551
|
He X, He Y, Xi B, Zheng J, Zeng X, Cai Q, OuYang Y, Wang C, Zhou X, Huang H, Deng W, Xin S, Huang Q, Liu H. LncRNAs expression in preeclampsia placenta reveals the potential role of LncRNAs contributing to preeclampsia pathogenesis. PLoS One 2013; 8:e81437. [PMID: 24312300 PMCID: PMC3842959 DOI: 10.1371/journal.pone.0081437] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 10/14/2013] [Indexed: 01/08/2023] Open
Abstract
Background Long non-coding RNAs (lncRNAs) are an important class of pervasive genes involved in a variety of biological functions. They are aberrantly expressed in many types of diseases. In this study, we aimed to investigate the lncRNA profiles in preeclampsia. Preeclampsia has been observed in patients with molar pregnancy where a fetus is absent, which demonstrate that the placenta is sufficient to cause this condition. Thus, we analyzed the lncRNA profiles in preeclampsia placentas. Methodology/Principal Findings In this study, we described the lncRNA profiles in six preeclampsia placentas (T) and five normal pregnancy placentas (N) using microarray. With abundant and varied probes accounting for 33,045 LncRNAs in our microarray, 28,443 lncRNAs that were expressed at a specific level were detected. From the data, we found 738 lncRNAs that were differentially expressed (≥1.5-fold-change) among preeclampsia placentas compared with controls. Coding-non-coding gene co-expression networks (CNC network) were constructed based on the correlation analysis between the differentially expressed lncRNAs and mRNAs. According to the CNC network and GO analysis of differentially expressed lncRNAs/mRNAs, we selected three lncRNAs to analyze the relationship between lncRNAs and preeclampsia. LOC391533, LOC284100, and CEACAMP8 were evaluated using qPCR in 40 preeclampsia placentas and 40 controls. These results revealed that three lncRNAs were aberrantly expressed in preeclampsia placentas compared with controls. Conclusions/Significance Our study is the first study to determine the genome-wide lncRNAs expression patterns in preeclampsia placenta using microarray. These results revealed that clusters of lncRNAs were aberrantly expressed in preeclampsia placenta compared with controls, which indicated that lncRNAs differentially expressed in preeclampsia placenta might play a partial or key role in preeclampsia development. Misregulation of LOC391533, LOC284100, and CEACAMP8 might contribute to the mechanism underlying preeclampsia. Taken together, this study may provide potential targets for the future treatment of preeclampsia and novel insights into preeclampsia biology.
Collapse
Affiliation(s)
- Xiaoju He
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- * (XH); (HL)
| | - Yinyan He
- Department of Obstetrics & Gynecology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Binrong Xi
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Jiusheng Zheng
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Xiaoming Zeng
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Qinhua Cai
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Yu OuYang
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Chen Wang
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Xiaofei Zhou
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Huiying Huang
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Wei Deng
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Siming Xin
- School of Medicine, Nanchang University, Nanchang, China
| | - Qixiang Huang
- School of Medicine, Nanchang University, Nanchang, China
| | - Huai Liu
- Department of Obstetrics, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- * (XH); (HL)
| |
Collapse
|
552
|
LncRNA pathway involved in premature preterm rupture of membrane (PPROM): an epigenomic approach to study the pathogenesis of reproductive disorders. PLoS One 2013; 8:e79897. [PMID: 24312190 PMCID: PMC3842261 DOI: 10.1371/journal.pone.0079897] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 09/26/2013] [Indexed: 12/11/2022] Open
Abstract
Preterm birth (PTB) is a live birth delivered before 37 weeks of gestation (GW). About one-third of PTBs result from the preterm premature rupture of membranes (PPROM). Up to the present, the pathogenic mechanisms underlying PPROM are not clearly understood. Here, we investigated the differential expression of long chain non-coding RNAs (lncRNAs) in placentas of PTBs with PPROM, and their possible involvement in the pathogenic pathways leading to PPROM. A total number of 1954, 776, and 1050 lncRNAs were identified with a microarray from placentas of PPROM (group A), which were compared to full-term birth (FTB) (group B), PTB (group C), and premature rupture of membrane (PROM) (group D) at full-term, respectively. Instead of investigating the individual pathogenic role of each lncRNA involved in the molecular mechanism underlying PPROM, we have focused on investigating the metabolic pathways and their functions to explore what is the likely association and how they are possibly involved in the development of PPROM. Six groups, including up-regulation and down-regulation in the comparisons of A vs. B, A vs. C, and A vs. D, of pathways were analyzed. Our results showed that 22 pathways were characterized as up-regulated 7 down-regulated in A vs. C, 18 up-regulated and 15 down-regulated in A vs. D, and 33 up-regulated and 7 down-regulated in A vs. B. Functional analysis showed pathways of infection and inflammatory response, ECM-receptor interactions, apoptosis, actin cytoskeleton, and smooth muscle contraction are the major pathogenic mechanisms involved in the development of PPROM. Characterization of these pathways through identification of lncRNAs opened new avenues for further investigating the epigenomic mechanisms of lncRNAs in PPROM as well as PTB.
Collapse
|
553
|
Rao P, Benito E, Fischer A. MicroRNAs as biomarkers for CNS disease. Front Mol Neurosci 2013; 6:39. [PMID: 24324397 PMCID: PMC3840814 DOI: 10.3389/fnmol.2013.00039] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/31/2013] [Indexed: 01/23/2023] Open
Abstract
For many neurological diseases, the efficacy and outcome of treatment depend on early detection. Diagnosis is currently based on the detection of symptoms and neuroimaging abnormalities, which appear at relatively late stages in the pathogenesis. However, the underlying molecular responses to genetic and environmental insults begin much earlier and non-coding RNA networks are critically involved in these cellular regulatory mechanisms. Profiling RNA expression patterns could thus facilitate presymptomatic disease detection. Obtaining indirect readouts of pathological processes is particularly important for brain disorders because of the lack of direct access to tissue for molecular analyses. Living neurons and other CNS cells secrete microRNA and other small non-coding RNA into the extracellular space packaged in exosomes, microvesicles, or lipoprotein complexes. This discovery, together with the rapidly evolving massive sequencing technologies that allow detection of virtually all RNA species from small amounts of biological material, has allowed significant progress in the use of extracellular RNA as a biomarker for CNS malignancies, neurological, and psychiatric diseases. There is also recent evidence that the interactions between external stimuli and brain pathological processes may be reflected in peripheral tissues, facilitating their use as potential diagnostic markers. In this review, we explore the possibilities and challenges of using microRNA and other small RNAs as a signature for neurodegenerative and other neuropsychatric conditions.
Collapse
Affiliation(s)
- Pooja Rao
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen Göttingen, Germany
| | | | | |
Collapse
|
554
|
Sun Y, Shen S, Tang H, Xiang J, Peng Y, Tang A, Li N, Zhou W, Wang Z, Zhang D, Xiang B, Ge J, Li G, Wu M, Li X. miR-429 identified by dynamic transcriptome analysis is a new candidate biomarker for colorectal cancer prognosis. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2013; 18:54-64. [PMID: 24237355 DOI: 10.1089/omi.2012.0132] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is a common malignant gastrointestinal cancer. Efforts for preventive and personalized medicine have intensified in the last decade with attention to novel forms of biomarkers. In the present study, microRNA and genetic analyses were performed in tandem for differential transcriptome profiling between primary tumors with or without nodes or distant metastases. Serial Test Cluster (STC) analysis demonstrated that 20 genes and two microRNAs showed distinctive expression patterns associated with the tumor, node, and metastasis (TNM) stage. The selected target genes were characterized by GO and Pathway analysis. A microRNA-target gene network analysis showed that miR-429 resided in the center of the network, indicating that miR-429 might serve important roles in the development of CRC. Real-time PCR and tissue microarrays showed that miR-429 had a dynamic expression pattern during the CRC progression stage, and was significantly downregulated in stage II and stage III clinical progression. The low expression of miR-429 was correlated with poor prognosis for CRC. Taken together, miR-429 warrant further clinical translation research as a candidate biomarker for CRC prognosis. Additional downstream targets and attendant gene function also need to be discerned to design a sound critical path to personalized medicine for persons susceptible to, or diagnosed with CRC.
Collapse
Affiliation(s)
- Yingnan Sun
- 1 Department of Gastroenterology, The Third Xiangya Hospital, Central South University , Changsha, Hunan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
555
|
Gadelha MR, Kasuki L, Dénes J, Trivellin G, Korbonits M. MicroRNAs: Suggested role in pituitary adenoma pathogenesis. J Endocrinol Invest 2013; 36:889-95. [PMID: 24317305 DOI: 10.1007/bf03346759] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that represent a major class of molecular regulators. miRNAs have been implicated in the pathogenesis of several human tumors, including pituitary adenomas. Altered expression of miRNAs has been described in pituitary adenomas, and specific miRNA signatures are related to clinical and therapeutic characteristics of the tumors. The data suggest that miRNAs influence various genes known to be associated with the pathogenesis of pituitary adenomas and in this review we summarize these currently available studies focusing on miRNAs in pituitary adenomas.
Collapse
Affiliation(s)
- M R Gadelha
- Division of Endocrinology, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
556
|
Feldstein O, Nizri T, Doniger T, Jacob J, Rechavi G, Ginsberg D. The long non-coding RNA ERIC is regulated by E2F and modulates the cellular response to DNA damage. Mol Cancer 2013; 12:131. [PMID: 24168400 PMCID: PMC4176120 DOI: 10.1186/1476-4598-12-131] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 10/01/2013] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The human genome encodes thousands of unique long non-coding RNAs (lncRNAs), and these transcripts are emerging as critical regulators of gene expression and cell fate. However, the transcriptional regulation of their expression is not fully understood. The pivotal transcription factor E2F1 which can induce both proliferation and cell death, is a critical downstream target of the tumor suppressor, RB. The retinoblastoma pathway is often inactivated in human tumors resulting in deregulated E2F activity. RESULTS Here, we report that lncRNA XLOC 006942, which we named ERIC, is regulated by E2F1 and, most probably, also E2F3. We show that expression levels of ERIC were elevated upon activation of exogenous E2F1, E2F3 or endogenous E2Fs. Moreover, knockdown of either E2F1 or E2F3 reduced ERIC levels and endogenous E2F1 binds ERIC's promoter. Expression of ERIC was cell cycle regulated and peaked in G1 in an E2F1-dependent manner. Inhibition of ERIC expression increased E2F1-mediated apoptosis, suggesting that E2F1 and ERIC constitute a negative feedback loop that modulates E2F1 activity. Furthermore, ERIC levels were increased following DNA damage by the chemotherapeutic drug Etoposide, and inhibition of ERIC expression enhanced Etoposide -induced apoptosis. CONCLUSIONS Our data identify ERIC as a novel lncRNA that is transcriptionally regulated by E2Fs, and restricts apoptosis induced by E2F1, as well as by DNA damage.
Collapse
Affiliation(s)
| | | | | | | | | | - Doron Ginsberg
- The Mina and Everard Goodman Faculty of Life Science, Bar Ilan University, Ramat Gan 52900, Israel.
| |
Collapse
|
557
|
Fujiya M, Konishi H, Mohamed Kamel MK, Ueno N, Inaba Y, Moriichi K, Tanabe H, Ikuta K, Ohtake T, Kohgo Y. microRNA-18a induces apoptosis in colon cancer cells via the autophagolysosomal degradation of oncogenic heterogeneous nuclear ribonucleoprotein A1. Oncogene 2013; 33:4847-56. [PMID: 24166503 DOI: 10.1038/onc.2013.429] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 08/12/2013] [Accepted: 08/26/2013] [Indexed: 12/16/2022]
Abstract
It is well known that microRNAs (miRs) are abnormally expressed in various cancers and target the messenger RNAs (mRNAs) of cancer-associated genes. While (miRs) are abnormally expressed in various cancers, whether miRs directly target oncogenic proteins is unknown. The present study investigated the inhibitory effects of miR-18a on colon cancer progression, which was considered to be mediated through its direct binding and degradation of heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1). An MTT assay and xenograft model demonstrated that the transfection of miR-18a induced apoptosis in SW620 cells. A binding assay revealed direct binding between miR-18a and hnRNP A1 in the cytoplasm of SW620 cells, which inhibited the oncogenic functions of hnRNP A1. A competitor RNA, which included the complementary sequence of the region of the miR-18a-hnRNP A1 binding site, repressed the effects of miR-18a on the induction of cancer cell apoptosis. In vitro single and in vivo double isotope assays demonstrated that miR-18a induced the degradation of hnRNP A1. An immunocytochemical study of hnRNP A1 and LC3-II and the inhibition of autophagy by 3-methyladenine and ATG7, p62 and BAG3 siRNA showed that miR-18a and hnRNP A1 formed a complex that was degraded through the autophagolysosomal pathway. This is the first report showing a novel function of a miR in the autophagolysosomal degradation of an oncogenic protein resulting from the creation of a complex consisting of the miR and a RNA-binding protein, which suppressed cancer progression.
Collapse
Affiliation(s)
- M Fujiya
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - H Konishi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - M K Mohamed Kamel
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - N Ueno
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Y Inaba
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - K Moriichi
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - H Tanabe
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - K Ikuta
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - T Ohtake
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| | - Y Kohgo
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical College, Asahikawa, Japan
| |
Collapse
|
558
|
MicroRNAs and long non-coding RNAs: prospects in diagnostics and therapy of cancer. Radiol Oncol 2013; 47:311-8. [PMID: 24294175 PMCID: PMC3814275 DOI: 10.2478/raon-2013-0062] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 08/20/2013] [Indexed: 12/11/2022] Open
Abstract
Background Non-coding RNAs (ncRNAs) are key regulatory molecules in cellular processes, and are potential biomarkers in many diseases. Currently, microRNAs and long non-coding RNAs are being pursued as diagnostic and prognostic biomarkers, and as therapeutic tools in cancer, since their expression profiling is able to distinguish different cancer types and classify their sub-types. Conclusions There are numerous studies confirming involvement of ncRNAs in cancer initiation, development and progression, but have only been recently identified as new diagnostic and prognostic tools. This can be beneficial in future medical cancer treatment options, since ncRNAs are natural antisense interactors included in regulation of many genes connected to survival and proliferation. Research is directed in development of useful markers for diagnosis and prognosis in cancer and in developing new RNA-based cancer therapies, of which some are already in clinical trials.
Collapse
|
559
|
Shu D, Khisamutdinov EF, Zhang L, Guo P. Programmable folding of fusion RNA in vivo and in vitro driven by pRNA 3WJ motif of phi29 DNA packaging motor. Nucleic Acids Res 2013; 42:e10. [PMID: 24084081 PMCID: PMC3902900 DOI: 10.1093/nar/gkt885] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Misfolding and associated loss of function are common problems in constructing fusion RNA complexes due to changes in energy landscape and the nearest-neighbor principle. Here we report the incorporation and application of the pRNA-3WJ motif of the phi29 DNA packaging motor into fusion RNA with controllable and predictable folding. The motif included three discontinuous ∼18 nucleotide (nt) fragments, displayed a distinct low folding energy (Shu D et al., Nature Nanotechnology, 2011, 6:658–667), and folded spontaneously into a leading core that enabled the correct folding of other functionalities fused to the RNA complex. Three individual fragments dispersed at any location within the sequence allowed the other RNA functional modules to fold into their original structures with authentic functions, as tested by Hepatitis B virus ribozyme, siRNA, and aptamers for malachite green (MG), spinach, and streptavidin (STV). Only nine complementary nucleotides were present for any two of the three ∼18-nt fragments, but the three 9 bp branches were so powerful that they disrupted other double strands with more than 15 bp within the fusion RNA. This system enabled the production of fusion complexes harboring multiple RNA functionalities with correct folding for potential applications in biotechnology, nanomedicine and nanotechnology. We also applied this system to investigate the principles governing the folding of RNA in vivo and in vitro. Temporal production of RNA sequences during in vivo transcription caused RNA to fold into different conformations that could not be predicted with routine principles derived from in vitro studies.
Collapse
Affiliation(s)
- Dan Shu
- Nanobiotechnology Center, Markey Cancer Center, and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | |
Collapse
|
560
|
Zhang H, Chen X, Wang C, Xu Z, Wang Y, Liu X, Kang Z, Ji W. Long non-coding genes implicated in response to stripe rust pathogen stress in wheat (Triticum aestivum L.). Mol Biol Rep 2013; 40:6245-53. [PMID: 24065539 DOI: 10.1007/s11033-013-2736-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 09/14/2013] [Indexed: 02/01/2023]
Abstract
The non-protein-coding genes have been reported as a critical control role in the regulation of gene expression in abiotic stress. We previously identified four expressed sequence tags numbered S18 (EL773024), S73 (EL773035), S106 (EL773041) and S108 (EL773042) from a SSH-cDNA library of bread wheat Shaanmai 139 infected with Puccinia striiformis f. sp. tritici (Pst). Here, we isolated four cDNA clones and referred them as TalncRNA18, TalncRNA73, TalncRNA106 and TalncRNA108 (GenBank: KC549675-KC549678). These cDNA separately consisted of 1,393, 667, 449 and 647 nucleotides but without any open reading frame. The alignment result showed that TalncRNA18 is a partial cDNA of E3 ubiquitin-protein ligase UPL1-like gene, TalncRNA73 is an antisense transcript of hypothetical protein, TalncRNA108 is a homolog to RRNA intron-encoded homing endonuclease, and lncRNA106 had no similarly sequence. Quantitative RT-PCR studies confirmed that these four lncRNAs were differentially expressed in three near isogenic lines. TalncRNA108 was significantly stepwise decreased at early stage of inoculation with Pst, while the others were upregulated, especially at 1 and 3 dpi (days post-inoculation). Using Chinese Spring nulli-tetrasomic lines and its ditelosomic lines, TalncRNA73 and TalncRNA108 were located to wheat chromosome 7A and the short arm of chromosome 4B, respectively, while TalncRNA18 and TalncRNA106 were located to chromosome 5B. Comparing the sequence of DNA and cDNA of four lncRNAs with polymerase chain reaction primers, the results showed that all of them have no introns. The kinetics analyses of lncRNAs expression as a result of pathogen challenge in immune resistant genotype indicated that they may play the roles of modulating or silencing the protein-coding gene into pathogen-defence response.
Collapse
Affiliation(s)
- Hong Zhang
- State Key Laboratory of Crop Stress Biology for Arid Area, College of Agronomy, Northwest A&F University, Yangling, 712100, Shaanxi, China,
| | | | | | | | | | | | | | | |
Collapse
|
561
|
Murata A, Harada Y, Fukuzumi T, Nakatani K. Fluorescent indicator displacement assay of ligands targeting 10 microRNA precursors. Bioorg Med Chem 2013; 21:7101-6. [PMID: 24084297 DOI: 10.1016/j.bmc.2013.09.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/04/2013] [Accepted: 09/04/2013] [Indexed: 11/15/2022]
Abstract
Fluorescent indicator displacement (FID) assay is a rapid and convenient assay for identifying new ligands that bind to the target molecules. In our previous studies, we have shown that a series of 2,7-diaminoalkoxy xanthone and thioxanthone derivatives can be used as fluorescent indicators for detecting the interaction between RNA and a ligand. The xanthone and thioxanthone fluorochromes showed efficient fluorescence quenching upon binding to target RNA. Subsequent displacement of the bound-fluorochrome with a ligand that binds more strongly to the target RNA led to the recovery of the fluorescence by releasing the fluorochrome from RNA. Here we report a pilot screening of a chemical library that contains 9600 structurally diverse compounds for molecules that bind to a specific miRNA precursor using the FID assay.
Collapse
Affiliation(s)
- Asako Murata
- Department of Regulatory Bioorganic Chemistry, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki 567-0047, Japan
| | | | | | | |
Collapse
|
562
|
Lu X, Fang Y, Wang Z, Xie J, Zhan Q, Deng X, Chen H, Jin J, Peng C, Li H, Shen B. Downregulation of gas5 increases pancreatic cancer cell proliferation by regulating CDK6. Cell Tissue Res 2013; 354:891-6. [PMID: 24026436 DOI: 10.1007/s00441-013-1711-x] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/19/2013] [Indexed: 01/03/2023]
Abstract
Recent studies have revealed that long non-coding RNAs (lncRNAs) play important roles in cancer biology and that lncRNA gas5 (growth arrest-specific 5) regulates breast cancer cell growth. However, the role of gas5 in pancreatic cancer progression remains largely unknown. In the current study, we assay the expression level of gas5 in pancreatic cancer tissues and define the role of gas5 in the regulation of pancreatic cancer cell proliferation. We verify that the expression level of gas5 is significantly decreased in pancreatic cancer tissues compared with normal control. Overexpression of gas5 in pancreatic cancer cells inhibits cell proliferation, whereas gas5 inhibition induces a significant decrease in G0/G1 phase and an increase in S phase. We further demonstrate that gas5 negatively regulates CDK6 (cyclin-dependent kinase 6) expression in vitro and in vivo. More importantly, knockdown of CDK6 partially abrogates gas5-siRNA-induced cell proliferation. These data suggest an important role of gas5 in the molecular etiology of pancreatic cancer and implicate the potential application of gas5 in pancreatic cancer therapy.
Collapse
Affiliation(s)
- Xiongxiong Lu
- Department of General Surgery, RuiJin Hospital, Shanghai Jiaotong University School of Medicine, 197 RuiJin.Er.Road, ShangHai, 200025, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
563
|
Ronchetti D, Mosca L, Cutrona G, Tuana G, Gentile M, Fabris S, Agnelli L, Ciceri G, Matis S, Massucco C, Colombo M, Reverberi D, Recchia AG, Bossio S, Negrini M, Tassone P, Morabito F, Ferrarini M, Neri A. Small nucleolar RNAs as new biomarkers in chronic lymphocytic leukemia. BMC Med Genomics 2013; 6:27. [PMID: 24004562 PMCID: PMC3766210 DOI: 10.1186/1755-8794-6-27] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 09/02/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Small nucleolar RNAs (snoRNAs) and small Cajal body-specific RNAs are non-coding RNAs involved in the maturation of other RNA molecules. Alterations of sno/scaRNA expression may play a role in cancerogenesis. This study elucidates the patterns of sno/scaRNA expression in 211 chronic lymphocytic leukemia (CLL) patients (Binet stage A) also in comparison with those of different normal B-cell subsets. METHODS The patterns of sno/scaRNA expression in highly purified CD19+ B-cells of 211 CLL patients and in 18 normal B-cell samples--6 from peripheral blood, and 12 from tonsils (4 germinal center, 2 marginal zone, 3 switched memory and 3 naïve B-cells)--were analyzed on the Affymetrix GeneChip® Human Gene 1.0 ST array. RESULTS CLLs display a sno/scaRNAs expression profile similar to normal memory, naïve and marginal-zone B-cells, with the exception of a few down-regulated transcripts (SNORA31, -6, -62, and -71C). Our analyses also suggest some heterogeneity in the pattern of sno/scaRNAs expression which is apparently unrelated to the major biological (ZAP-70 and CD38), molecular (IGHV mutation) and cytogenetic markers. Moreover, we found that SNORA70F was significantly down-regulated in poor prognostic subgroups and this phenomenon was associated with the down-regulation of its host gene COBLL1. Finally, we generated an independent model based on SNORA74A and SNORD116-18 expression, which appears to distinguish two different prognostic CLL groups. CONCLUSIONS These data extend the view of sno/scaRNAs deregulation in cancer and may contribute to discover novel biomarkers associated with the disease and potentially useful to predict the clinical outcome of early stage CLL patients.
Collapse
MESH Headings
- B-Lymphocyte Subsets/metabolism
- Biomarkers, Tumor/genetics
- Cell Nucleus/genetics
- Gene Expression Profiling
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Neoplasm Staging
- Prognosis
- RNA, Small Nucleolar/genetics
- Risk
- Transcription, Genetic
Collapse
Affiliation(s)
- Domenica Ronchetti
- Department of Clinical Sciences and Community Health, University of Milano, Milano, Italy
| | - Laura Mosca
- Department of Clinical Sciences and Community Health, University of Milano, Milano, Italy
| | | | - Giacomo Tuana
- Department of Clinical Sciences and Community Health, University of Milan, Hematology 1 CTMO, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, F. Sforza, 35-20122, Milano, Italy
| | - Massimo Gentile
- U.O.C. di Ematologia, Azienda Ospedaliera di Cosenza, Cosenza, Italy
| | - Sonia Fabris
- Department of Clinical Sciences and Community Health, University of Milano, Milano, Italy
| | - Luca Agnelli
- Department of Clinical Sciences and Community Health, University of Milano, Milano, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Hematology 1 CTMO, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, F. Sforza, 35-20122, Milano, Italy
| | - Gabriella Ciceri
- Department of Clinical Sciences and Community Health, University of Milano, Milano, Italy
| | - Serena Matis
- Scientific Direction, IRCCS S Martino-IST, Genova, Italy
| | | | - Monica Colombo
- Scientific Direction, IRCCS S Martino-IST, Genova, Italy
| | | | | | - Sabrina Bossio
- U.O.C. di Ematologia, Azienda Ospedaliera di Cosenza, Cosenza, Italy
| | - Massimo Negrini
- Department of Experimental and Clinical Medicine, University of Ferrara, Ferrara, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | | | | | - Antonino Neri
- Department of Clinical Sciences and Community Health, University of Milano, Milano, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Hematology 1 CTMO, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, F. Sforza, 35-20122, Milano, Italy
| |
Collapse
|
564
|
Chen X, Yan GY. Novel human lncRNA-disease association inference based on lncRNA expression profiles. ACTA ACUST UNITED AC 2013; 29:2617-24. [PMID: 24002109 DOI: 10.1093/bioinformatics/btt426] [Citation(s) in RCA: 446] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
MOTIVATION More and more evidences have indicated that long-non-coding RNAs (lncRNAs) play critical roles in many important biological processes. Therefore, mutations and dysregulations of these lncRNAs would contribute to the development of various complex diseases. Developing powerful computational models for potential disease-related lncRNAs identification would benefit biomarker identification and drug discovery for human disease diagnosis, treatment, prognosis and prevention. RESULTS In this article, we proposed the assumption that similar diseases tend to be associated with functionally similar lncRNAs. Then, we further developed the method of Laplacian Regularized Least Squares for LncRNA-Disease Association (LRLSLDA) in the semisupervised learning framework. Although known disease-lncRNA associations in the database are rare, LRLSLDA still obtained an AUC of 0.7760 in the leave-one-out cross validation, significantly improving the performance of previous methods. We also illustrated the performance of LRLSLDA is not sensitive (even robust) to the parameters selection and it can obtain a reliable performance in all the test classes. Plenty of potential disease-lncRNA associations were publicly released and some of them have been confirmed by recent results in biological experiments. It is anticipated that LRLSLDA could be an effective and important biological tool for biomedical research. AVAILABILITY The code of LRLSLDA is freely available at http://asdcd.amss.ac.cn/Software/Details/2.
Collapse
Affiliation(s)
- Xing Chen
- National Center for Mathematics and Interdisciplinary Sciences and Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | | |
Collapse
|
565
|
Beckedorff FC, Ayupe AC, Crocci-Souza R, Amaral MS, Nakaya HI, Soltys DT, Menck CFM, Reis EM, Verjovski-Almeida S. The intronic long noncoding RNA ANRASSF1 recruits PRC2 to the RASSF1A promoter, reducing the expression of RASSF1A and increasing cell proliferation. PLoS Genet 2013; 9:e1003705. [PMID: 23990798 PMCID: PMC3749938 DOI: 10.1371/journal.pgen.1003705] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 06/24/2013] [Indexed: 01/01/2023] Open
Abstract
The down-regulation of the tumor-suppressor gene RASSF1A has been shown to increase cell proliferation in several tumors. RASSF1A expression is regulated through epigenetic events involving the polycomb repressive complex 2 (PRC2); however, the molecular mechanisms modulating the recruitment of this epigenetic modifier to the RASSF1 locus remain largely unknown. Here, we identify and characterize ANRASSF1, an endogenous unspliced long noncoding RNA (lncRNA) that is transcribed from the opposite strand on the RASSF1 gene locus in several cell lines and tissues and binds PRC2. ANRASSF1 is transcribed through RNA polymerase II and is 5′-capped and polyadenylated; it exhibits nuclear localization and has a shorter half-life compared with other lncRNAs that bind PRC2. ANRASSF1 endogenous expression is higher in breast and prostate tumor cell lines compared with non-tumor, and an opposite pattern is observed for RASSF1A. ANRASSF1 ectopic overexpression reduces RASSF1A abundance and increases the proliferation of HeLa cells, whereas ANRASSF1 silencing causes the opposite effects. These changes in ANRASSF1 levels do not affect the RASSF1C isoform abundance. ANRASSF1 overexpression causes a marked increase in both PRC2 occupancy and histone H3K27me3 repressive marks, specifically at the RASSF1A promoter region. No effect of ANRASSF1 overexpression was detected on PRC2 occupancy and histone H3K27me3 at the promoter regions of RASSF1C and the four other neighboring genes, including two well-characterized tumor suppressor genes. Additionally, we demonstrated that ANRASSF1 forms an RNA/DNA hybrid and recruits PRC2 to the RASSF1A promoter. Together, these results demonstrate a novel mechanism of epigenetic repression of the RASSF1A tumor suppressor gene involving antisense unspliced lncRNA, in which ANRASSF1 selectively represses the expression of the RASSF1 isoform overlapping the antisense transcript in a location-specific manner. In a broader perspective, our findings suggest that other non-characterized unspliced intronic lncRNAs transcribed in the human genome might contribute to a location-specific epigenetic modulation of genes. RASSF1A is a tumor suppressor gene whose expression is repressed through epigenetic events in a wide range of different cancers. Repression is effected by DNA hypermethylation of the RASSF1A promoter, which in turn is triggered through histone H3K9/H3K27 trimethylation repressive marks. The addition of the H3K27me3 mark at the RASSF1A promoter locus involves the polycomb repressive complex 2 (PRC2). The molecular mechanisms that control the recruitment of PRC2 to the promoter to initiate H3K27 trimethylation and repress RASSF1A expression have not been described. Here, we identified a long noncoding RNA (lncRNA), termed ANRASSF1 for antisense noncoding RASSF1, that is transcribed from the opposite strand of the RASSF1A gene and is responsible for recruiting PRC2 to the RASSF1A promoter region in a highly location-specific manner. No effect of ANRASSF1 was detected on the promoter of the RASSF1C isoform or the promoters of the four other genes within the vicinity of RASSF1, including two other well-characterized tumor suppressor genes. This work provides evidence that the epigenetic modulation of the tumor suppressor gene RASSF1A is dependent on the lncRNA ANRASSF1 and highlights the importance of further studies on the involvement of ANRASSF1 in tumorigenesis.
Collapse
Affiliation(s)
- Felipe C. Beckedorff
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Ana C. Ayupe
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Renan Crocci-Souza
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Murilo S. Amaral
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Helder I. Nakaya
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Daniela T. Soltys
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Carlos F. M. Menck
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Eduardo M. Reis
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brasil
- Instituto Nacional de Ciência e Tecnologia em Oncogenômica, São Paulo, São Paulo, Brasil
| | - Sergio Verjovski-Almeida
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brasil
- Instituto Nacional de Ciência e Tecnologia em Oncogenômica, São Paulo, São Paulo, Brasil
- * E-mail:
| |
Collapse
|
566
|
|
567
|
Mustafi D, Kevany BM, Bai X, Maeda T, Sears JE, Khalil AM, Palczewski K. Evolutionarily conserved long intergenic non-coding RNAs in the eye. Hum Mol Genet 2013; 22:2992-3002. [PMID: 23562822 PMCID: PMC3699063 DOI: 10.1093/hmg/ddt156] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 04/02/2013] [Indexed: 02/01/2023] Open
Abstract
The discovery that the mammalian transcriptome encodes thousands of long intergenic non-coding (linc) RNA transcripts, together with recent evidence that lincRNAs can regulate protein-coding genes, has added a new level of complexity to cellular transcriptional/translational regulation. Indeed several reports now link mutations in lincRNAs to heritable human disorders. Here, we identified a subset of lincRNAs in terminally differentiated adult human retinal neurons based on their sequence conservation across species. RNA sequencing of eye tissue from several mammalian species with varied rod/cone photoreceptor content identified 18 lincRNAs that were highly conserved across these species. Sixteen of the 18 were conserved in human retinal tissue with 14 of these also conserved in the macular region. A subset of lincRNAs exhibited restricted tissue expression profiles in mice, with preferential expression in the retina. Mouse models with different populations of retinal cells as well as in situ hybridization provided evidence that these lincRNAs localized to specific retinal compartments, most notably to the photoreceptor neuronal layer. Computational genomic loci and promoter region analyses provided a basis for regulated expression of these conserved lincRNAs in retinal post-mitotic neurons. This combined approach identified several lincRNAs that could be critical for retinal and visual maintenance in adults.
Collapse
Affiliation(s)
| | | | | | - Tadao Maeda
- Department of Ophthalmology and Visual Sciences and
| | - Jonathan E. Sears
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44106-4965, USA and
| | - Ahmad M. Khalil
- Center for RNA Molecular Biology
- Department of Genetics and Genome Sciences, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106-4965, USA
| | | |
Collapse
|
568
|
Cruickshanks HA, Vafadar-Isfahani N, Dunican DS, Lee A, Sproul D, Lund JN, Meehan RR, Tufarelli C. Expression of a large LINE-1-driven antisense RNA is linked to epigenetic silencing of the metastasis suppressor gene TFPI-2 in cancer. Nucleic Acids Res 2013; 41:6857-69. [PMID: 23703216 PMCID: PMC3737543 DOI: 10.1093/nar/gkt438] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 04/29/2013] [Accepted: 04/29/2013] [Indexed: 12/18/2022] Open
Abstract
LINE-1 retrotransposons are abundant repetitive elements of viral origin, which in normal cells are kept quiescent through epigenetic mechanisms. Activation of LINE-1 occurs frequently in cancer and can enable LINE-1 mobilization but also has retrotransposition-independent consequences. We previously reported that in cancer, aberrantly active LINE-1 promoters can drive transcription of flanking unique sequences giving rise to LINE-1 chimeric transcripts (LCTs). Here, we show that one such LCT, LCT13, is a large transcript (>300 kb) running antisense to the metastasis-suppressor gene TFPI-2. We have modelled antisense RNA expression at TFPI-2 in transgenic mouse embryonic stem (ES) cells and demonstrate that antisense RNA induces silencing and deposition of repressive histone modifications implying a causal link. Consistent with this, LCT13 expression in breast and colon cancer cell lines is associated with silencing and repressive chromatin at TFPI-2. Furthermore, we detected LCT13 transcripts in 56% of colorectal tumours exhibiting reduced TFPI-2 expression. Our findings implicate activation of LINE-1 elements in subsequent epigenetic remodelling of surrounding genes, thus hinting a novel retrotransposition-independent role for LINE-1 elements in malignancy.
Collapse
Affiliation(s)
- Hazel A. Cruickshanks
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, University of Nottingham, Centre for Biomedical Sciences, Nottingham NG7 2RD, UK, School of Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK, Breakthrough Research Unit, University of Edinburgh, Edinburgh EH4 2XU, UK and Centre for Genetics and Genomics, University of Nottingham, Queens Medical Centre, Nottingham NG7 2RD, UK
| | - Natasha Vafadar-Isfahani
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, University of Nottingham, Centre for Biomedical Sciences, Nottingham NG7 2RD, UK, School of Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK, Breakthrough Research Unit, University of Edinburgh, Edinburgh EH4 2XU, UK and Centre for Genetics and Genomics, University of Nottingham, Queens Medical Centre, Nottingham NG7 2RD, UK
| | - Donncha S. Dunican
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, University of Nottingham, Centre for Biomedical Sciences, Nottingham NG7 2RD, UK, School of Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK, Breakthrough Research Unit, University of Edinburgh, Edinburgh EH4 2XU, UK and Centre for Genetics and Genomics, University of Nottingham, Queens Medical Centre, Nottingham NG7 2RD, UK
| | - Andy Lee
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, University of Nottingham, Centre for Biomedical Sciences, Nottingham NG7 2RD, UK, School of Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK, Breakthrough Research Unit, University of Edinburgh, Edinburgh EH4 2XU, UK and Centre for Genetics and Genomics, University of Nottingham, Queens Medical Centre, Nottingham NG7 2RD, UK
| | - Duncan Sproul
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, University of Nottingham, Centre for Biomedical Sciences, Nottingham NG7 2RD, UK, School of Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK, Breakthrough Research Unit, University of Edinburgh, Edinburgh EH4 2XU, UK and Centre for Genetics and Genomics, University of Nottingham, Queens Medical Centre, Nottingham NG7 2RD, UK
| | - Jonathan N. Lund
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, University of Nottingham, Centre for Biomedical Sciences, Nottingham NG7 2RD, UK, School of Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK, Breakthrough Research Unit, University of Edinburgh, Edinburgh EH4 2XU, UK and Centre for Genetics and Genomics, University of Nottingham, Queens Medical Centre, Nottingham NG7 2RD, UK
| | - Richard R. Meehan
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, University of Nottingham, Centre for Biomedical Sciences, Nottingham NG7 2RD, UK, School of Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK, Breakthrough Research Unit, University of Edinburgh, Edinburgh EH4 2XU, UK and Centre for Genetics and Genomics, University of Nottingham, Queens Medical Centre, Nottingham NG7 2RD, UK
| | - Cristina Tufarelli
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, University of Nottingham, Centre for Biomedical Sciences, Nottingham NG7 2RD, UK, School of Graduate Entry Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK, MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK, Breakthrough Research Unit, University of Edinburgh, Edinburgh EH4 2XU, UK and Centre for Genetics and Genomics, University of Nottingham, Queens Medical Centre, Nottingham NG7 2RD, UK
| |
Collapse
|
569
|
McHale CM, Zhang L, Thomas R, Smith MT. Analysis of the transcriptome in molecular epidemiology studies. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2013; 54:500-517. [PMID: 23907930 PMCID: PMC5142298 DOI: 10.1002/em.21798] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 06/07/2013] [Accepted: 06/08/2013] [Indexed: 05/29/2023]
Abstract
The human transcriptome is complex, comprising multiple transcript types, mostly in the form of non-coding RNA (ncRNA). The majority of ncRNA is of the long form (lncRNA, ≥ 200 bp), which plays an important role in gene regulation through multiple mechanisms including epigenetics, chromatin modification, control of transcription factor binding, and regulation of alternative splicing. Both mRNA and ncRNA exhibit additional variability in the form of alternative splicing and RNA editing. All aspects of the human transcriptome can potentially be dysregulated by environmental exposures. Next-generation RNA sequencing (RNA-Seq) is the best available methodology to measure this although it has limitations, including experimental bias. The third phase of the MicroArray Quality Control Consortium project (MAQC-III), also called Sequencing Quality Control (SeQC), aims to address these limitations through standardization of experimental and bioinformatic methodologies. A limited number of toxicogenomic studies have been conducted to date using RNA-Seq. This review describes the complexity of the human transcriptome, the application of transcriptomics by RNA-Seq or microarray in molecular epidemiology studies, and limitations of these approaches including the type of cell or tissue analyzed, experimental variation, and confounding. By using good study designs with precise, individual exposure measurements, sufficient power and incorporation of phenotypic anchors, studies in human populations can identify biomarkers of exposure and/or early effect and elucidate mechanisms of action underlying associated diseases, even at low doses. Analysis of datasets at the pathway level can compensate for some of the limitations of RNA-Seq and, as more datasets become available, will increasingly elucidate the exposure-disease continuum.
Collapse
Affiliation(s)
- Cliona M McHale
- Division of Environmental Health Sciences, Genes and Environment Laboratory, School of Public Health, University of California, Berkeley, California 94720, USA.
| | | | | | | |
Collapse
|
570
|
Shu Y, Shu D, Haque F, Guo P. Fabrication of pRNA nanoparticles to deliver therapeutic RNAs and bioactive compounds into tumor cells. Nat Protoc 2013; 8:1635-59. [PMID: 23928498 DOI: 10.1038/nprot.2013.097] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RNA nanotechnology is a term that refers to the design, fabrication and use of nanoparticles that are mainly composed of RNAs via bottom-up self-assembly. The packaging RNA (pRNA) of the bacteriophage phi29 DNA packaging motor has been developed into a nanodelivery platform. This protocol describes the synthesis, assembly and functionalization of pRNA nanoparticles on the basis of three 'toolkits' derived from pRNA structural features: interlocking loops for hand-in-hand interactions, palindrome sequences for foot-to-foot interactions and an RNA three-way junction for branch extension. siRNAs, ribozymes, aptamers, chemical ligands, fluorophores and other functionalities can also be fused to the pRNA before the assembly of the nanoparticles, so as to ensure the production of homogeneous nanoparticles and the retention of appropriate folding and function of the incorporated modules. The resulting self-assembled multivalent pRNA nanoparticles are thermodynamically and chemically stable, and they remain intact at ultralow concentrations. Gene-silencing effects are progressively enhanced with increasing numbers of siRNAs in each pRNA nanoparticle. Systemic injection of the pRNA nanoparticles into xenograft-bearing mice has revealed strong binding to tumors without accumulation in vital organs or tissues. The pRNA-based nanodelivery scaffold paves a new way for nanotechnological application of pRNA-based nanoparticles for disease detection and treatment. The time required for completing one round of this protocol is 3-4 weeks when including in vitro functional assays, or 2-3 months when including in vivo studies.
Collapse
Affiliation(s)
- Yi Shu
- Nanobiotechnology Center, Markey Cancer Center, Lexington, Kentucky, USA
| | | | | | | |
Collapse
|
571
|
Garitano-Trojaola A, Agirre X, Prósper F, Fortes P. Long non-coding RNAs in haematological malignancies. Int J Mol Sci 2013; 14:15386-422. [PMID: 23887658 PMCID: PMC3759866 DOI: 10.3390/ijms140815386] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 06/28/2013] [Accepted: 07/09/2013] [Indexed: 12/20/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are functional RNAs longer than 200 nucleotides in length. LncRNAs are as diverse as mRNAs and they normally share the same biosynthetic machinery based on RNA polymerase II, splicing and polyadenylation. However, lncRNAs have low coding potential. Compared to mRNAs, lncRNAs are preferentially nuclear, more tissue specific and expressed at lower levels. Most of the lncRNAs described to date modulate the expression of specific genes by guiding chromatin remodelling factors; inducing chromosomal loopings; affecting transcription, splicing, translation or mRNA stability; or serving as scaffolds for the organization of cellular structures. They can function in cis, cotranscriptionally, or in trans, acting as decoys, scaffolds or guides. These functions seem essential to allow cell differentiation and growth. In fact, many lncRNAs have been shown to exert oncogenic or tumor suppressor properties in several cancers including haematological malignancies. In this review, we summarize what is known about lncRNAs, the mechanisms for their regulation in cancer and their role in leukemogenesis, lymphomagenesis and hematopoiesis. Furthermore, we discuss the potential of lncRNAs in diagnosis, prognosis and therapy in cancer, with special attention to haematological malignancies.
Collapse
Affiliation(s)
- Andoni Garitano-Trojaola
- Laboratory of Myeloproliferative Syndromes, Oncology Area, Foundation for Applied Medical Research, University of Navarra, Pamplona 31008, Spain; E-Mails: (A.G.-T.); (X.A.); (F.P.)
| | - Xabier Agirre
- Laboratory of Myeloproliferative Syndromes, Oncology Area, Foundation for Applied Medical Research, University of Navarra, Pamplona 31008, Spain; E-Mails: (A.G.-T.); (X.A.); (F.P.)
| | - Felipe Prósper
- Laboratory of Myeloproliferative Syndromes, Oncology Area, Foundation for Applied Medical Research, University of Navarra, Pamplona 31008, Spain; E-Mails: (A.G.-T.); (X.A.); (F.P.)
- Hematology Service and Area of Cell Therapy, University of Navarra Clinic, University of Navarra, Pamplona 31008, Spain
| | - Puri Fortes
- Department of Hepatology and Gene Therapy, Foundation for Applied Medical Research, University of Navarra, Pamplona 31008, Spain
| |
Collapse
|
572
|
Intercellular communication by exosome-derived microRNAs in cancer. Int J Mol Sci 2013; 14:14240-69. [PMID: 23839094 PMCID: PMC3742242 DOI: 10.3390/ijms140714240] [Citation(s) in RCA: 399] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 02/08/2023] Open
Abstract
The development of human cancers is a multistep process in which normal cells acquire characteristics that ultimately lead to their conversion into cancer cells. Many obstacles must be overcome for this process to occur; of these obstacles, is the ability to survive an inhospitable microenvironment. It is recognized that the intercommunication between tumor cells and their surrounding microenvironment is essential to overcoming this obstacle and for the tumor to progress, metastasize and establish itself at distant sites. Exosomes are membrane-derived vesicles that have recently been recognized as important mediators of intercellular communication, as they carry lipids, proteins, mRNAs and microRNAs that can be transferred to a recipient cell via fusion of the exosome with the target cell membrane. In the context of cancer cells, this process entails the transfer of cancer-promoting cellular contents to surrounding cells within the tumor microenvironment or into the circulation to act at distant sites, thereby enabling cancer progression. In this process, the transfer of exosomal microRNAs to a recipient cell where they can regulate target gene expression is of particular interest, both in understanding the basic biology of cancer progression and for the development of therapeutic approaches. This review discusses the exosome-mediated intercellular communication via microRNAs within the tumor microenvironment in human cancers, with a particular focus on breast cancer exosomes.
Collapse
|
573
|
Dave RK, Dinger ME, Andrew M, Askarian-Amiri M, Hume DA, Kellie S. Regulated expression of PTPRJ/CD148 and an antisense long noncoding RNA in macrophages by proinflammatory stimuli. PLoS One 2013; 8:e68306. [PMID: 23840844 PMCID: PMC3695918 DOI: 10.1371/journal.pone.0068306] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 05/28/2013] [Indexed: 12/28/2022] Open
Abstract
PTPRJ/CD148 is a tyrosine phosphatase that has tumour suppressor-like activity. Quantitative PCR of various cells and tissues revealed that it is preferentially expressed in macrophage-enriched tissues. Within lymphoid tissues immunohistochemistry revealed that PTPRJ/CD148 co-localised with F4/80, indicating that macrophages most strongly express the protein. Macrophages express the highest basal level of ptprj, and this is elevated further by treatment with LPS and other Toll-like receptor ligands. In contrast, CSF-1 treatment reduced basal and stimulated Ptprj expression in human and mouse cells, and interferon also repressed Ptprj expression. We identified a 1006 nucleotide long noncoding RNA species, Ptprj-as1 that is transcribed antisense to Ptprj. Ptprj-as1 was highly expressed in macrophage-enriched tissue and was transiently induced by Toll-like receptor ligands with a similar time course to Ptprj. Finally, putative transcription factor binding sites in the promoter region of Ptprj were identified.
Collapse
Affiliation(s)
- Richa K. Dave
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Australia
- The University of Queensland, Cooperative Research Centre for Chronic Inflammatory Diseases (CRC-CID), Brisbane, Australia
- The University of Queensland, Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, Australia
| | - Marcel E. Dinger
- The University of Queensland Diamantina Institute, Brisbane, Australia
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Megan Andrew
- The University of Queensland, Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, Australia
| | - Marjan Askarian-Amiri
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Australia
| | - David A. Hume
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Australia
- The University of Queensland, Cooperative Research Centre for Chronic Inflammatory Diseases (CRC-CID), Brisbane, Australia
- The Roslin Institute, University of Edinburgh, Roslin, Scotland, United Kingdom
| | - Stuart Kellie
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Australia
- The University of Queensland, Cooperative Research Centre for Chronic Inflammatory Diseases (CRC-CID), Brisbane, Australia
- The University of Queensland, Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, Australia
| |
Collapse
|
574
|
Zhang Z, Zhao Z, Liu B, Li D, Zhang D, Chen H, Liu D. Systems biomedicine: It’s your turn—Recent progress in systems biomedicine. QUANTITATIVE BIOLOGY 2013. [DOI: 10.1007/s40484-013-0009-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
575
|
Abstract
Long non-coding RNAs (lncRNAs) have been found to perform various functions in a wide variety of important biological processes. To make easier interpretation of lncRNA functionality and conduct deep mining on these transcribed sequences, it is convenient to classify lncRNAs into different groups. Here, we summarize classification methods of lncRNAs according to their four major features, namely, genomic location and context, effect exerted on DNA sequences, mechanism of functioning and their targeting mechanism. In combination with the presently available function annotations, we explore potential relationships between different classification categories, and generalize and compare biological features of different lncRNAs within each category. Finally, we present our view on potential further studies. We believe that the classifications of lncRNAs as indicated above are of fundamental importance for lncRNA studies, helpful for further investigation of specific lncRNAs, for formulation of new hypothesis based on different features of lncRNA and for exploration of the underlying lncRNA functional mechanisms.
Collapse
Affiliation(s)
- Lina Ma
- CAS Key Laboratory of Genome Sciences and Information; Beijing Institute of Genomics; Chinese Academy of Sciences; Beijing, China
| | - Vladimir B. Bajic
- King Abdullah University of Science and Technology (KAUST); Computational Bioscience Research Center; Computer, Electrical and Mathematical Sciences and Engineering Division; Thuwal, Kingdom of Saudi Arabia
| | - Zhang Zhang
- CAS Key Laboratory of Genome Sciences and Information; Beijing Institute of Genomics; Chinese Academy of Sciences; Beijing, China
- Correspondence to: Zhang Zhang,
| |
Collapse
|
576
|
Whiteside EJ, Seim I, Pauli JP, O'Keeffe AJ, Thomas PB, Carter SL, Walpole CM, Fung JNT, Josh P, Herington AC, Chopin LK. Identification of a long non-coding RNA gene, growth hormone secretagogue receptor opposite strand, which stimulates cell migration in non-small cell lung cancer cell lines. Int J Oncol 2013; 43:566-74. [PMID: 23722988 DOI: 10.3892/ijo.2013.1969] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 03/07/2013] [Indexed: 11/06/2022] Open
Abstract
The molecular mechanisms involved in non‑small cell lung cancer tumourigenesis are largely unknown; however, recent studies have suggested that long non-coding RNAs (lncRNAs) are likely to play a role. In this study, we used public databases to identify an mRNA-like, candidate long non-coding RNA, GHSROS (GHSR opposite strand), transcribed from the antisense strand of the ghrelin receptor gene, growth hormone secretagogue receptor (GHSR). Quantitative real-time RT-PCR revealed higher expression of GHSROS in lung cancer tissue compared to adjacent, non-tumour lung tissue. In common with many long non-coding RNAs, GHSROS is 5' capped and 3' polyadenylated (mRNA-like), lacks an extensive open reading frame and harbours a transposable element. Engineered overexpression of GHSROS stimulated cell migration in the A549 and NCI-H1299 non-small cell lung cancer cell lines, but suppressed cell migration in the Beas-2B normal lung-derived bronchoepithelial cell line. This suggests that GHSROS function may be dependent on the oncogenic context. The identification of GHSROS, which is expressed in lung cancer and stimulates cell migration in lung cancer cell lines, contributes to the growing number of non-coding RNAs that play a role in the regulation of tumourigenesis and metastatic cancer progression.
Collapse
Affiliation(s)
- Eliza J Whiteside
- Ghrelin Research Group, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
577
|
Kornienko AE, Guenzl PM, Barlow DP, Pauler FM. Gene regulation by the act of long non-coding RNA transcription. BMC Biol 2013; 11:59. [PMID: 23721193 PMCID: PMC3668284 DOI: 10.1186/1741-7007-11-59] [Citation(s) in RCA: 561] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 05/15/2013] [Indexed: 12/20/2022] Open
Abstract
Long non-protein-coding RNAs (lncRNAs) are proposed to be the largest transcript class in the mouse and human transcriptomes. Two important questions are whether all lncRNAs are functional and how they could exert a function. Several lncRNAs have been shown to function through their product, but this is not the only possible mode of action. In this review we focus on a role for the process of lncRNA transcription, independent of the lncRNA product, in regulating protein-coding-gene activity in cis. We discuss examples where lncRNA transcription leads to gene silencing or activation, and describe strategies to determine if the lncRNA product or its transcription causes the regulatory effect.
Collapse
Affiliation(s)
- Aleksandra E Kornienko
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25,3, 1090, Vienna, Austria
| | | | | | | |
Collapse
|
578
|
Jørgensen MG, Thomason MK, Havelund J, Valentin-Hansen P, Storz G. Dual function of the McaS small RNA in controlling biofilm formation. Genes Dev 2013; 27:1132-45. [PMID: 23666921 DOI: 10.1101/gad.214734.113] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Many bacterial small RNAs (sRNAs) regulate gene expression through base-pairing with mRNAs, and it has been assumed that these sRNAs act solely by this one mechanism. Here we report that the multicellular adhesive (McaS) sRNA of Escherichia coli uniquely acts by two different mechanisms: base-pairing and protein titration. Previous work established that McaS base pairs with the mRNAs encoding master transcription regulators of curli and flagella synthesis, respectively, resulting in down-regulation and up-regulation of these important cell surface structures. In this study, we demonstrate that McaS activates synthesis of the exopolysaccharide β-1,6 N-acetyl-D-glucosamine (PGA) by binding the global RNA-binding protein CsrA, a negative regulator of pgaA translation. The McaS RNA bears at least two CsrA-binding sequences, and inactivation of these sites compromises CsrA binding, PGA regulation, and biofilm formation. Moreover, ectopic McaS expression leads to induction of two additional CsrA-repressed genes encoding diguanylate cyclases. Collectively, our study shows that McaS is a dual-function sRNA with roles in the two major post-transcriptional regulons controlled by the RNA-binding proteins Hfq and CsrA.
Collapse
Affiliation(s)
- Mikkel Girke Jørgensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | | | | | | | | |
Collapse
|
579
|
Li X, Wu Z, Fu X, Han W. Long Noncoding RNAs: Insights from Biological Features and Functions to Diseases. Med Res Rev 2013; 33:517-53. [PMID: 22318902 DOI: 10.1002/med.21254] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the past decade, genome-wide transcriptomic studies have shown that the mammalian genome is pervasively transcribed and produces many thousands of transcriptomes without bias from previous genome annotations. This finding, together with the discovery of a plethora of unexpected RNAs that have no obvious coding capacities, have challenged the traditional views that proteins are the main protagonists of cellular functions and that RNA is merely an intermediary between DNA sequence and its encoded protein. There are many different kinds of products that are generated by this pervasive transcription; this review focuses on long noncoding RNAs (lncRNAs) that have shown spatial and temporal specific patterns of expression and regulation in a wide variety of cells and tissues, adding significant complexity to the understanding of their biological roles. Recent research has shed new light onto the biological function significance of lncRNAs. Here, we review the rapidly advancing field of lncRNAs, describing their biological features and their roles in regulation of gene expression. Moreover, we highlight some recent advances in our understanding of ncRNA-mediated regulation of stem cell pluripotency, morphogenesis, and development, focusing mainly on the regulatory roles of lncRNAs. Finally, we consider the potential medical implications, and the potential use of lncRNAs in drug development and discovery and in the identification of molecular markers of diseases, including cancer.
Collapse
Affiliation(s)
- Xiaolei Li
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing, 100853, China
| | | | | | | |
Collapse
|
580
|
Liu TT, Zhu D, Chen W, Deng W, He H, He G, Bai B, Qi Y, Chen R, Deng XW. A global identification and analysis of small nucleolar RNAs and possible intermediate-sized non-coding RNAs in Oryza sativa. MOLECULAR PLANT 2013; 6:830-46. [PMID: 22986792 PMCID: PMC3716300 DOI: 10.1093/mp/sss087] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 07/26/2012] [Indexed: 05/20/2023]
Abstract
Accumulating evidence suggests that non-coding RNAs (ncRNAs) are both widespread and functionally important in many eukaryotic organisms. In this study, we employed a special size fractionation and cDNA library construction method followed by 454 deep sequencing to systematically profile rice intermediate-size ncRNAs. Our analysis resulted in the identification of 1349 ncRNAs in total, including 754 novel ncRNAs of an unknown functional category. Chromosome distribution of all identified ncRNAs showed no strand bias, and displayed a pattern similar to that observed in protein-coding genes with few chromosome dependencies. More than half of the ncRNAs were centered around the plus-strand of the 5' and 3' termini of the coding regions. The majority of the novel ncRNAs were rice specific, while 78% of the small nucleolar RNAs (snoRNAs) were conserved. Tandem duplication drove the expansion of over half of the snoRNA gene families. Furthermore, 90% of the snoRNA candidates were shown to produce small RNAs between 20-30 nt, 80% of which were associated with ARGONAUT proteins generally, and AGO1b in particular. Overall, our findings provide a comprehensive view of an intermediate-size non-coding transcriptome in a monocot species, which will serve as a useful platform for an in-depth analysis of ncRNA functions.
Collapse
Affiliation(s)
- Ting-Ting Liu
- Peking-Yale Joint Research Center for Plant Molecular Genetics and Agrobiotechnology, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
- These authors contributed equally to this work
| | - Danmeng Zhu
- Peking-Yale Joint Research Center for Plant Molecular Genetics and Agrobiotechnology, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
- These authors contributed equally to this work
| | - Wei Chen
- Peking-Yale Joint Research Center for Plant Molecular Genetics and Agrobiotechnology, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
- These authors contributed equally to this work
| | - Wei Deng
- Bioinformatics Laboratory and National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing 100101, China
| | - Hang He
- Peking-Yale Joint Research Center for Plant Molecular Genetics and Agrobiotechnology, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
- Shenzhen Institute of Molecular Crop Design,
Shenzhen 518107, China
| | - Guangming He
- Peking-Yale Joint Research Center for Plant Molecular Genetics and Agrobiotechnology, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
| | - Baoyan Bai
- Bioinformatics Laboratory and National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing 100101, China
| | - Yijun Qi
- National Institute of Biological Sciences, No. 7 zhongguancun Life Science Park Road, Beijing 102206, China
| | - Runsheng Chen
- Bioinformatics Laboratory and National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing 100101, China
| | - Xing Wang Deng
- Peking-Yale Joint Research Center for Plant Molecular Genetics and Agrobiotechnology, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing 100871, China
- National Institute of Biological Sciences, No. 7 zhongguancun Life Science Park Road, Beijing 102206, China
- Shenzhen Institute of Molecular Crop Design,
Shenzhen 518107, China
- To whom correspondence should be addressed. D.Z. E-mail , tel. +86 10 62988781, Fax +86 10 62980457; X.W.D. E-mail , tel. (203) 432–8909, fax (203) 432–8908
| |
Collapse
|
581
|
Rijlaarsdam MA, Rijlaarsdam DJ, Gillis AJM, Dorssers LCJ, Looijenga LHJ. miMsg: a target enrichment algorithm for predicted miR–mRNA interactions based on relative ranking of matched expression data. Bioinformatics 2013; 29:1638-46. [DOI: 10.1093/bioinformatics/btt246] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
582
|
Pacifici M, Delbue S, Ferrante P, Jeansonne D, Kadri F, Nelson S, Velasco-Gonzalez C, Zabaleta J, Peruzzi F. Cerebrospinal fluid miRNA profile in HIV-encephalitis. J Cell Physiol 2013; 228:1070-5. [PMID: 23042033 DOI: 10.1002/jcp.24254] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 09/27/2012] [Indexed: 12/14/2022]
Abstract
MicroRNAs are short non-coding RNAs that modulate gene expression by translational repression. Because of their high stability in intracellular as well as extracellular environments, miRNAs have recently emerged as important biomarkers in several human diseases. However, they have not been tested in the cerebrospinal fluid (CSF) of HIV-1 positive individuals. Here, we present results of a study aimed at determining the feasibility of detecting miRNAs in the CSF of HIV-infected individuals with and without encephalitis (HIVE). We also evaluated similarities and differences between CSF and brain tissue miRNAs in the same clinical setting. We utilized a high throughput approach of miRNA detection arrays and identified differentially expressed miRNAs in the frontal cortex of three cases each of HIV+, HIVE, and HIV- controls, and CSF of 10 HIV-positive and 10 HIV-negative individuals. For the CSF samples, the group of HIV+ individuals contained nine cases of HIV-associated neurological disorders (HAND) and, among those, four had HIVE. All the HIV-negative samples had non-viral acute disseminate encephalomyelitis. A total of 66 miRNAs were found differentially regulated in HIV+ compared to HIV- groups. The greatest difference in miRNA expression was observed when four cases of HIVE were compared to five non-HIVE cases, previously normalized with the HIV-negative group. After statistical analyses, 11 miRNAs were fund significantly up-regulated in HIVE. Although more clinical samples should be examined, this work represents the first report of CSF miRNAs in HIV-infection and offers the basis for future investigation.
Collapse
Affiliation(s)
- Marco Pacifici
- LSU Health Sciences Center, School of Medicine, Stanley S Scott Cancer Center, Louisiana State University, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
583
|
Zhang W, Huang C, Gong Z, Zhao Y, Tang K, Li X, Fan S, Shi L, Li X, Zhang P, Zhou Y, Huang D, Liang F, Zhang X, Wu M, Cao L, Wang J, Li Y, Xiong W, Zeng Z, Li G. Expression of LINC00312, a long intergenic non-coding RNA, is negatively correlated with tumor size but positively correlated with lymph node metastasis in nasopharyngeal carcinoma. J Mol Histol 2013; 44:545-54. [PMID: 23529758 DOI: 10.1007/s10735-013-9503-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/12/2013] [Indexed: 12/17/2022]
Abstract
The long intergenic non-coding RNA LINC00312, also called NAG7, was first cloned by our group. Our previous studies have found that LINC00312 could inhibit proliferation and induce apoptosis in nasopharyngeal carcinoma (NPC) cells but also stimulate NPC cell invasion. However, the relevance of LINC00312 in NPC progression or in patient outcomes has not been reported. This study aims to assess the possible correlations of LINC00312 expression with NPC progression and its potential prognostic predictive ability in NPC outcomes. A NPC tissue microarray, which included 561 normal and NPC tissue cores, was used to detect LINC00312 expression, and we found that LINC00312 was significantly down-regulated in NPC tissues compared with non-cancerous nasopharyngeal epithelium tissues. Positive expression of LINC00312 was negatively correlated with tumor size (P < 0.001) but positively correlated with lymph node metastasis (P = 0.002). A receiver operating characteristic (ROC) analysis revealed that LINC00312 expression could distinguish non-cancerous patients from NPC patients (P < 0.001, sensitivity: 72.1 %, specificity: 87.7 %). We also found that LINC00312 was strongly negatively correlated with EBER-1, a non-coding RNA transcribed by Epstein-Barr Virus, in NPC (r = -0.384, P < 0.001). In the final logistic regression analysis model, the abnormal expression of LINC00312 and EBER-1 were found to be independent contributors to nasopharyngeal carcinogenesis (P < 0.001, P < 0.001, respectively). A survival analysis revealed that LINC00312 could predict a good prognosis of no lymph node metastasis (Disease Free Survival (DFS): P = 0.005, Overall Survival (OS): P = 0.001) and a poor prognosis of lymph node metastasis (DFS: P = 0.011, OS: P = 0.001) in NPC patients. Low expression of LINC00312 was an independent risk factor for OS in multivariate analyses (P = 0.017). These observations indicated that LINC00312 could represent a potential biomarker for metastasis, progression and prognosis in NPC.
Collapse
Affiliation(s)
- Wenling Zhang
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
584
|
Abstract
The gene expression programs that establish and maintain specific cell states in humans are controlled by thousands of transcription factors, cofactors, and chromatin regulators. Misregulation of these gene expression programs can cause a broad range of diseases. Here, we review recent advances in our understanding of transcriptional regulation and discuss how these have provided new insights into transcriptional misregulation in disease.
Collapse
Affiliation(s)
- Tong Ihn Lee
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Richard A. Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts
| |
Collapse
|
585
|
Benetatos L, Hatzimichael E, Londin E, Vartholomatos G, Loher P, Rigoutsos I, Briasoulis E. The microRNAs within the DLK1-DIO3 genomic region: involvement in disease pathogenesis. Cell Mol Life Sci 2013; 70:795-814. [PMID: 22825660 PMCID: PMC11114045 DOI: 10.1007/s00018-012-1080-8] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Revised: 06/26/2012] [Accepted: 06/28/2012] [Indexed: 02/07/2023]
Abstract
The mammalian genome is transcribed in a developmentally regulated manner, generating RNA strands ranging from long to short non-coding RNA (ncRNAs). NcRNAs generated by intergenic sequences and protein-coding loci, represent up to 98 % of the human transcriptome. Non-coding transcripts comprise short ncRNAs such as microRNAs, piwi-interacting RNAs, small nucleolar RNAs and long intergenic RNAs, most of which exercise a strictly controlled negative regulation of expression of protein-coding genes. In humans, the DLK1-DIO3 genomic region, located on human chromosome 14 (14q32) contains the paternally expressed imprinted genes DLK1, RTL1, and DIO3 and the maternally expressed imprinted genes MEG3 (Gtl2), MEG8 (RIAN), and antisense RTL1 (asRTL1). This region hosts, in addition to two long intergenic RNAs, the MEG3 and MEG8, one of the largest microRNA clusters in the genome, with 53 miRNAs in the forward strand and one (mir-1247) in the reverse strand. Many of these miRNAs are differentially expressed in several pathologic processes and various cancers. A better understanding of the pathophysiologic importance of the DLK1-DIO3 domain-containing microRNA cluster may contribute to innovative therapeutic strategies in a range of diseases. Here we present an in-depth review of this vital genomic region, and examine the role the microRNAs of this region may play in controlling tissue homeostasis and in the pathogenesis of some human diseases, mostly cancer, when aberrantly expressed. The potential clinical implications of this data are also discussed.
Collapse
Affiliation(s)
- Leonidas Benetatos
- Transfusion Unit, General Hospital of Preveza, Selefkias 2, 48100, Preveza, Greece.
| | | | | | | | | | | | | |
Collapse
|
586
|
Abstract
In this review, we focus on the roles of long noncoding RNAs (lncRNAs), including cellular and viral lncRNAs, in virus replication in infected cells. We survey the interactions and functions of several cellular lncRNAs such as XIST, HOTAIR, NEAT1, BIC, and several virus-encoded lncRNAs.
Collapse
Affiliation(s)
- Quan Zhang
- Molecular Virology Section, Laboratory of Molecular Microbiology, National Institutes of Allergy and Infectious Diseases, the National Institutes of Health, Bethesda, Maryland 20892-0460, USA
| | | |
Collapse
|
587
|
Kim T, Reitmair A. Non-Coding RNAs: Functional Aspects and Diagnostic Utility in Oncology. Int J Mol Sci 2013; 14:4934-68. [PMID: 23455466 PMCID: PMC3634484 DOI: 10.3390/ijms14034934] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/09/2013] [Accepted: 02/18/2013] [Indexed: 02/06/2023] Open
Abstract
Noncoding RNAs (ncRNAs) have been found to have roles in a large variety of biological processes. Recent studies indicate that ncRNAs are far more abundant and important than initially imagined, holding great promise for use in diagnostic, prognostic, and therapeutic applications. Within ncRNAs, microRNAs (miRNAs) are the most widely studied and characterized. They have been implicated in initiation and progression of a variety of human malignancies, including major pathologies such as cancers, arthritis, neurodegenerative disorders, and cardiovascular diseases. Their surprising stability in serum and other bodily fluids led to their rapid ascent as a novel class of biomarkers. For example, several properties of stable miRNAs, and perhaps other classes of ncRNAs, make them good candidate biomarkers for early cancer detection and for determining which preneoplastic lesions are likely to progress to cancer. Of particular interest is the identification of biomarker signatures, which may include traditional protein-based biomarkers, to improve risk assessment, detection, and prognosis. Here, we offer a comprehensive review of the ncRNA biomarker literature and discuss state-of-the-art technologies for their detection. Furthermore, we address the challenges present in miRNA detection and quantification, and outline future perspectives for development of next-generation biodetection assays employing multicolor alternating-laser excitation (ALEX) fluorescence spectroscopy.
Collapse
Affiliation(s)
- Taiho Kim
- Nesher Technologies, Inc., 2100 W. 3rd St. Los Angeles, CA 90057, USA.
| | | |
Collapse
|
588
|
Frese KS, Katus HA, Meder B. Next-generation sequencing: from understanding biology to personalized medicine. BIOLOGY 2013; 2:378-98. [PMID: 24832667 PMCID: PMC4009863 DOI: 10.3390/biology2010378] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 01/21/2013] [Accepted: 02/04/2013] [Indexed: 12/11/2022]
Abstract
Within just a few years, the new methods for high-throughput next-generation sequencing have generated completely novel insights into the heritability and pathophysiology of human disease. In this review, we wish to highlight the benefits of the current state-of-the-art sequencing technologies for genetic and epigenetic research. We illustrate how these technologies help to constantly improve our understanding of genetic mechanisms in biological systems and summarize the progress made so far. This can be exemplified by the case of heritable heart muscle diseases, so-called cardiomyopathies. Here, next-generation sequencing is able to identify novel disease genes, and first clinical applications demonstrate the successful translation of this technology into personalized patient care.
Collapse
Affiliation(s)
- Karen S Frese
- Department of Internal Medicine III, University of Heidelberg, Heidelberg 69120, Germany.
| | - Hugo A Katus
- Department of Internal Medicine III, University of Heidelberg, Heidelberg 69120, Germany.
| | - Benjamin Meder
- Department of Internal Medicine III, University of Heidelberg, Heidelberg 69120, Germany.
| |
Collapse
|
589
|
Long non-coding RNA in cancer. Int J Mol Sci 2013; 14:4655-69. [PMID: 23443164 PMCID: PMC3634483 DOI: 10.3390/ijms14034655] [Citation(s) in RCA: 290] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 01/03/2013] [Accepted: 01/31/2013] [Indexed: 12/31/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are pervasively transcribed in the genome and are emerging as new players in tumorigenesis due to their various functions in transcriptional, posttranscriptional and epigenetic mechanisms of gene regulation. LncRNAs are deregulated in a number of cancers, demonstrating both oncogenic and tumor suppressive roles, thus suggesting their aberrant expression may be a substantial contributor in cancer development. In this review, we will summarize their emerging role in human cancer and discuss their perspectives in diagnostics as potential biomarkers.
Collapse
|
590
|
Crujeiras AB, Díaz-Lagares A, Carreira MC, Amil M, Casanueva FF. Oxidative stress associated to dysfunctional adipose tissue: a potential link between obesity, type 2 diabetes mellitus and breast cancer. Free Radic Res 2013; 47:243-56. [PMID: 23409968 DOI: 10.3109/10715762.2013.772604] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus and breast cancer are two important health problems. Type 2 diabetes (T2DM) and obesity are closely linked with both being associated with breast cancer. Despite abundant epidemiological data, there is no definitive evidence regarding the mechanisms responsible for this association. The proposed mechanisms by which diabetes affects breast cancer risk and prognosis are the same as the mechanisms hypothesised for the contribution of obesity to breast cancer risk. The obesity-induced inflammation promoted by adipose tissue dysfunction is a key feature, which is thought to be an important link between obesity and cancer. Inflammation induces an increase in free radicals and subsequently promotes oxidative stress, which may create a microenvironment favourable to the tumor development in obese persons. Oxidative stress is also proposed as the link between obesity and diabetes mellitus. Therefore, obesity-related oxidative stress could be a direct cause of neoplastic transformation associated with obesity and T2DM in breast cancer cells. This review is focused on the role of obesity-related oxidative stress in the context of chronic inflammation, on the time of breast cancer onset and progression, which provide targets for preventive and therapeutic strategies in the fields of diabetes and obesity-related breast cancer.
Collapse
Affiliation(s)
- A B Crujeiras
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute, Barcelona, Spain.
| | | | | | | | | |
Collapse
|
591
|
Webster ALH, Yan MSC, Marsden PA. Epigenetics and cardiovascular disease. Can J Cardiol 2013; 29:46-57. [PMID: 23261320 DOI: 10.1016/j.cjca.2012.10.023] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/26/2012] [Accepted: 10/28/2012] [Indexed: 12/18/2022] Open
Abstract
A commonly-assumed paradigm holds that the primary genetic determinant of cardiovascular disease resides within the DNA sequence of our genes. This paradigm can be challenged. For example, how do sequence changes in the non-coding region of the genome influence phenotype? Why are all diseases not shared between identical twins? Part of the answer lies in the fact that the environment or exogenous stimuli clearly influence disease susceptibility, but it was unclear in the past how these effects were signalled to the static DNA code. Epigenetics is providing a newer perspective on these issues. Epigenetics refers to chromatin-based mechanisms important in the regulation of gene expression that do not involve changes to the DNA sequence per se. The field can be broadly categorized into three areas: DNA base modifications (including cytosine methylation and cytosine hydroxymethylation), post-translational modifications of histone proteins, and RNA-based mechanisms that operate in the nucleus. Cardiovascular disease pathways are now being approached from the epigenetic perspective, including those associated with atherosclerosis, angiogenesis, ischemia-reperfusion damage, and the cardiovascular response to hypoxia and shear stress, among many others. With increasing interest and expanding partnerships in the field, we can expect new insights to emerge from epigenetic perspectives of cardiovascular health. This paper reviews the principles governing epigenetic regulation, discusses their presently-understood importance in cardiovascular disease, and considers the growing significance we are likely to attribute to epigenetic contributions in the future, as they provide new mechanistic insights and a host of novel clinical applications.
Collapse
Affiliation(s)
- Andrew L H Webster
- Keenan Research Centre and Li Ka Shing Knowledge Institute, St Michael's Hospital, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
592
|
Lukiw WJ, Andreeva TV, Grigorenko AP, Rogaev EI. Studying micro RNA Function and Dysfunction in Alzheimer's Disease. Front Genet 2013; 3:327. [PMID: 23390425 PMCID: PMC3565163 DOI: 10.3389/fgene.2012.00327] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 12/28/2012] [Indexed: 01/18/2023] Open
Abstract
Alzheimer’s disease (AD) is a tragic, progressive, age-related neurological dysfunction, representing one of the most prevalent neurodegenerative disorders in industrialized societies. Globally, 5 million new cases of AD are diagnosed annually, with one new AD case being reported every 7 s. Most recently there has been a surge in the study of the regulatory mechanisms of the AD process, and the particular significance of small non-coding ∼22 ribonucleotide RNAs called micro RNAs (miRNAs). Abundant data have profiled miRNA patterns in healthy, aging brain, in mild cognitive impairment (MCI), and in the moderate- and late-stages of AD. The major mode of action of miRNA is to interact, via base-pair complementarity, with ribonucleotides located within the 3′ untranslated region (3′-UTR) of multiple target messenger RNAs (mRNAs), and in doing so decrease the capability of that specific mRNA to be expressed. Many miRNAs are highly cell- and tissue-specific. The human brain appears to use only a highly specific fraction of all known human miRNAs, whose speciation and complexity are defined as a discrete subset of all known small non-coding RNAs (sncRNAs) in the brain. In general, in contrast to normally, aging human brain, in AD a family of pathogenically up-regulated miRNAs appear to be down-regulating the expression certain brain-essential mRNA targets, including key regulatory genes involved interactively in neuroinflammation, synaptogenesis, neurotrophic functions, and amyloidogenesis. These up-regulated, NF-kB-sensitive miRNAs, involved in the innate immune and inflammatory response and synaptic, neurotrophic, and amyloidogenic functions include miRNA-9, miRNA-125b, miRNA-146a, and miRNA-155. Other miRNAs of the miRNA-15/107 family, miRNA-153 and miRNA-190, and others, will be discussed. Overall, this manuscript will review the known contribution of miRNAs to aging brain function and the role they appear to play in the incidence and progression of AD.
Collapse
Affiliation(s)
- Walter J Lukiw
- Department of Neurology, LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, LA, USA ; Department of Ophthalmology, LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | | | | | | |
Collapse
|
593
|
Clifford RL, Singer CA, John AE. Epigenetics and miRNA emerge as key regulators of smooth muscle cell phenotype and function. Pulm Pharmacol Ther 2013; 26:75-85. [PMID: 22800879 PMCID: PMC4076625 DOI: 10.1016/j.pupt.2012.07.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 07/02/2012] [Accepted: 07/04/2012] [Indexed: 10/28/2022]
Abstract
Regulation of phenotypic plasticity in smooth muscle requires an understanding of the mechanisms regulating phenotype-specific genes and the processes dysregulated during pathogenesis. Decades of study in airway smooth muscle has provided extensive knowledge of the gene expression patterns and signaling pathways necessary to maintain and alter smooth muscle cell phenotype. With this solid foundation, the importance and complexity of inheritable epigenetic modifications and mechanisms silencing gene expression have now emerged as fundamental components regulating aspects of inflammation, proliferation and remodeling.
Collapse
Affiliation(s)
- Rachel L. Clifford
- University of Nottingham Division of Respiratory Medicine and Nottingham Respiratory Research Unit Clinical Sciences Building, City Hospital Hucknall Road, Nottingham NG5 1PB, England, UK
| | - Cherie A. Singer
- University of Nevada School of Medicine Center for Molecular Medicine 573 Department of Pharmacology, Reno, NV 89557, USA
| | - Alison E. John
- Corresponding Author University of Nottingham Division of Respiratory Medicine and Nottingham Respiratory Research Unit Clinical Sciences Building, City Hospital Hucknall Road, Nottingham NG5 1PB, England, UK Tel:+44 115 8231106 Fax: +44 115 8231946
| |
Collapse
|
594
|
NEAT1 long noncoding RNA and paraspeckle bodies modulate HIV-1 posttranscriptional expression. mBio 2013; 4:e00596-12. [PMID: 23362321 PMCID: PMC3560530 DOI: 10.1128/mbio.00596-12] [Citation(s) in RCA: 300] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Most of the human genome is transcribed into protein-noncoding RNAs (ncRNAs), including small ncRNAs and long ncRNAs (lncRNAs). Over the past decade, rapidly emerging evidence has increasingly supported the view that lncRNAs serve key regulatory and functional roles in mammal cells. HIV-1 replication relies on various cell functions. To date, while the involvement of host protein factors and microRNAs (miRNAs) in the HIV-1 life cycle has been extensively studied, the relationship between lncRNAs and HIV-1 remains uncharacterized. Here, we have profiled 83 disease-related lncRNAs in HIV-1-infected T cells. We found NEAT1 to be one of several lncRNAs whose expression is changed by HIV-1 infection, and we have characterized its role in HIV-1 replication. We In the abstract, added definition of INS OK, or should "cis-acting" be added?report here that the knockdown of NEAT1 enhances virus production through increased nucleus-to-cytoplasm export of Rev-dependent instability element (INS)-containing HIV-1 mRNAs. IMPORTANCE Long protein-noncoding RNAs (lncRNAs) play roles in regulating gene expression and modulating protein activities. There is emerging evidence that lncRNAs are involved in the replication of viruses. To our knowledge, this report is the first to characterize a role contributed by an lncRNA, NEAT1, to HIV-1 replication. NEAT1 is essential for the integrity of the nuclear paraspeckle substructure. Based on our findings from NEAT1 knockdown, we have identified the nuclear paraspeckle body as another important subcellular organelle for HIV-1 replication.
Collapse
|
595
|
Cairns MJ, Kocerha J. Advances in non-coding RNA profiling for neurological diseases. Front Genet 2013; 4:5. [PMID: 23355845 PMCID: PMC3554880 DOI: 10.3389/fgene.2013.00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/02/2013] [Indexed: 11/23/2022] Open
Affiliation(s)
- Murray J Cairns
- School of Biomedical Sciences and Pharmacy, and Hunter Medical Research Institute, University of Newcastle Callaghan, NSW, Australia ; Schizophrenia Research Institute Sydney, NSW, Australia
| | | |
Collapse
|
596
|
Youngson NA, Morris MJ. What obesity research tells us about epigenetic mechanisms. Philos Trans R Soc Lond B Biol Sci 2013; 368:20110337. [PMID: 23166398 PMCID: PMC3539363 DOI: 10.1098/rstb.2011.0337] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathophysiology of obesity is extremely complex and is associated with extensive gene expression changes in tissues throughout the body. This situation, combined with the fact that all gene expression changes are thought to have associated epigenetic changes, means that the links between obesity and epigenetics will undoubtedly be vast. Much progress in identifying epigenetic changes induced by (or inducing) obesity has already been made, with candidate and genome-wide approaches. These discoveries will aid the clinician through increasing our understanding of the inheritance, development and treatment of obesity. However, they are also of great value for epigenetic researchers, as they have revealed mechanisms of environmental interactions with epigenetics that can produce or perpetuate a disease state. Here, we will review the evidence for four mechanisms through which epigenetics contributes to obesity: as downstream effectors of environmental signals; through abnormal global epigenetic state driving obesogenic expression patterns; through facilitating developmental programming and through transgenerational epigenetic inheritance.
Collapse
Affiliation(s)
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, High Street, Kensington, New South Wales 2052, Australia
| |
Collapse
|
597
|
Alvarez ML, Distefano JK. The role of non-coding RNAs in diabetic nephropathy: potential applications as biomarkers for disease development and progression. Diabetes Res Clin Pract 2013; 99:1-11. [PMID: 23102915 DOI: 10.1016/j.diabres.2012.10.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 09/11/2012] [Accepted: 10/05/2012] [Indexed: 01/10/2023]
Abstract
Diabetic nephropathy, a progressive kidney disease that develops secondary to diabetes, is the major cause of chronic kidney disease in developed countries, and contributes significantly to increased morbidity and mortality among individuals with diabetes. Although the causes of diabetic nephropathy are not fully understood, recent studies demonstrate a role for epigenetic factors in the development of the disease. For example, non-coding RNA (ncRNA) molecules, including microRNAs (miRNAs), have been shown to be functionally important in modulating renal response to hyperglycemia and progression of diabetic nephropathy. Characterization of miRNA expression in diabetic nephropathy from studies of animal models of diabetes, and in vitro investigations using different types of kidney cells also support this role. The goal of this review, therefore, is to summarize the current state of knowledge of specific ncRNAs involved in the development of diabetic nephropathy, with a focus on the potential role of miRNAs to serve as sensitive, non-invasive biomarkers of kidney disease and progression. Non-coding RNAs are currently recognized as potentially important regulators of genes involved in processes related to the development of diabetic nephropathy, and as such, represent viable targets for both clinical diagnostic strategies and therapeutic intervention.
Collapse
Affiliation(s)
- M Lucrecia Alvarez
- Diabetes, Cardiovascular and Metabolic Diseases Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, United States
| | | |
Collapse
|
598
|
Guo X, Gao L, Liao Q, Xiao H, Ma X, Yang X, Luo H, Zhao G, Bu D, Jiao F, Shao Q, Chen R, Zhao Y. Long non-coding RNAs function annotation: a global prediction method based on bi-colored networks. Nucleic Acids Res 2013; 41:e35. [PMID: 23132350 PMCID: PMC3554231 DOI: 10.1093/nar/gks967] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Revised: 09/05/2012] [Accepted: 09/25/2012] [Indexed: 02/01/2023] Open
Abstract
More and more evidences demonstrate that the long non-coding RNAs (lncRNAs) play many key roles in diverse biological processes. There is a critical need to annotate the functions of increasing available lncRNAs. In this article, we try to apply a global network-based strategy to tackle this issue for the first time. We develop a bi-colored network based global function predictor, long non-coding RNA global function predictor ('lnc-GFP'), to predict probable functions for lncRNAs at large scale by integrating gene expression data and protein interaction data. The performance of lnc-GFP is evaluated on protein-coding and lncRNA genes. Cross-validation tests on protein-coding genes with known function annotations indicate that our method can achieve a precision up to 95%, with a suitable parameter setting. Among the 1713 lncRNAs in the bi-colored network, the 1625 (94.9%) lncRNAs in the maximum connected component are all functionally characterized. For the lncRNAs expressed in mouse embryo stem cells and neuronal cells, the inferred putative functions by our method highly match those in the known literature.
Collapse
Affiliation(s)
- Xingli Guo
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Lin Gao
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Qi Liao
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Hui Xiao
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Xiaoke Ma
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Xiaofei Yang
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Haitao Luo
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Guoguang Zhao
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Dechao Bu
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Fei Jiao
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Qixiang Shao
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - RunSheng Chen
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| | - Yi Zhao
- School of computer science and technology, XIDIAN
UNIVERSITY, 2 South Taibai Road, Xi’an Shaanxi, 710071, PR China,
Bioinformatics Research Group, Key Laboratory of Intelligent Information
Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese
Academy of Sciences, No.6 Kexueyuan South Road Zhongguancun, Haidian District, Beijing,
100190, PR China, School of Software Engineering, XIDIAN UNIVERSITY, 2 South
Taibai Road, Xi‘an Shaanxi, 710071, PR China, Institute of Biochemistry
and Molecular Biology, School of Medicine, Ningbo University, 818 Fenghua Road Ningbo
Zhejiang, 315211, PR China, University of Chinese Academy of Sciences, No.19A
Yuquan Road, Beijing, 100049, PR China, Department of Biochemistry and Molecular
Biology, Binzhou Medical College, 346 GuanHai Road, Yantai Shandong, 264003, PR China,
Department of Immunology, School of Medical Science and Laboratory Medicine,
Jiangsu University, 301 Xuefu Road, Zhenjiang Jiangsu, 212013, PR China and
Bioinformatics Laboratory and National Laboratory of Biomacromolecules,
Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District,
Beijing, 100101, PR China
| |
Collapse
|
599
|
Ortogero N, Hennig GW, Langille C, Ro S, McCarrey JR, Yan W. Computer-assisted annotation of murine Sertoli cell small RNA transcriptome. Biol Reprod 2013; 88:3. [PMID: 23136297 PMCID: PMC4434943 DOI: 10.1095/biolreprod.112.102269] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 07/17/2012] [Accepted: 11/05/2012] [Indexed: 01/02/2023] Open
Abstract
Mammalian genomes encode a large number of small noncoding RNAs (sncRNAs) that play regulatory roles during development and adulthood by affecting gene expression. Several sncRNA species, including microRNAs (miRNAs), piwi-interacting RNAs (piRNAs), endogenous small interfering RNAs (endo-siRNAs), and small nucleolar RNAs (snoRNAs), are abundantly expressed in the testis and required for normal testicular development and spermatogenesis. To evaluate global changes in sncRNA expression, the next-generation sequencing (NGS)-based sncRNA transcriptomic analysis has become routine, because it allows rapid determination of the small RNA transcriptome of a particular testicular cell type. However, annotation of small RNA NGS reads can be challenging due to the volume of reads obtained, which is usually in the millions. Therefore, we developed a computer-assisted sncRNA annotation protocol that could identify not only known sncRNAs but also previously uncharacterized ones. Using this protocol, we annotated NGS reads of a Sertoli cell sncRNA library, and we report to our knowledge the first comprehensive annotation of the sncRNA transcriptome of immature murine Sertoli cells. Moreover, the computer-assisted sncRNA annotation pipeline that we report is applicable for annotating NGS reads derived from other cell types and/or sequencing platforms.
Collapse
Affiliation(s)
- Nicole Ortogero
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Grant W. Hennig
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Chad Langille
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - John R. McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
600
|
Lee S, Vasudevan S. Post-transcriptional stimulation of gene expression by microRNAs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 768:97-126. [PMID: 23224967 DOI: 10.1007/978-1-4614-5107-5_7] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
MicroRNAs are small noncoding RNA regulatory molecules that control gene expression by guiding associated effector complexes to other RNAs via sequence-specific recognition of target sites. Misregulation of microRNAs leads to a wide range of diseases including cancers, inflammatory and developmental disorders. MicroRNAs were found to mediate deadenylation-dependent decay and translational repression of messages through partially complementary microRNA target sites in the 3'-UTR (untranslated region). A growing series of studies has demonstrated that microRNAs and their associated complexes (microRNPs) elicit alternate functions that enable stimulation of gene expression in addition to their assigned repressive roles. These reports, discussed in this chapter, indicate that microRNA-mediated effects via natural 3' and 5'-UTRs can be selective and controlled, dictated by the RNA sequence context, associated complex, and cellular conditions. Similar to the effects of repression, upregulated gene expression by microRNAs varies from small refinements to significant amplifications in expression. An emerging theme from this literature is that microRNAs have a versatile range of abilities to manipulate post-transcriptional control mechanisms leading to controlled gene expression. These studies reveal new potentials for microRNPs in gene expression control that develop as responses to specific cellular conditions.
Collapse
|