosurgery, Neuro-Oncology Research Group, VU University Medical Center, 1007MB Amsterdam, the Netherlands; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Gerrit A Meijer
- Department of Pathology, VU University Medical Center, 1007MB Amsterdam, the Netherlands
| | - D Michiel Pegtel
- Department of Pathology, VU University Medical Center, 1007MB Amsterdam, the Netherlands; Exosomes Research Group, VU University Medical Center, 1007MB Amsterdam, the Netherlands.
| |
Collapse
|
217
|
LncRBase: an enriched resource for lncRNA information. PLoS One 2014; 9:e108010. [PMID: 25233092 PMCID: PMC4169474 DOI: 10.1371/journal.pone.0108010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/11/2014] [Indexed: 11/19/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are noncoding transcripts longer than 200 nucleotides, which show evidence of pervasive transcription and participate in a plethora of cellular regulatory processes. Although several noncoding transcripts have been functionally annotated as lncRNAs within the genome, not all have been proven to fulfill the criteria for a functional regulator and further analyses have to be done in order to include them in a functional cohort. LncRNAs are being classified and reclassified in an ongoing annotation process, and the challenge is fraught with ambiguity, as newer evidences of their biogenesis and functional implication come into light. In our effort to understand the complexity of this still enigmatic biomolecule, we have developed a new database entitled "LncRBase" where we have classified and characterized lncRNAs in human and mouse. It is an extensive resource of human and mouse lncRNA transcripts belonging to fourteen distinct subtypes, with a total of 83,201 entries for mouse and 133,361 entries for human: among these, we have newly annotated 8,507 mouse and 14,813 human non coding RNA transcripts (from UCSC and H-InvDB 8.0) as lncRNAs. We have especially considered protein coding gene loci which act as hosts for non coding transcripts. LncRBase includes different lncRNA transcript variants of protein coding genes within LncRBase. LncRBase provides information about the genomic context of different lncRNA subtypes, their interaction with small non coding RNAs (ncRNAs) viz. piwi interacting RNAs (piRNAs) and microRNAs (miRNAs) and their mode of regulation, via association with diverse other genomic elements. Adequate knowledge about genomic origin and molecular features of lncRNAs is essential to understand their functional and behavioral complexities. Overall, LncRBase provides a thorough study on various aspects of lncRNA origin and function and a user-friendly interface to search for lncRNA information. LncRBase is available at http://bicresources.jcbose.ac.in/zhumur/lncrbase.
Collapse
|
218
|
Abstract
In the last decades, the developmental origins of health and disease (DOHaD) have emerged as a vigorous field combining experimental, clinical, epidemiological and public health research. Its goal is to understand how events in early life shape later morbidity risk, especially of non-communicable chronic diseases. As these diseases become the major cause of morbidity and mortality worldwide, research arising from DOHaD is likely to gain significance to public health and economic development. But action may be hindered by the lack of a firm mechanistic explanation and of a conceptual basis, especially regarding the evolutionary significance of the DOHaD phenomenon. In this article, we provide a succinct historical review of the research into the relationship between development and later disease, consider the evolutionary and developmental significance and discuss the underlying mechanisms of the DOHaD phenomenon. DOHaD should be viewed as a part of a broader biological mechanism of plasticity by which organisms, in response to cues such as nutrition or hormones, adapt their phenotype to environment. These responses may be divided into those for immediate benefit and those aimed at prediction of a future environment: disease occurs in the mismatch between predicted and realized future. The likely mechanisms that enable plasticity involve epigenetic processes, affecting the expression of genes associated with regulatory pathways. There is now evidence that epigenetic marks may be inherited and so contribute to non-genomic heritable disease risk. We end by discussing the global significance of the DOHaD phenomenon and its potential applications for public health purposes.
Collapse
|
219
|
Abstract
Numerous studies over the past decade have identified increasing numbers of long noncoding RNAs (lncRNAs) across many organisms. Research since has shown that lncRNAs constitute an important layer of genome regulation in diverse biological processes and disease. Here, we discuss the common emerging theme of lncRNAs interfacing with epigenetic machinery. This, in turn, modulates the activity and localization of the epigenetic machinery during cell fate specification.
Collapse
Affiliation(s)
- John L Rinn
- Harvard University, Department of Stem Cell and Regenerative Biology, Cambridge, Massachusetts 02138
| |
Collapse
|
220
|
Dynamic transcription of long non-coding RNA genes during CD4+ T cell development and activation. PLoS One 2014; 9:e101588. [PMID: 25003630 PMCID: PMC4086894 DOI: 10.1371/journal.pone.0101588] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/07/2014] [Indexed: 11/19/2022] Open
Abstract
Background Recent evidence shows that long non-coding RNA (LncRNA) play important regulatory roles in many biology process, including cell development, activation and oncogenesis. However, the roles of these LncRNAs in the development and activation of CD4+ T cells, which is an important component of immune response, remain unknown. Results To predict the function of LncRNA in the development and activation of CD4+ T cells, first, we examined the expression profiles of LncRNAs and mRNAs in CD4−CD8− (DN), CD4+CD8+ (DP), CD4+CD8−, and activated CD4+CD8− T cells in a microarray analysis and verified these results by real time PCRs (qPCR). We found that the expression of hundreds of LncRNAs significantly changed in each process of developmental transition, including DN into DP, DP into CD4+CD8−, and CD4+CD8− into activated CD4+ T cells. A Kendall distance analysis suggested that the expression of LncRNAs in DN, DP, CD4+CD8− T cells and activated CD4+ T cells were correlated with the expression of mRNAs in these T cells. The Blat algorithm and GO analysis suggested that LncRNAs may exert important roles in the development and activation of CD4+ T cells. These roles included proliferation, homeostasis, maturation, activation, migration, apoptosis and calcium ion transportation. Conclusion The present study found that the expression profiles of LncRNAs in different stages of CD4+ T cells are distinguishable. LncRNAs are involved in the key biological process in CD4+ T cell development and activation.
Collapse
|
221
|
Giza DE, Vasilescu C, Calin GA. MicroRNAs and ceRNAs: therapeutic implications of RNA networks. Expert Opin Biol Ther 2014; 14:1285-93. [PMID: 24941322 DOI: 10.1517/14712598.2014.920812] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION A new concept of gene regulation, in which competitive endogenous RNAs (ceRNAs) compete for common microRNAs (miRNAs), suggests that mRNA targets have an active role as key elements in the regulation of miRNA availability within cells. ceRNAs are considered to be natural decoys of miRNA activity and can influence the expression of multiple miRNAs. AREAS COVERED A new complex network of indirect interaction among the RNA transcripts competing for the same pool of miRNAs has been described; in this network, the nodes are the targets, and the links between the nodes are the miRNAs the targets have in common, which form smaller subnetworks. The incidence, state and severity of cancer can be evaluated on the basis of this network signature. The study of these new genome-scale regulatory networks involving miRNAs and ceRNAs may provide information that researchers can use to fine-tune these networks to improve responses to cancer therapy and/or develop new therapeutic interventions. EXPERT OPINION Combinational approaches based on complex regulatory ceRNA networks (ceRNETs) may be one of the most promising strategies for silencing important mediators of cancer-promoting pathways. Targeting a single miRNA may in fact represent a combined intervention that acts on the feedback and compensatory pathways that can impair treatment response or cause treatment resistance.
Collapse
Affiliation(s)
- Dana Elena Giza
- The University of Texas MD Anderson Cancer Center, Department of Experimental Therapeutics , Unit 1950, 1515 Holcombe Blvd, Houston, TX, 77030-4009 , USA +1 713 792 5461 ; +1 713 792 1204 ;
| | | | | |
Collapse
|
222
|
Clark BS, Blackshaw S. Long non-coding RNA-dependent transcriptional regulation in neuronal development and disease. Front Genet 2014; 5:164. [PMID: 24936207 PMCID: PMC4047558 DOI: 10.3389/fgene.2014.00164] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 05/18/2014] [Indexed: 01/17/2023] Open
Abstract
Comprehensive analysis of the mammalian transcriptome has revealed that long non-coding RNAs (lncRNAs) may make up a large fraction of cellular transcripts. Recent years have seen a surge of studies aimed at functionally characterizing the role of lncRNAs in development and disease. In this review, we discuss new findings implicating lncRNAs in controlling development of the central nervous system (CNS). The evolution of the higher vertebrate brain has been accompanied by an increase in the levels and complexities of lncRNAs expressed within the developing nervous system. Although a limited number of CNS-expressed lncRNAs are now known to modulate the activity of proteins important for neuronal differentiation, the function of the vast majority of neuronal-expressed lncRNAs is still unknown. Topics of intense current interest include the mechanism by which CNS-expressed lncRNAs might function in epigenetic and transcriptional regulation during neuronal development, and how gain and loss of function of individual lncRNAs contribute to neurological diseases.
Collapse
Affiliation(s)
- Brian S Clark
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Seth Blackshaw
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Ophthalmology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Department of Neurology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Center for High-Throughput Biology, Johns Hopkins University School of Medicine Baltimore, MD, USA ; Institute for Cell Engineering, Johns Hopkins University School of Medicine Baltimore, MD, USA
| |
Collapse
|
223
|
Liu PY, Erriquez D, Marshall GM, Tee AE, Polly P, Wong M, Liu B, Bell JL, Zhang XD, Milazzo G, Cheung BB, Fox A, Swarbrick A, Hüttelmaier S, Kavallaris M, Perini G, Mattick JS, Dinger ME, Liu T. Effects of a novel long noncoding RNA, lncUSMycN, on N-Myc expression and neuroblastoma progression. J Natl Cancer Inst 2014; 106:dju113. [PMID: 24906397 DOI: 10.1093/jnci/dju113] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Patients with neuroblastoma due to the amplification of a 130-kb genomic DNA region containing the MYCN oncogene have poor prognoses. METHODS Bioinformatics data were used to discover a novel long noncoding RNA, lncUSMycN, at the 130-kb amplicon. RNA-protein pull-down assays were used to identify proteins bound to lncUSMycN RNA. Kaplan-Meier survival analysis, multivariable Cox regression, and two-sided log-rank test were used to examine the prognostic value of lncUSMycN and NonO expression in three cohorts of neuroblastoma patients (n = 47, 88, and 476, respectively). Neuroblastoma-bearing mice were treated with antisense oligonucleotides targeting lncUSMycN (n = 12) or mismatch sequence (n = 13), and results were analyzed by multiple comparison two-way analysis of variance. All statistical tests were two-sided. RESULTS Bioinformatics data predicted lncUSMycN gene and RNA, and reverse-transcription polymerase chain reaction confirmed its three exons and two introns. The lncUSMycN gene was coamplified with MYCN in 88 of 341 human neuroblastoma tissues. lncUSMycN RNA bound to the RNA-binding protein NonO, leading to N-Myc RNA upregulation and neuroblastoma cell proliferation. High levels of lncUSMycN and NonO expression in human neuroblastoma tissues independently predicted poor patient prognoses (lncUSMycN: hazard ratio [HR] = 1.87, 95% confidence interval [CI] = 1.06 to 3.28, P = .03; NonO: HR = 2.48, 95% CI = 1.34 to 4.57, P = .004). Treatment with antisense oligonucleotides targeting lncUSMycN in neuroblastoma-bearing mice statistically significantly hindered tumor progression (P < .001). CONCLUSIONS Our data demonstrate the important roles of lncUSMycN and NonO in regulating N-Myc expression and neuroblastoma oncogenesis and provide the first evidence that amplification of long noncoding RNA genes can contribute to tumorigenesis.
Collapse
Affiliation(s)
- Pei Y Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Daniela Erriquez
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Glenn M Marshall
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Andrew E Tee
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Patsie Polly
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Mathew Wong
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Bing Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Jessica L Bell
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Xu D Zhang
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Giorgio Milazzo
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Belamy B Cheung
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Archa Fox
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Alexander Swarbrick
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Stefan Hüttelmaier
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Maria Kavallaris
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Giovanni Perini
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - John S Mattick
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Marcel E Dinger
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Tao Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL).
| |
Collapse
|
224
|
P53-regulated long non-coding RNA TUG1 affects cell proliferation in human non-small cell lung cancer, partly through epigenetically regulating HOXB7 expression. Cell Death Dis 2014; 5:e1243. [PMID: 24853421 PMCID: PMC4047917 DOI: 10.1038/cddis.2014.201] [Citation(s) in RCA: 355] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 04/03/2014] [Accepted: 04/08/2014] [Indexed: 01/01/2023]
Abstract
Recently, a novel class of transcripts, long non-coding RNAs (lncRNAs), is being identified at a rapid pace. These RNAs have critical roles in diverse biological processes, including tumorigenesis. Here we report that taurine-upregulated gene 1 (TUG1), a 7.1-kb lncRNA, recruiting and binding to polycomb repressive complex 2 (PRC2), is generally downregulated in non-small cell lung carcinoma (NSCLC) tissues. In a cohort of 192 NSCLC patients, the lower expression of TUG1 was associated with a higher TNM stage and tumor size, as well as poorer overall survival (P<0.001). Univariate and multivariate analyses revealed that TUG1 expression serves as an independent predictor for overall survival (P<0.001). Further experiments revealed that TUG1 expression was induced by p53, and luciferase and chromatin immunoprecipitation (ChIP) assays confirmed that TUG1 was a direct transcriptional target of p53. TUG1 knockdown significantly promoted the proliferation in vitro and in vivo. Moreover, the lncRNA-mediated regulation of the expression of HOX genes in tumorigenesis and development has been recently receiving increased attention. Interestingly, inhibition of TUG1 could upregulate homeobox B7 (HOXB7) expression; ChIP assays demonstrated that the promoter of HOXB7 locus was bound by EZH2 (enhancer of zeste homolog 2), a key component of PRC2, and was H3K27 trimethylated. This TUG1-mediated growth regulation is in part due to specific modulation of HOXB7, thus participating in AKT and MAPK pathways. Together, these results suggest that p53-regulated TUG1 is a growth regulator, which acts in part through control of HOXB7. The p53/TUG1/PRC2/HOXB7 interaction might serve as targets for NSCLC diagnosis and therapy.
Collapse
|
225
|
Khorkova O, Myers AJ, Hsiao J, Wahlestedt C. Natural antisense transcripts. Hum Mol Genet 2014; 23:R54-63. [PMID: 24838284 DOI: 10.1093/hmg/ddu207] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Recent years have seen the increasing understanding of the crucial role of RNA in the functioning of the eukaryotic genome. These discoveries, fueled by the achievements of the FANTOM, and later GENCODE and ENCODE consortia, led to the recognition of the important regulatory roles of natural antisense transcripts (NATs) arising from what was previously thought to be 'junk DNA'. Roughly defined as non-coding regulatory RNA transcribed from the opposite strand of a coding gene locus, NATs are proving to be a heterogeneous group with high potential for therapeutic application. Here, we attempt to summarize the rapidly growing knowledge about this important non-coding RNA subclass.
Collapse
Affiliation(s)
- Olga Khorkova
- OPKO Health Inc., 10320 USA Today Way, Miramar, FL 33025, USA
| | | | - Jane Hsiao
- OPKO Health Inc., 10320 USA Today Way, Miramar, FL 33025, USA
| | - Claes Wahlestedt
- Department of Psychiatry and Behavioral Sciences and Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
226
|
Editing our way to regeneration. Cell Tissue Res 2014; 356:533-7. [PMID: 24803027 DOI: 10.1007/s00441-014-1844-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 02/07/2014] [Indexed: 10/25/2022]
Abstract
Transcription is the primary regulatory step to gene expression. However, there are numerous post-transcriptional mechanisms that are also crucial for developing the transcritptome, and the subsequent proteome, signature of any physiological setting. Organ and tissue regeneration is one such physiological setting that requires the rapid development of an environment that can supply all of the necessary molecular and cellular signalling needs necessary to attenuate infection, remove dead or necrotic cells, provide structural stability and finally replenish the compromised area with functional cells. The post-transcriptional regulatory mechanisms that have the ability to heavily influence the molecular and cellular pathways associated with regeneration are slowly being characterized. This mini-review will further clarify the possible regulation of regeneration through adenosine-to-inosine (A-I) RNA editing; a post-transcriptional mechanism that can affect the molecular and cellular pathways associated with functional restoration of damaged tissues and organs through discrete nucleotide changes in RNA transcripts. It is hoped that the intriguing links made between A-I RNA editing and regeneration in this mini-review will encourage further comparative studies into this infant field of research.
Collapse
|
227
|
Li DK, Tisdale S, Lotti F, Pellizzoni L. SMN control of RNP assembly: from post-transcriptional gene regulation to motor neuron disease. Semin Cell Dev Biol 2014; 32:22-9. [PMID: 24769255 DOI: 10.1016/j.semcdb.2014.04.026] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/17/2014] [Indexed: 11/25/2022]
Abstract
At the post-transcriptional level, expression of protein-coding genes is controlled by a series of RNA regulatory events including nuclear processing of primary transcripts, transport of mature mRNAs to specific cellular compartments, translation and ultimately, turnover. These processes are orchestrated through the dynamic association of mRNAs with RNA binding proteins and ribonucleoprotein (RNP) complexes. Accurate formation of RNPs in vivo is fundamentally important to cellular development and function, and its impairment often leads to human disease. The survival motor neuron (SMN) protein is key to this biological paradigm: SMN is essential for the biogenesis of various RNPs that function in mRNA processing, and genetic mutations leading to SMN deficiency cause the neurodegenerative disease spinal muscular atrophy. Here we review the expanding role of SMN in the regulation of gene expression through its multiple functions in RNP assembly. We discuss advances in our understanding of SMN activity as a chaperone of RNPs and how disruption of SMN-dependent RNA pathways can cause motor neuron disease.
Collapse
Affiliation(s)
- Darrick K Li
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Sarah Tisdale
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Francesco Lotti
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Livio Pellizzoni
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
228
|
Abstract
The ability to sequence genomes and characterize their products has begun to reveal the central role for regulatory RNAs in biology, especially in complex organisms. It is now evident that the human genome contains not only protein-coding genes, but also tens of thousands of non-protein coding genes that express small and long ncRNAs (non-coding RNAs). Rapid progress in characterizing these ncRNAs has identified a diverse range of subclasses, which vary widely in size, sequence and mechanism-of-action, but share a common functional theme of regulating gene expression. ncRNAs play a crucial role in many cellular pathways, including the differentiation and development of cells and organs and, when mis-regulated, in a number of diseases. Increasing evidence suggests that these RNAs are a major area of evolutionary innovation and play an important role in determining phenotypic diversity in animals.
Collapse
|
229
|
DNA Repair and Cell Differentiation—Does Getting Older Means Getting Wiser as Well? BIOTECHNOL BIOTEC EQ 2014. [DOI: 10.2478/v10133-010-0039-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
230
|
Sun Q, Liu H, Chen Z. The fine tuning role of microRNA-RNA interaction in odontoblast differentiation and disease. Oral Dis 2014; 21:142-8. [DOI: 10.1111/odi.12237] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/26/2014] [Accepted: 03/12/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Q Sun
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM); School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - H Liu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM); School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - Z Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM); School and Hospital of Stomatology; Wuhan University; Wuhan China
| |
Collapse
|
231
|
Luk ACS, Chan WY, Rennert OM, Lee TL. Long noncoding RNAs in spermatogenesis: insights from recent high-throughput transcriptome studies. Reproduction 2014; 147:R131-41. [PMID: 24713396 DOI: 10.1530/rep-13-0594] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Spermatogenesis is a complex developmental process in which undifferentiated spermatogonia are differentiated into spermatocytes and spermatids through two rounds of meiotic division and finally giving rise to mature spermatozoa (sperm). These processes involve many testis- or male germ cell-specific gene products that undergo strict developmental regulations. As a result, identifying critical, regulatory genes controlling spermatogenesis provide the clues not only to the regulatory mechanism of spermatogenesis at the molecular level, but also to the identification of candidate genes for infertility or contraceptives development. Despite the biological importance in male germ cell development, the underlying mechanisms of stage-specific gene regulation and cellular transition during spermatogenesis remain largely elusive. Previous genomic studies on transcriptome profiling were largely limited to protein-coding genes. Importantly, protein-coding genes only account for a small percentage of transcriptome; the majority are noncoding transcripts that do not translate into proteins. Although small noncoding RNAs (ncRNAs) such as microRNAs, siRNAs, and Piwi-interacting RNAs are extensively investigated in male germ cell development, the role of long ncRNAs (lncRNAs), commonly defined as ncRNAs longer than 200 bp, is relatively unexplored. Herein, we summarize recent transcriptome studies on spermatogenesis and show examples that a subset of noncoding transcript population, known as lncRNAs, constitutes a novel regulatory target in spermatogenesis.
Collapse
Affiliation(s)
- Alfred Chun-Shui Luk
- School of Biomedical Sciences, Room 622A, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | | | |
Collapse
|
232
|
|
233
|
An mRNA-derived noncoding RNA targets and regulates the ribosome. Mol Cell 2014; 54:147-155. [PMID: 24685157 PMCID: PMC3988847 DOI: 10.1016/j.molcel.2014.02.024] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/26/2014] [Accepted: 02/12/2014] [Indexed: 12/02/2022]
Abstract
The structural and functional repertoire of small non-protein-coding RNAs (ncRNAs) is central for establishing gene regulation networks in cells and organisms. Here, we show that an mRNA-derived 18-nucleotide-long ncRNA is capable of downregulating translation in Saccharomyces cerevisiae by targeting the ribosome. This 18-mer ncRNA binds to polysomes upon salt stress and is crucial for efficient growth under hyperosmotic conditions. Although the 18-mer RNA originates from the TRM10 locus, which encodes a tRNA methyltransferase, genetic analyses revealed the 18-mer RNA nucleotide sequence, rather than the mRNA-encoded enzyme, as the translation regulator. Our data reveal the ribosome as a target for a small regulatory ncRNA and demonstrate the existence of a yet unkown mechanism of translation regulation. Ribosome-targeted small ncRNAs are found in all domains of life and represent a prevalent but so far largely unexplored class of regulatory molecules. This study reveals the yeast ribosome as direct target for small regulatory ncRNAs An 18-nt-long exon-derived RNA fragment from the TRM10 locus binds to ribosomes This 18-mer ncRNA inhibits global protein biosynthesis in vivo and in vitro This translation attenuation is crucial for adaption under hyperosmotic stress
Collapse
|
234
|
Zhang EB, Han L, Yin DD, Kong R, De W, Chen J. c-Myc-induced, long, noncoding H19 affects cell proliferation and predicts a poor prognosis in patients with gastric cancer. Med Oncol 2014; 31:914. [PMID: 24671855 DOI: 10.1007/s12032-014-0914-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/06/2014] [Indexed: 01/01/2023]
Abstract
Gastric cancer (GC) is one of the most frequent cancers worldwide. Recent studies have shown that long noncoding RNAs (lncRNAs) play critical roles in multiple biological processes, including oncogenesis. The present study aimed to evaluate the potential role of lncRNA H19 in GC. qRT-PCR was performed to investigate the expression of H19 in tumor tissues and corresponding non-tumor lung tissues from 80 patients with GC and in GC cell lines. The Kaplan-Meier method and Cox proportional hazards analysis were used to evaluate the association between H19 expression and overall survival time (OS). The biological significance of H19 was evaluated using siRNAs in vitro. We also constructed a c-Myc plasmid to investigate the cause of the altered expression of H19 in the progression of GC. The results show that lncRNA H19 is overexpressed in tumor tissues compared with adjacent normal tissues. An advanced tumor-node-metastasis stage was positively correlated with increased H19 expression (P < 0.001), and a high H19 expression was associated with poor OS and can be regarded as an independent predictor of the OS of GC patients (P = 0.042). MTT and colony formation assays confirmed that H19 expression affects GC cell proliferation in vitro. Furthermore, exogenous c-Myc significantly induces H19 expression, and the expression of H19 was positively correlated with the c-Myc levels in the 80 samples used in our study (Pearson correlation coefficient = -0.687). In conclusion, our study demonstrates that the altered expression of lncRNA H19, which is induced by c-Myc, is involved in the development and progression of GC by regulating cell proliferation and shows that H19 may be a potential diagnostic and prognostic target in patients with GC.
Collapse
Affiliation(s)
- Er-Bao Zhang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
235
|
Hu YW, Yang JY, Ma X, Chen ZP, Hu YR, Zhao JY, Li SF, Qiu YR, Lu JB, Wang YC, Gao JJ, Sha YH, Zheng L, Wang Q. A lincRNA-DYNLRB2-2/GPR119/GLP-1R/ABCA1-dependent signal transduction pathway is essential for the regulation of cholesterol homeostasis. J Lipid Res 2014; 55:681-97. [PMID: 24493833 DOI: 10.1194/jlr.m044669] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Accumulated evidence shows that G protein-coupled receptor 119 (GPR119) plays a key role in glucose and lipid metabolism. Here, we explored the effect of GPR119 on cholesterol metabolism and inflammation in THP-1 macrophages and atherosclerotic plaque progression in apoE(-/-) mice. We found that oxidized LDL (Ox-LDL) significantly induced long intervening noncoding RNA (lincRNA)-DYNLRB2-2 expression, resulting in the upregulation of GPR119 and ABCA1 expression through the glucagon-like peptide 1 receptor signaling pathway. GPR119 significantly decreased cellular cholesterol content and increased apoA-I-mediated cholesterol efflux in THP-1 macrophage-derived foam cells. In vivo, apoE(-/-) mice were randomly divided into two groups and infected with lentivirus (LV)-Mock or LV-GPR119 for 8 weeks. GPR119-treated mice showed decreased liver lipid content and plasma TG, interleukin (IL)-1β, IL-6, and TNF-α levels, whereas plasma levels of apoA-I were significantly increased. Consistent with this, atherosclerotic lesion development was significantly inhibited by infection of apoE(-/-) mice with LV-GPR119. Our findings clearly indicate that, Ox-LDL significantly induced lincRNA-DYNLRB2-2 expression, which promoted ABCA1-mediated cholesterol efflux and inhibited inflammation through GPR119 in THP-1 macrophage-derived foam cells. Moreover, GPR119 decreased lipid and serum inflammatory cytokine levels, decreasing atherosclerosis in apoE(-/-) mice. These suggest that GPR119 may be a promising candidate as a therapeutic agent.
Collapse
Affiliation(s)
- Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Swaminathan S, Murray DD, Kelleher AD. miRNAs and HIV: unforeseen determinants of host-pathogen interaction. Immunol Rev 2014; 254:265-80. [PMID: 23772625 DOI: 10.1111/imr.12077] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Our understanding of the complexity of gene regulation has significantly improved in the last decade as the role of small non-coding RNAs, called microRNAs (miRNAs), has been appreciated. These 19-22 nucleotide RNA molecules are critical regulators of mRNA translation and turnover. The miRNAs bind via a protein complex to the 3' untranslated region (3' UTR) of mRNA, ultimately leading to mRNA translational inhibition, degradation, or repression. Although many mechanisms by which human immunodeficiency virus-1 (HIV-1) infection eventually induces catastrophic immune destruction have been elucidated, the important role that miRNAs play in HIV-1 pathogenesis is only now emerging. Accumulating evidence demonstrates that changes to endogenous miRNA levels following infection is important: in maintaining HIV-1 latency in resting CD4(+) T cells, potentially affect immune function via changes to cytokines such as interleukin-2 (IL-2) and IL-10 and may predict disease progression. We review the roles that both viral and host miRNAs play in different cell types and disease conditions that are important in HIV-1 infection and discuss how miRNAs affect key immunomodulatory molecules contributing to immune dysfunction. Further, we discuss whether miRNAs may be used as novel biomarkers in serum and the potential to modulate miRNA levels as a unique approach to combating this pathogen.
Collapse
Affiliation(s)
- Sanjay Swaminathan
- Laboratory of Human Retrovirology, Applied and Developmental Research Directorate (ADD), Science Application International Corporation (SAIC)-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | |
Collapse
|
237
|
Washietl S, Kellis M, Garber M. Evolutionary dynamics and tissue specificity of human long noncoding RNAs in six mammals. Genome Res 2014; 24:616-28. [PMID: 24429298 PMCID: PMC3975061 DOI: 10.1101/gr.165035.113] [Citation(s) in RCA: 294] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Long intergenic noncoding RNAs (lincRNAs) play diverse regulatory roles in human development and disease, but little is known about their evolutionary history and constraint. Here, we characterize human lincRNA expression patterns in nine tissues across six mammalian species and multiple individuals. Of the 1898 human lincRNAs expressed in these tissues, we find orthologous transcripts for 80% in chimpanzee, 63% in rhesus, 39% in cow, 38% in mouse, and 35% in rat. Mammalian-expressed lincRNAs show remarkably strong conservation of tissue specificity, suggesting that it is selectively maintained. In contrast, abundant splice-site turnover suggests that exact splice sites are not critical. Relative to evolutionarily young lincRNAs, mammalian-expressed lincRNAs show higher primary sequence conservation in their promoters and exons, increased proximity to protein-coding genes enriched for tissue-specific functions, fewer repeat elements, and more frequent single-exon transcripts. Remarkably, we find that ∼20% of human lincRNAs are not expressed beyond chimpanzee and are undetectable even in rhesus. These hominid-specific lincRNAs are more tissue specific, enriched for testis, and faster evolving within the human lineage.
Collapse
Affiliation(s)
- Stefan Washietl
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02140, USA
| | | | | |
Collapse
|
238
|
The ribosomal protein rpl26 promoter is required for its 3' sense terminus ncRNA transcription in Schizosaccharomyces pombe, implicating a new transcriptional mechanism for ncRNAs. Biochem Biophys Res Commun 2014; 444:86-91. [PMID: 24434141 DOI: 10.1016/j.bbrc.2014.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 01/09/2014] [Indexed: 11/20/2022]
Abstract
Transcriptome studies have revealed that many non-coding RNAs (ncRNAs) are located near the 3' sense terminus of protein-coding genes. However, the transcription and function of these RNAs remain elusive. Here, we identify a 3' sense termini-associated sRNA (TASR) downstream of rpl26 in Schizosaccharomyces pombe (S. pombe). Structure and function assays indicate that the TASR is an H/ACA box snoRNA required for 18S rRNA pseudouridylation at U121 and U305 sites and is therefore a cognate of snR49 from the budding yeast. Transcriptional studies show that pre-snR49 overlaps most of the coding sequence (CDS) of rpl26. Using scanning deletion analysis within promoter region, we show that the rpl26 promoter is required for the 3' TASR transcription. Interestingly, chromosomal synteny of rpl26-snR49 is found in the Schizosaccharomyces groups. Taken together, we have revealed a new transcriptional mechanism for 3' sense TASRs, which are transcribed by the same promoter as their upstream protein genes. These results further suggest that the origin and function of 3' sense ncRNAs are associated with upstream genes in higher eukaryotes.
Collapse
|
239
|
Jašarević E, Geary DC, Rosenfeld CS. Sexually selected traits: a fundamental framework for studies on behavioral epigenetics. ILAR J 2014; 53:253-69. [PMID: 23744965 DOI: 10.1093/ilar.53.3-4.253] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence suggests that epigenetic-based mechanisms contribute to various aspects of sex differences in brain and behavior. The major obstacle in establishing and fully understanding this linkage is identifying the traits that are most susceptible to epigenetic modification. We have proposed that sexual selection provides a conceptual framework for identifying such traits. These are traits involved in intrasexual competition for mates and intersexual choice of mating partners and generally entail a combination of male-male competition and female choice. These behaviors are programmed during early embryonic and postnatal development, particularly during the transition from the juvenile to adult periods, by exposure of the brain to steroid hormones, including estradiol and testosterone. We evaluate the evidence that endocrine-disrupting compounds, including bisphenol A, can interfere with the vital epigenetic and gene expression pathways and with the elaboration of sexually selected traits with epigenetic mechanisms presumably governing the expression of these traits. Finally, we review the evidence to suggest that these steroid hormones can induce a variety of epigenetic changes in the brain, including the extent of DNA methylation, histone protein alterations, and even alterations of noncoding RNA, and that many of the changes differ between males and females. Although much previous attention has focused on primary sex differences in reproductive behaviors, such as male mounting and female lordosis, we outline why secondary sex differences related to competition and mate choice might also trace their origins back to steroid-induced epigenetic programming in disparate regions of the brain.
Collapse
Affiliation(s)
- Eldin Jašarević
- Department of Psychological Sciences, the Interdisciplinary Neuroscience Program, and the Bond Life Sciences Center, University of Missouri, Columbia 65211, USA
| | | | | |
Collapse
|
240
|
Rossi MN, Antonangeli F. LncRNAs: New Players in Apoptosis Control. Int J Cell Biol 2014; 2014:473857. [PMID: 24627686 PMCID: PMC3929073 DOI: 10.1155/2014/473857] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/22/2013] [Indexed: 12/26/2022] Open
Abstract
The discovery that the mammalian genome is largely transcribed and that almost half of the polyadenylated RNAs is composed of noncoding RNAs has attracted the attention of the scientific community. Growing amount of data suggests that long noncoding RNAs (lncRNAs) are a new class of regulators involved not only in physiological processes, such as imprinting and differentiation, but also in cancer progression and neurodegeneration. Apoptosis is a well regulated type of programmed cell death necessary for correct organ development and tissue homeostasis. Indeed, cancer cells often show an inhibition of the apoptotic pathways and it is now emerging that overexpression or downregulation of different lncRNAs in specific types of tumors sensitize cancer cells to apoptotic stimuli. In this review we summarize the latest studies on lncRNAs and apoptosis with major attention to those performed in cancer cells and in healthy cells upon differentiation. We discuss the new perspectives of using lncRNAs as targets of anticancer drugs. Finally, considering that lncRNA levels have been reported to have a correlation with specific cancer types, we argue the possibility of using lncRNAs as tumor biomarkers.
Collapse
Affiliation(s)
- Marianna Nicoletta Rossi
- 1Pasteur Institute-Fondazione Cenci Bolognetti, Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Fabrizio Antonangeli
- 2Pasteur Institute-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
- *Fabrizio Antonangeli:
| |
Collapse
|
241
|
David L, Clauder-Münster S, Steinmetz LM. High-density tiling microarray analysis of the full transcriptional activity of yeast. Methods Mol Biol 2014; 1205:257-73. [PMID: 25213250 DOI: 10.1007/978-1-4939-1363-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Understanding the relationship between DNA sequence variation and phenotypic variation in complex or quantitative traits is one of the major challenges in modern biology. We are witnessing a deluge of DNA sequence information and association studies of genetic polymorphisms with phenotypes of interest in families and populations. In addition, it has become clear that large portions of eukaryotic genomes beyond protein-coding genes are transcribed, generating numerous noncoding RNA (ncRNA) molecules whose functions remain mostly unknown.DNA oligonucleotide microarrays constitute a powerful technology for studying the expression of genes in different organisms. The Saccharomyces cerevisiae tiling array presents a significant advance over previous array-based platforms. It has a high density of overlapping probes that start on average every 8 bp along each strand of the genome, enabling precise definition of transcript structure. Furthermore, the array includes probes specific for the polymorphic positions of another, distantly related yeast strain, allowing accurate measurement of allele-specific expression in a hybrid of the two strains. This technology thus allows high-resolution, quantitative, strand- and allele-specific measurements of transcription from a full eukaryotic genome. In this chapter, we describe the methods for extracting RNA, synthesizing first-strand cDNA, fragmenting, and labeling of samples for hybridization to the tiling array. Combining genome-wide information on variation in DNA sequence with variation in transcript structure and levels promises to increase our understanding of the genotype-to-phenotype relationship.
Collapse
Affiliation(s)
- Lior David
- Department of Animal Sciences, R.H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | | | | |
Collapse
|
242
|
Kaeuferle T, Bartel S, Dehmel S, Krauss-Etschmann S. MicroRNA methodology: advances in miRNA technologies. Methods Mol Biol 2014; 1169:121-30. [PMID: 24957235 DOI: 10.1007/978-1-4939-0882-0_12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
There is an emerging trend in microRNA research and thus substantial progress in microRNA technologies. In this chapter we provide insights into the main microRNA specific methodologies and critical steps of microRNA expression profiling, target gene identification, and functional confirmation of microRNA effects up to in vivo application of antagomirs.
Collapse
Affiliation(s)
- Theresa Kaeuferle
- Dr. von Hauner'sche Kinderklinik, Klinikum der Universität München, 80337, Munich, Germany
| | | | | | | |
Collapse
|
243
|
Winterling C, Koch M, Koeppel M, Garcia-Alcalde F, Karlas A, Meyer TF. Evidence for a crucial role of a host non-coding RNA in influenza A virus replication. RNA Biol 2013; 11:66-75. [PMID: 24440876 DOI: 10.4161/rna.27504] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence suggests the non-protein coding human genome is of vital importance for human cell function. Besides small RNAs, the diverse class of long non-coding RNAs (lncRNAs) recently came into focus. However, their relevance for infection, a major evolutionary driving force, remains elusive. Using two commercially available microarray systems, namely NCode™ and Sureprint™ G3, we identified differential expression of 42 ncRNAs during influenza A virus (IAV) infection in human lung epithelial cells. This included several classes of lncRNAs, including large intergenic ncRNAs (lincRNAs). As analyzed by qRT-PCR, expression of one lincRNA, which we termed virus inducible lincRNA (VIN), is induced by several IAV strains (H1N1, H3N2, H7N7) as well as vesicular stomatitis virus. However, we did not observe an induction of VIN by influenza B virus, treatment with RNA mimics, or IFNβ. Thus, VIN expression seems to be a specific response to certain viral infections. RNA fractionation and RNA-FISH experiments revealed that VIN is localized to the host cell nucleus. Most importantly, we show that abolition of VIN by RNA interference restricts IAV replication and viral protein synthesis, highlighting the relevance of this lincRNA for productive IAV infection. Our observations suggest that viral pathogens interfere with the non-coding portion of the human genome, thereby guaranteeing their successful propagation, and that the expression of VIN correlates with their virulence. Consequently, our study provides a novel approach for understanding virus pathogenesis in greater detail, which will enable future design of new antiviral strategies targeting the host's non-protein coding genome.
Collapse
Affiliation(s)
- Carla Winterling
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Manuel Koch
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Max Koeppel
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Fernando Garcia-Alcalde
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Alexander Karlas
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| |
Collapse
|
244
|
Abstract
Post-transcriptional cleavage of RNA molecules to generate smaller fragments is a widespread mechanism that enlarges the structural and functional complexity of cellular RNomes. Substrates for such RNA fragmentations are coding as well as non-protein-coding RNAs. In particular, fragments derived from both precursor and mature tRNAs represent one of the rapidly growing classes of post-transcriptional RNA pieces. Importantly, these tRNA fragments possess distinct expression patterns, abundance, cellular localizations, or biological roles compared with their parental tRNA molecules. Here we review recent reports on tRNA cleavage and attempt to categorize tRNA pieces according to their origin and cellular function. The biological scope of tRNA-derived fragments ranges from translation control, over RNA silencing, to regulating apoptosis, and thus clearly enlarges the functional repertoire of ncRNA biology.
Collapse
Affiliation(s)
- Jennifer Gebetsberger
- Department of Chemistry and Biochemistry; University of Bern; Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences; University of Bern; Bern, Switzerland
| | - Norbert Polacek
- Department of Chemistry and Biochemistry; University of Bern; Bern, Switzerland; Division of Genomics and RNomics; Medical University Innsbruck; Innsbruck, Austria
| |
Collapse
|
245
|
Takahashi Y, Sawada G, Kurashige J, Uchi R, Matsumura T, Ueo H, Takano Y, Eguchi H, Sudo T, Sugimachi K, Yamamoto H, Doki Y, Mori M, Mimori K. Amplification of PVT-1 is involved in poor prognosis via apoptosis inhibition in colorectal cancers. Br J Cancer 2013; 110:164-71. [PMID: 24196785 PMCID: PMC3887297 DOI: 10.1038/bjc.2013.698] [Citation(s) in RCA: 256] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 12/16/2022] Open
Abstract
Background: We previously conducted gene expression microarray analyses to identify novel indicators for colorectal cancer (CRC) metastasis and prognosis from which we identified PVT-1 as a candidate gene. PVT-1, which encodes a long noncoding RNA, mapped to chromosome 8q24 whose copy-number amplification is one of the most frequent events in a wide variety of malignant diseases. However, PVT-1 molecular mechanism of action remains unclear. Methods: We conducted cell proliferation and invasion assays using colorectal cancer cell lines transfected with PVT-1siRNA or negative control siRNA. Gene expression microarray analyses on these cell lines were also carried out to investigate the molecular function of PVT-1. Further, we investigated the impact of PVT-1 expression on the prognosis of 164 colorectal cancer patients by qRT–PCR. Results: CRC cells transfected with PVT-1 siRNA exhibited significant loss of their proliferation and invasion capabilities. In these cells, the TGF-β signalling pathway and apoptotic signals were significantly activated. In addition, univariate and multivariate analysis revealed that PVT-1 expression level was an independent risk factor for overall survival of colorectal cancer patients. Conclusion: PVT-1, which maps to 8q24, generates antiapoptotic activity in CRC, and abnormal expression of PVT-1 was a prognostic indicator for CRC patients.
Collapse
Affiliation(s)
- Y Takahashi
- 1] Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan [2] Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - G Sawada
- 1] Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan [2] Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - J Kurashige
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan
| | - R Uchi
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan
| | - T Matsumura
- 1] Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan [2] Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - H Ueo
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan
| | - Y Takano
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan
| | - H Eguchi
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan
| | - T Sudo
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan
| | - K Sugimachi
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan
| | - H Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Y Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - M Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - K Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara 4546, Beppu 874-0838, Japan
| |
Collapse
|
246
|
Abstract
Long non-coding RNAs (lncRNAs) are transcripts longer than ~200 nucleotides with little or no protein-coding capacity. Growing evidence shows that lncRNAs present important function in development and are associated with many human diseases such as cancers, Alzheimer disease, and heart diseases. Transcribed ultraconserved region (T-UCR) transcripts are a novel class of lncRNAs transcribed from ultraconserved regions (UCRs). UCRs are absolutely conserved (100%) between the orthologous regions of the human, rat, and mouse genomes. The UCRs are frequently located at fragile sites and at genomic regions involved in cancers. Recent data suggest that T-UCRs are altered at the transcriptional level in human tumorigenesis and the aberrant T-UCRs expression profiles can be used to differentiate human cancer types. The profound understanding of T-UCRs can throw new light on the pathogenesis of human cancers.
Collapse
Affiliation(s)
- Jiang Chen Peng
- Department of Gastroenterology; Renji Hospital; Shanghai Jiao-Tong University School of Medicine; Shanghai Institute of Digestive Disease; Shanghai Inflammatory Bowel Disease Research Center; Shanghai, PR China
| | - Jun Shen
- Department of Gastroenterology; Renji Hospital; Shanghai Jiao-Tong University School of Medicine; Shanghai Institute of Digestive Disease; Shanghai Inflammatory Bowel Disease Research Center; Shanghai, PR China
| | - Zhi Hua Ran
- Department of Gastroenterology; Renji Hospital; Shanghai Jiao-Tong University School of Medicine; Shanghai Institute of Digestive Disease; Shanghai Inflammatory Bowel Disease Research Center; Shanghai, PR China
| |
Collapse
|
247
|
Gualtieri A, Andreola F, Sciamanna I, Sinibaldi-Vallebona P, Serafino A, Spadafora C. Increased expression and copy number amplification of LINE-1 and SINE B1 retrotransposable elements in murine mammary carcinoma progression. Oncotarget 2013; 4:1882-1893. [PMID: 24231191 PMCID: PMC3875756 DOI: 10.18632/oncotarget.1188] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 08/07/2013] [Indexed: 12/13/2022] Open
Abstract
In higher eukaryotic genomes, Long Interspersed Nuclear Element 1 (LINE-1) retrotransposons and endogenous retroviruses represent large families of repeated elements encoding reverse transcriptase (RT) proteins. Short Interspersed Nuclear Element B1 (SINE B1) retrotrasposons do not encode RT, but use LINE-1-derived RT for their retrotransposition. We previously showed that many cancer types have an abundant endogenous RT activity. Inhibition of that activity, by either RNA interference-dependent silencing of active LINE-1 elements or by RT inhibitory drugs, reduced proliferation and promoted differentiation in cancer cells, indicating that LINE-1-encoded RT is required for tumor progression. Using MMTV-PyVT transgenic mice as a well-defined model of breast cancer progression, we now report that both LINE-1 and SINE B1 retrotransposons are up-regulated at a very early stage of tumorigenesis; LINE-1-encoded RT product and enzymatic activity were detected in tumor tissues as early as stage 1, preceding the widespread appearance of histological alterations and specific cancer markers, and further increased in later progression stages, while neither was present in non-pathological breast tissues. Importantly, both LINE-1 and SINE B1 retrotransposon families undergo copy number amplification during tumor progression. These findings therefore indicate that RT activity is distinctive of breast cancer cells and that, furthermore, LINE-1 and SINE B1 undergo copy number amplification during cancer progression.
Collapse
|
248
|
Yoshihama M, Nakao A, Kenmochi N. snOPY: a small nucleolar RNA orthological gene database. BMC Res Notes 2013; 6:426. [PMID: 24148649 PMCID: PMC4015994 DOI: 10.1186/1756-0500-6-426] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/21/2013] [Indexed: 02/05/2023] Open
Abstract
Background Small nucleolar RNAs (snoRNAs) are a class of non-coding RNAs that guide the modification of specific nucleotides in ribosomal RNAs (rRNAs) and small nuclear RNAs (snRNAs). Although most non-coding RNAs undergo post-transcriptional modifications prior to maturation, the functional significance of these modifications remains unknown. Here, we introduce the snoRNA orthological gene database (snOPY) as a tool for studying RNA modifications. Findings snOPY provides comprehensive information about snoRNAs, snoRNA gene loci, and target RNAs. It also contains data for orthologues from various species, which enables users to analyze the evolution of snoRNA genes. In total, 13,770 snoRNA genes, 10,345 snoRNA gene loci, and 133 target RNAs have been registered. Users can search and access the data efficiently using a simple web interface with a series of internal links. snOPY is freely available on the web at http://snoopy.med.miyazaki-u.ac.jp. Conclusions snOPY is the database that provides information about the small nucleolar RNAs and their orthologues. It will help users to study RNA modifications and snoRNA gene evolution.
Collapse
Affiliation(s)
| | | | - Naoya Kenmochi
- Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| |
Collapse
|
249
|
Liu Y, Luo D, Zhao H, Zhu Z, Hu W, Cheng CHK. Inheritable and precise large genomic deletions of non-coding RNA genes in zebrafish using TALENs. PLoS One 2013; 8:e76387. [PMID: 24130773 PMCID: PMC3794983 DOI: 10.1371/journal.pone.0076387] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 08/30/2013] [Indexed: 01/15/2023] Open
Abstract
Transcription activator-like effector nucleases (TALENs) have so far been applied to disrupt protein-coding genes which constitute only 2-3% of the genome in animals. The majority (70-90%) of the animal genome is actually transcribed as non-coding RNAs (ncRNAs), yet the lack of efficient tools to knockout ncRNA genes hinders studies on their in vivo functions. Here we have developed novel strategies using TALENs to achieve precise and inheritable large genomic deletions and knockout of ncRNA genes in zebrafish. We have demonstrated that individual miRNA genes could be disrupted using one pair of TALENs, whereas large microRNA (miRNA) gene clusters and long non-coding RNA (lncRNA) genes could be precisely deleted using two pairs of TALENs. We have generated large genomic deletions of two miRNA clusters (the 1.2 kb miR-17-92 cluster and the 79.8 kb miR-430 cluster) and one long non-coding RNA (lncRNA) gene (the 9.0 kb malat1), and the deletions are transmitted through the germline. Taken together, our results establish TALENs as a robust tool to engineer large genomic deletions and knockout of ncRNA genes, thus opening up new avenues in the application of TALENs to study the genome in vivo.
Collapse
Affiliation(s)
- Yun Liu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences Core Laboratory, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Daji Luo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- Department of Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Hui Zhao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences Core Laboratory, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Zuoyan Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Wei Hu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- * E-mail: (WH); (CC)
| | - Christopher H. K. Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences Core Laboratory, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- * E-mail: (WH); (CC)
| |
Collapse
|
250
|
Long non-coding RNA-guided regulation in organisms. SCIENCE CHINA-LIFE SCIENCES 2013; 56:891-6. [DOI: 10.1007/s11427-013-4558-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 09/16/2013] [Indexed: 12/24/2022]
|