501
|
Esguerra JLS, Mollet IG, Salunkhe VA, Wendt A, Eliasson L. Regulation of Pancreatic Beta Cell Stimulus-Secretion Coupling by microRNAs. Genes (Basel) 2014; 5:1018-31. [PMID: 25383562 PMCID: PMC4276924 DOI: 10.3390/genes5041018] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/01/2014] [Accepted: 10/21/2014] [Indexed: 12/31/2022] Open
Abstract
Increased blood glucose after a meal is countered by the subsequent increased release of the hypoglycemic hormone insulin from the pancreatic beta cells. The cascade of molecular events encompassing the initial sensing and transport of glucose into the beta cell, culminating with the exocytosis of the insulin large dense core granules (LDCVs) is termed "stimulus-secretion coupling." Impairment in any of the relevant processes leads to insufficient insulin release, which contributes to the development of type 2 diabetes (T2D). The fate of the beta cell, when exposed to environmental triggers of the disease, is determined by the possibility to adapt to the new situation by regulation of gene expression. As established factors of post-transcriptional regulation, microRNAs (miRNAs) are well-recognized mediators of beta cell plasticity and adaptation. Here, we put focus on the importance of comprehending the transcriptional regulation of miRNAs, and how miRNAs are implicated in stimulus-secretion coupling, specifically those influencing the late stages of insulin secretion. We suggest that efficient beta cell adaptation requires an optimal balance between transcriptional regulation of miRNAs themselves, and miRNA-dependent gene regulation. The increased knowledge of the beta cell transcriptional network inclusive of non-coding RNAs such as miRNAs is essential in identifying novel targets for the treatment of T2D.
Collapse
Affiliation(s)
- Jonathan L S Esguerra
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| | - Inês G Mollet
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| | - Vishal A Salunkhe
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| | - Anna Wendt
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| | - Lena Eliasson
- Unit of Islet Cell Exocytosis, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, CRC 91-11, Jan Waldenströms gata 35, 205 02 Malmö, Sweden.
| |
Collapse
|
502
|
Carnero E, Barriocanal M, Segura V, Guruceaga E, Prior C, Börner K, Grimm D, Fortes P. Type I Interferon Regulates the Expression of Long Non-Coding RNAs. Front Immunol 2014; 5:548. [PMID: 25414701 PMCID: PMC4222131 DOI: 10.3389/fimmu.2014.00548] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/14/2014] [Indexed: 12/22/2022] Open
Abstract
Interferons (IFNs) are key players in the antiviral response. IFN sensing by the cell activates transcription of IFN-stimulated genes (ISGs) able to induce an antiviral state by affecting viral replication and release. IFN also induces the expression of ISGs that function as negative regulators to limit the strength and duration of IFN response. The ISGs identified so far belong to coding genes. However, only a small proportion of the transcriptome corresponds to coding transcripts and it has been estimated that there could be as many coding as long non-coding RNAs (lncRNAs). To address whether IFN can also regulate the expression of lncRNAs, we analyzed the transcriptome of HuH7 cells treated or not with IFNα2 by expression arrays. Analysis of the arrays showed increased levels of several well-characterized coding genes that respond to IFN both at early or late times. Furthermore, we identified several IFN-stimulated or -downregulated lncRNAs (ISRs and IDRs). Further validation showed that ISR2, 8, and 12 expression mimics that of their neighboring genes GBP1, IRF1, and IL6, respectively, all related to the IFN response. These genes are induced in response to different doses of IFNα2 in different cell lines at early (ISR2 or 8) or later (ISR12) time points. IFNβ also induced the expression of these lncRNAs. ISR2 and 8 were also induced by an influenza virus unable to block the IFN response but not by other wild-type lytic viruses tested. Surprisingly, both ISR2 and 8 were significantly upregulated in cultured cells and livers from patients infected with HCV. Increased levels of ISR2 were also detected in patients chronically infected with HIV. This is relevant as genome-wide guilt-by-association studies predict that ISR2, 8, and 12 may function in viral processes, in the IFN pathway and the antiviral response. Therefore, we propose that these lncRNAs could be induced by IFN to function as positive or negative regulators of the antiviral response.
Collapse
Affiliation(s)
- Elena Carnero
- Department of Gene therapy and Hepatology, Center for Applied Medical Research (CIMA), University of Navarra , Pamplona , Spain
| | - Marina Barriocanal
- Department of Gene therapy and Hepatology, Center for Applied Medical Research (CIMA), University of Navarra , Pamplona , Spain
| | - Victor Segura
- Bioinformatics Unit, Center for Applied Medical Research (CIMA), University of Navarra , Pamplona , Spain
| | - Elizabeth Guruceaga
- Bioinformatics Unit, Center for Applied Medical Research (CIMA), University of Navarra , Pamplona , Spain
| | - Celia Prior
- Department of Gene therapy and Hepatology, Center for Applied Medical Research (CIMA), University of Navarra , Pamplona , Spain
| | - Kathleen Börner
- Centre for Infectious Diseases/Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks , Heidelberg , Germany ; German Center for Infection Research (DZIF) , Heidelberg , Germany
| | - Dirk Grimm
- Centre for Infectious Diseases/Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks , Heidelberg , Germany
| | - Puri Fortes
- Department of Gene therapy and Hepatology, Center for Applied Medical Research (CIMA), University of Navarra , Pamplona , Spain
| |
Collapse
|
503
|
Shahryari A, Rafiee MR, Fouani Y, Oliae NA, Samaei NM, Shafiee M, Semnani S, Vasei M, Mowla SJ. Two novel splice variants of SOX2OT, SOX2OT-S1, and SOX2OT-S2 are coupregulated with SOX2 and OCT4 in esophageal squamous cell carcinoma. Stem Cells 2014; 32:126-34. [PMID: 24105929 DOI: 10.1002/stem.1542] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 08/12/2013] [Indexed: 01/18/2023]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as new regulators of stem cell pluripotency and tumorigenesis. The SOX2 gene, a master regulator of pluripotency, is embedded within the third intron of a lncRNA known as SOX2 overlapping transcript (SOX2OT). SOX2OT has been suspected to participate in regulation of SOX2 expression and/or other related processes; nevertheless, its potential involvement in tumor initiation and/or progression is unclear. Here, we have evaluated a possible correlation between expression patterns of SOX2OT and those of master regulators of pluripotency, SOX2 and OCT4, in esophageal squamous cell carcinoma (ESCC) tissue samples. We have also examined its potential function in the human embryonic carcinoma stem cell line, NTERA2 (NT2), which highly expresses SOX2OT, SOX2, and OCT4. Our data revealed a significant coupregulation of SOX2OT along with SOX2 and OCT4 in tumor samples, compared to the non-tumor tissues obtained from the margin of same tumors. We also identified two novel splice variants of SOX2OT (SOX2OT-S1 and SOX2OT-S2) which coupregulated with SOX2 and OCT4 in ESCCs. Suppressing SOX2OT variants caused a profound alteration in cell cycle distribution, including a 5.9 and 6.9 time increase in sub-G1 phase of cell cycle for SOX2OT-S1 and SOX2OT-S2, respectively. The expression of all variants was significantly diminished, upon the induction of neural differentiation in NT2 cells, suggesting their potential functional links to the undifferentiated state of the cells. Our data suggest a part for SOX2OT spliced variants in tumor initiation and/or progression as well as regulating pluripotent state of stem cells.
Collapse
Affiliation(s)
- Alireza Shahryari
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
504
|
Ruan J, Liu X, Xiong X, Zhang C, Li J, Zheng H, Huang C, Shi Q, Weng Y. miR‑107 promotes the erythroid differentiation of leukemia cells via the downregulation of Cacna2d1. Mol Med Rep 2014; 11:1334-9. [PMID: 25373460 DOI: 10.3892/mmr.2014.2865] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 09/12/2014] [Indexed: 11/06/2022] Open
Abstract
microRNAs (miRNAs) have been reported to be involved in various human diseases. They may have uses in diagnosis and as therapeutic targets, thus the discovery of novel miRNAs has the potential to provide clinical tools or shed light on novel mechanisms. In the current study, miR‑107 was revealed to be downregulated in chronic myeloid leukemia cells. Overexpression of miR‑107 in K562 and KCL‑22 chronic myeloid leukemia cells promotes erythroid differentiation, while having no effect on cell proliferation. Further bioinformatics predicted that one target of miR‑107 may be Cacna2d1, a calcium channel protein. A luciferase reporter assay and quantitative polymerase chain reaction were utilized to confirm that Cacna2d1 is a target molecule of miR-107. The effect of miR‑107 on K562 and KCL‑22 cells was mediated through the downregulation of Cacna2d1, as rescued expression of Cacna2d1 reversed the effects of miR‑107. In summary, the current study identified a novel miRNA that is involved in chronic myeloid leukemia cell erythroid differentiation and the associated mechanisms, making it a potential therapeutic target in the treatment of chronic myeloid leukemia.
Collapse
Affiliation(s)
- Jie Ruan
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xinguang Liu
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xingdong Xiong
- Institute of Aging Research, Guangdong Medical College, Dongguan, Guandong 523808, P.R. China
| | - Chunlong Zhang
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jiangbin Li
- Institute of Aging Research, Guangdong Medical College, Dongguan, Guandong 523808, P.R. China
| | - Huiling Zheng
- Institute of Aging Research, Guangdong Medical College, Dongguan, Guandong 523808, P.R. China
| | - Chirong Huang
- Institute of Laboratory Medicine, Guangdong Medical College, Dongguan, Guandong 523808, P.R. China
| | - Qiong Shi
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yaguang Weng
- Department of Clinical Laboratory, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
505
|
van Dam S, Craig T, de Magalhães JP. GeneFriends: a human RNA-seq-based gene and transcript co-expression database. Nucleic Acids Res 2014; 43:D1124-32. [PMID: 25361971 PMCID: PMC4383890 DOI: 10.1093/nar/gku1042] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Co-expression networks have proven effective at assigning putative functions to genes based on the functional annotation of their co-expressed partners, in candidate gene prioritization studies and in improving our understanding of regulatory networks. The growing number of genome resequencing efforts and genome-wide association studies often identify loci containing novel genes and there is a need to infer their functions and interaction partners. To facilitate this we have expanded GeneFriends, an online database that allows users to identify co-expressed genes with one or more user-defined genes. This expansion entails an RNA-seq-based co-expression map that includes genes and transcripts that are not present in the microarray-based co-expression maps, including over 10 000 non-coding RNAs. The results users obtain from GeneFriends include a co-expression network as well as a summary of the functional enrichment among the co-expressed genes. Novel insights can be gathered from this database for different splice variants and ncRNAs, such as microRNAs and lincRNAs. Furthermore, our updated tool allows candidate transcripts to be linked to diseases and processes using a guilt-by-association approach. GeneFriends is freely available from http://www.GeneFriends.org and can be used to quickly identify and rank candidate targets relevant to the process or disease under study.
Collapse
Affiliation(s)
- Sipko van Dam
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Thomas Craig
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
506
|
McMahon M, Contreras A, Ruggero D. Small RNAs with big implications: new insights into H/ACA snoRNA function and their role in human disease. WILEY INTERDISCIPLINARY REVIEWS-RNA 2014; 6:173-89. [PMID: 25363811 DOI: 10.1002/wrna.1266] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/25/2014] [Accepted: 08/07/2014] [Indexed: 12/26/2022]
Abstract
A myriad of structurally and functionally diverse noncoding RNAs (ncRNAs) have recently been implicated in numerous human diseases including cancer. Small nucleolar RNAs (snoRNAs), the most abundant group of intron-encoded ncRNAs, are classified into two families (box C/D snoRNAs and box H/ACA snoRNAs) and are required for post-transcriptional modifications on ribosomal RNA (rRNA). There is now a growing appreciation that nucleotide modifications on rRNA may impart regulatory potential to the ribosome; however, the functional consequence of site-specific snoRNA-guided modifications remains poorly defined. Discovered almost 20 years ago, H/ACA snoRNAs are required for the conversion of specific uridine residues to pseudouridine on rRNA. Interestingly, recent reports indicate that the levels of subsets of H/ACA snoRNAs required for pseudouridine modifications at specific sites on rRNA are altered in several diseases, particularly cancer. In this review, we describe recent advances in understanding the downstream consequences of H/ACA snoRNA-guided modifications on ribosome function, discuss the possible mechanism by which H/ACA snoRNAs may be regulated, and explore prospective expanding functions of H/ACA snoRNAs. Furthermore, we discuss the potential biological implications of alterations in H/ACA snoRNA expression in several human diseases.
Collapse
Affiliation(s)
- Mary McMahon
- School of Medicine and Department of Urology, Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
507
|
Avitabile C, Cimmino A, Romanelli A. Oligonucleotide analogues as modulators of the expression and function of noncoding RNAs (ncRNAs): emerging therapeutics applications. J Med Chem 2014; 57:10220-40. [PMID: 25280271 DOI: 10.1021/jm5006594] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
ncRNAs are emerging as key regulators of physiological and pathological processes and therefore have been identified as pharmacological targets and as markers for some diseases. Oligonucleotide analogues represent so far the most widely employed tool for the modulation of the expression of ncRNAs. In this perspective we briefly describe most of the known classes of ncRNAs and then we discuss the design and the applications of oligonucleotide analogues for their targeting. The effects of modifications of the chemical structure of the oligonucleotides on properties such as the binding affinity toward targets and off targets, and the stability to degradation and their biological effects (when known) are discussed. Examples of molecules currently used in clinical trials are also reported.
Collapse
Affiliation(s)
- Concetta Avitabile
- Università di Napoli "Federico II" , Dipartimento di Farmacia, via Mezzocannone 16, 80134 Napoli, Italy
| | | | | |
Collapse
|
508
|
Stachurska A, Zorro MM, van der Sijde MR, Withoff S. Small and Long Regulatory RNAs in the Immune System and Immune Diseases. Front Immunol 2014; 5:513. [PMID: 25368617 PMCID: PMC4202709 DOI: 10.3389/fimmu.2014.00513] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 10/02/2014] [Indexed: 12/14/2022] Open
Abstract
Cellular differentiation is regulated on the level of gene expression, and it is known that dysregulation of gene expression can lead to deficiencies in differentiation that contribute to a variety of diseases, particularly of the immune system. Until recently, it was thought that the dysregulation was governed by changes in the binding or activity of a class of proteins called transcription factors. However, the discovery of micro-RNAs and recent descriptions of long non-coding RNAs (lncRNAs) have given enormous momentum to a whole new field of biology: the regulatory RNAs. In this review, we describe these two classes of regulatory RNAs and summarize what is known about how they regulate aspects of the adaptive and innate immune systems. Finally, we describe what is known about the involvement of micro-RNAs and lncRNAs in three different autoimmune diseases (celiac disease, inflammatory bowel disease, and multiple sclerosis).
Collapse
Affiliation(s)
- Anna Stachurska
- Department of Genetics, University Medical Center Groningen, University of Groningen , Groningen , Netherlands
| | - Maria M Zorro
- Department of Genetics, University Medical Center Groningen, University of Groningen , Groningen , Netherlands
| | - Marijke R van der Sijde
- Department of Genetics, University Medical Center Groningen, University of Groningen , Groningen , Netherlands
| | - Sebo Withoff
- Department of Genetics, University Medical Center Groningen, University of Groningen , Groningen , Netherlands
| |
Collapse
|
509
|
Yang T, Liang Y, Lin Q, Liu J, Luo F, Li X, Zhou H, Zhuang S, Zhang H. miR-29 mediates TGFβ1-induced extracellular matrix synthesis through activation of PI3K-AKT pathway in human lung fibroblasts. J Cell Biochem 2014; 114:1336-42. [PMID: 23238947 DOI: 10.1002/jcb.24474] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 11/28/2012] [Indexed: 12/25/2022]
Abstract
TGFβ1 is very important in the synthesis and degradation of extracellular matrix, and also in the mediation of human lung fibroblasts proliferation, and miR-29 plays an important role in this process. To explore the interactions of miR-29 family members and TGFβ1, the effects of transforming growth factor TGFβ1 on the expression of miR-29 and whether miR-29 is involved in pro-survival signaling pathways mediated by TGFβ1 were examined in human lung fibroblasts. Treatment of the human embryonic lung fibroblast cell line IMR90 with TGFβ1 caused a decrease in expression of miR-29a/b/c by real-time PCR analysis. TGFβ1 stimulation increased cell proliferation, colony formation and up-regulated expression of COL1A1; transfecting with miR-29a/b/c mimics reverse TGFβ1-induced phenotype changes in IMR90 cells. Western blot analyses showed that TGFβ1 treatment unchanged total protein expression levels of PI3K or AKT, but the expression levels of p-PI3K, p-AKT, and COL1A1 were increased; and miR-19a/b/c mimics interfering blocked phosphorylation of PI3K or AKT and decreased expression of COL1A1 after TGFβ1 treatment. The results indicate that TGFβ1 beta uses the PI3k-Akt pathway in these embryonic fibroblasts and miR29 blocks this activation pathway. It indicates a novel biological function of the PI3K-Akt pathway in IMR90. Elevated expression of miR-29 may play an important role in the pathogenesis of diseases related to fibrogenic reactions in human lung fibroblasts.
Collapse
Affiliation(s)
- Tao Yang
- National Engineering Laboratory for Rice and By-product Deep Processing, Central South University of Forestry and Technology, Changsha 410004, Hunan Province, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
510
|
Dhahbi JM. Circulating small noncoding RNAs as biomarkers of aging. Ageing Res Rev 2014; 17:86-98. [PMID: 24607831 DOI: 10.1016/j.arr.2014.02.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/17/2014] [Accepted: 02/24/2014] [Indexed: 12/31/2022]
Abstract
Small noncoding RNAs (sncRNAs) mediate a variety of cellular functions in animals and plants. Deep sequencing has made it possible to obtain highly detailed information on the types and abundance of sncRNAs in biological specimens, leading to the discovery that sncRNAs circulate in the blood of humans and mammals. The most abundant types of circulating sncRNAs are microRNAs (miRNAs), 5' transfer RNA (tRNA) halves, and YRNA fragments, with minute amounts of other types that may nevertheless be significant. Of the more abundant circulating sncRNAs only miRNAs have well described functions, but characteristics of the others suggest specific processing and secretion as complexes that protect the RNA from degradation. The properties of circulating sncRNAs are consistent with their serving as signaling molecules, and investigations of circulating miRNAs support the view that they can enter cells and regulate cellular functions. The serum levels of specific sncRNAs change markedly with age, and these changes can be mitigated by calorie restriction (CR), indicating that levels are under physiologic control. The ability of circulating sncRNAs to transmit functions between cells and to regulate a broad spectrum of cellular functions, and the changes in their levels with age, implicate them in the manifestations of aging. Our understanding of the functions of circulating sncRNA, particularly in relation to aging, is currently at a very early stage; results to date suggest that more extensive investigation will yield important insights into mechanisms of aging.
Collapse
Affiliation(s)
- Joseph M Dhahbi
- Department of Biochemistry, University of California at Riverside, Riverside, CA 92521, USA; Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA.
| |
Collapse
|
511
|
Zhao M, Qiu Y, Yang B, Sun L, Hei K, Du X, Li Y. Long non-coding RNAs involved in gynecological cancer. Int J Gynecol Cancer 2014; 24:1140-5. [PMID: 25078335 DOI: 10.1097/igc.0000000000000212] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are defined as transcripts longer than 200 nucleotides with little or no protein-coding capacity. Previously, they were considered transcription byproducts without biological functions. Further studies have shown that lncRNAs are involved in multiple biological and pathological processes, including regulation of epigenetic, transcriptional, and posttranscriptional events. Long non-coding RNA expression patterns in various malignant tumors differ from those of benign tumors and normal tissue, and such alterations may promote or suppress tumorigenesis and cancer progression. The expression profiles of lncRNAs are abnormal in gynecological cancers, such as ovarian cancer, cervical cancer, and endometrial cancer, suggesting an important role for lncRNAs in tumorigenesis/progression of these cancers. Here, we summarized the research progress on identifying the biological functions of lncRNAs in tumorigenesis, progression, and metastasis in gynecological cancers. We provide references for exploring the clinical applications of lncRNAs as early diagnostic biomarkers or ideal therapeutic targets in gynecological cancers.
Collapse
Affiliation(s)
- Manyin Zhao
- *Department of Medical Microbiology, School of Basic Medical Sciences, †Second Affiliated Hospital, and ‡General Affiliated Hospital, Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
512
|
Toiyama Y, Okugawa Y, Goel A. DNA methylation and microRNA biomarkers for noninvasive detection of gastric and colorectal cancer. Biochem Biophys Res Commun 2014; 455:43-57. [PMID: 25128828 DOI: 10.1016/j.bbrc.2014.08.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 07/22/2014] [Accepted: 08/01/2014] [Indexed: 02/06/2023]
Abstract
Cancer initiation and progression is controlled by both genetic and epigenetic events. Epigenetics refers to the study of mechanisms that alter gene expression without permanently altering the DNA sequence. Epigenetic alterations are reversible and heritable, and include changes in histone modifications, DNA methylation, and non-coding RNA-mediated gene silencing. Disruption of epigenetic processes can lead to altered gene function and malignant cellular transformation. Aberrant epigenetic modifications occur at the earliest stages of neoplastic transformation and are now believed to be essential players in cancer initiation and progression. Recent advances in epigenetics have not only offered a deeper understanding of the underlying mechanism(s) of carcinogenesis, but have also allowed identification of clinically relevant putative biomarkers for the early detection, disease monitoring, prognosis and risk assessment of cancer patients. At this moment, DNA methylation and non-coding RNA including with microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) represent the largest body of available literature on epigenetic biomarkers with the highest potential for cancer diagnosis. Following identification of cell-free nucleic acids in systematic circulation, increasing evidence has demonstrated the potential of cell-free epigenetic biomarkers in the blood or other body fluids for cancer detection. In this article, we summarize the current state of knowledge on epigenetic biomarkers - primarily DNA methylation and non-coding RNAs - as potential substrates for cancer detection in gastric and colorectal cancer, the two most frequent cancers within the gastrointestinal tract. We also discuss the obstacles that have limited the routine use of epigenetic biomarkers in the clinical settings and provide our perspective on approaches that might help overcome these hurdles, so that these biomarkers can be readily developed for clinical management of cancer patients.
Collapse
Affiliation(s)
- Yuji Toiyama
- Gastrointestinal Cancer Research Laboratory, Department of Internal Medicine, Charles A. Sammons Cancer Center and Baylor Research Institute, Baylor University Medical Center, 3500 Gaston Avenue, Dallas, TX 75246, USA; Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Graduate School of Medicine, Mie University, Mie 514-8507, Japan
| | - Yoshinaga Okugawa
- Gastrointestinal Cancer Research Laboratory, Department of Internal Medicine, Charles A. Sammons Cancer Center and Baylor Research Institute, Baylor University Medical Center, 3500 Gaston Avenue, Dallas, TX 75246, USA; Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Graduate School of Medicine, Mie University, Mie 514-8507, Japan
| | - Ajay Goel
- Gastrointestinal Cancer Research Laboratory, Department of Internal Medicine, Charles A. Sammons Cancer Center and Baylor Research Institute, Baylor University Medical Center, 3500 Gaston Avenue, Dallas, TX 75246, USA.
| |
Collapse
|
513
|
Lipid nanoparticles as carriers for RNAi against viral infections: current status and future perspectives. BIOMED RESEARCH INTERNATIONAL 2014; 2014:161794. [PMID: 25184135 PMCID: PMC4145386 DOI: 10.1155/2014/161794] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 07/14/2014] [Accepted: 07/14/2014] [Indexed: 12/15/2022]
Abstract
The efforts made to develop RNAi-based therapies have led to productive research in the field of infections in humans, such as hepatitis C virus (HCV), hepatitis B virus (HBV), human immunodeficiency virus (HIV), human cytomegalovirus (HCMV), herpetic keratitis, human papillomavirus, or influenza virus. Naked RNAi molecules are rapidly digested by nucleases in the serum, and due to their negative surface charge, entry into the cell cytoplasm is also hampered, which makes necessary the use of delivery systems to exploit the full potential of RNAi therapeutics. Lipid nanoparticles (LNP) represent one of the most widely used delivery systems for in vivo application of RNAi due to their relative safety and simplicity of production, joint with the enhanced payload and protection of encapsulated RNAs. Moreover, LNP may be functionalized to reach target cells, and they may be used to combine RNAi molecules with conventional drug substances to reduce resistance or improve efficiency. This review features the current application of LNP in RNAi mediated therapy against viral infections and aims to explore possible future lines of action in this field.
Collapse
|
514
|
Zhao Y, Bhattacharjee S, Jones BM, Hill J, Dua P, Lukiw WJ. Regulation of neurotropic signaling by the inducible, NF-kB-sensitive miRNA-125b in Alzheimer's disease (AD) and in primary human neuronal-glial (HNG) cells. Mol Neurobiol 2014; 50:97-106. [PMID: 24293102 PMCID: PMC4038663 DOI: 10.1007/s12035-013-8595-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 11/13/2013] [Indexed: 12/12/2022]
Abstract
Inducible microRNAs (miRNAs) perform critical regulatory roles in central nervous system (CNS) development, aging, health, and disease. Using miRNA arrays, RNA sequencing, enhanced Northern dot blot hybridization technologies, Western immunoblot, and bioinformatics analysis, we have studied miRNA abundance and complexity in Alzheimer's disease (AD) brain tissues compared to age-matched controls. In both short post-mortem AD and in stressed primary human neuronal-glial (HNG) cells, we observe a consistent up-regulation of several brain-enriched miRNAs that are under transcriptional control by the pro-inflammatory transcription factor NF-kB. These include miRNA-9, miRNA-34a, miRNA-125b, miRNA-146a, and miRNA-155. Of the inducible miRNAs in this subfamily, miRNA-125b is among the most abundant and significantly induced miRNA species in human brain cells and tissues. Bioinformatics analysis indicated that an up-regulated miRNA-125b could potentially target the 3'untranslated region (3'-UTR) of the messenger RNA (mRNA) encoding (a) a 15-lipoxygenase (15-LOX; ALOX15; chr 17p13.3), utilized in the conversion of docosahexaneoic acid into neuroprotectin D1 (NPD1), and (b) the vitamin D3 receptor (VDR; VD3R; chr12q13.11) of the nuclear hormone receptor superfamily. 15-LOX and VDR are key neuromolecular factors essential in lipid-mediated signaling, neurotrophic support, defense against reactive oxygen and nitrogen species (reactive oxygen and nitrogen species), and neuroprotection in the CNS. Pathogenic effects appear to be mediated via specific interaction of miRNA-125b with the 3'-UTR region of the 15-LOX and VDR messenger RNAs (mRNAs). In AD hippocampal CA1 and in stressed HNG cells, 15-LOX and VDR down-regulation and a deficiency in neurotrophic support may therefore be explained by the actions of a single inducible, pro-inflammatory miRNA-125b. We will review the recent data on the pathogenic actions of this up-regulated miRNA-125b in AD and discuss potential therapeutic approaches using either anti-NF-kB or anti-miRNA-125b strategies. These may be of clinical relevance in the restoration of 15-LOX and VDR expression back to control levels and the re-establishment of homeostatic neurotrophic signaling in the CNS.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
| | - Surjyadipta Bhattacharjee
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
| | - Brandon M. Jones
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
| | - Jim Hill
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
- Department of Microbiology, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
| | - Prerna Dua
- Department of Health Information Management, Louisiana State University, Ruston, LA 71272 USA
| | - Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
- Department of Neurology, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
| |
Collapse
|
515
|
|
516
|
Kawamura M, Toiyama Y, Tanaka K, Inoue Y, Mohri Y, Kusunoki M. Can Circulating MicroRNAs Become the Test of Choice for Colorectal Cancer? CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0240-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
517
|
Blood born miRNAs signatures that can serve as disease specific biomarkers are not significantly affected by overall fitness and exercise. PLoS One 2014; 9:e102183. [PMID: 25010680 PMCID: PMC4092108 DOI: 10.1371/journal.pone.0102183] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/16/2014] [Indexed: 12/17/2022] Open
Abstract
Blood born micro(mi)RNA expression pattern have been reported for various human diseases with signatures specific for diseases. To evaluate these biomarkers, it is mandatory to know possible changes of miRNA signatures in healthy individuals under different physiological conditions. We analyzed the miRNA expression in peripheral blood of elite endurance athletes and moderatly active controls. Blood drawing was done before and after exhaustive exercise in each group. After Benjamini-Hochberg adjustment we did not find any miRNA with significant p-values when comparing miRNA expression between the different groups. We found, however, 24 different miRNAs with an expression fold change of minimum 1.5 in at least one of the comparisons (athletes before vs after exercise, athletes before exercise vs controls and athletes after exercise vs controls). The observed changes are not significant in contrast to the expression changes of the blood born miRNA expression reported for many human diseases. These data support the idea of disease associated miRNA patterns useful as biomarkers that are not readily altered by physiological conditions.
Collapse
|
518
|
Abstract
Diabetic nephropathy (DN), a severe microvascular complication frequently associated with both type 1 and type 2 diabetes mellitus, is a leading cause of renal failure. The condition can also lead to accelerated cardiovascular disease and macrovascular complications. Currently available therapies have not been fully efficacious in the treatment of DN, suggesting that further understanding of the molecular mechanisms underlying the pathogenesis of DN is necessary for the improved management of this disease. Although key signal transduction and gene regulation mechanisms have been identified, especially those related to the effects of hyperglycaemia, transforming growth factor β1 and angiotensin II, progress in functional genomics, high-throughput sequencing technology, epigenetics and systems biology approaches have greatly expanded our knowledge and uncovered new molecular mechanisms and factors involved in DN. These mechanisms include DNA methylation, chromatin histone modifications, novel transcripts and functional noncoding RNAs, such as microRNAs and long noncoding RNAs. In this Review, we discuss the significance of these emerging mechanisms, how they mediate the actions of growth factors to augment the expression of extracellular matrix and inflammatory genes associated with DN and their potential usefulness as diagnostic biomarkers or novel therapeutic targets for DN.
Collapse
Affiliation(s)
- Mitsuo Kato
- Department of Diabetes, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Rama Natarajan
- Department of Diabetes, Beckman Research Institute of City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
519
|
Zhao X, Zhu W, Zha W, Chen F, Wu Z, Liu Y, Huang M. Expression profiles and initial confirmation of long noncoding RNAs in Chinese patients with pulmonary adenocarcinoma. Onco Targets Ther 2014; 7:1195-204. [PMID: 25061321 PMCID: PMC4085304 DOI: 10.2147/ott.s64033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background The purpose of this study was to investigate differentially expressed long noncoding RNAs (lncRNAs) in pulmonary adenocarcinoma tissue and adjacent noncancerous tissue from Chinese patients using lncRNA expression microarray and preliminary analysis. Methods RNA extracted from three paired pulmonary adenocarcinoma tissue and adjacent noncancerous tissue specimens was used to synthesize double-stranded complementary DNA after labeling and hybridization. The complementary DNA was labeled and hybridized to the lncRNA expression microarray, and array data were analyzed for hierarchical clustering. Gene coexpression networks were constructed to identify interactions among genes. To validate the microarray findings, we measured the relative expression levels of four random differentially expressed lncRNAs in the same tissue used for microarray using real-time quantitative polymerase chain reaction. The expression level of one lncRNA, AK124939, in the paired pulmonary adenocarcinoma/adjacent noncancerous tissue of another 30 patients was measured using real-time quantitative polymerase chain reaction. The experimental data were further analyzed and compared with clinical features. Results Of 39,000 lncRNAs investigated, 704 were differentially expressed in pulmonary adenocarcinoma tissue; 385 were upregulated and 319 were downregulated compared with those in the adjacent noncancerous tissue (fold change ≥2 and ≤−2, P<0.05). AK124939 expression levels in poorly differentiated adenocarcinoma tissue were lower than those found in well to moderately differentiated adenocarcinoma tissue (P=0.05). Conclusion There are significant differences in the lncRNA expression profiles in Chinese patients with pulmonary adenocarcinoma. LncRNAs such as AK124939 may be anticancer factors related to the progression of pulmonary adenocarcinoma.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Wen Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Wangjian Zha
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Feifei Chen
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhenzhen Wu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yanan Liu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Mao Huang
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
520
|
Scalabrin M, Siu Y, Asare-Okai PN, Fabris D. Structure-specific ribonucleases for MS-based elucidation of higher-order RNA structure. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2014; 25:1136-1145. [PMID: 24845355 PMCID: PMC6911265 DOI: 10.1007/s13361-014-0911-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/08/2014] [Accepted: 04/08/2014] [Indexed: 06/03/2023]
Abstract
Supported by high-throughput sequencing technologies, structure-specific nucleases are experiencing a renaissance as biochemical probes for genome-wide mapping of nucleic acid structure. This report explores the benefits and pitfalls of the application of Mung bean (Mb) and V1 nuclease, which attack specifically single- and double-stranded regions of nucleic acids, as possible structural probes to be employed in combination with MS detection. Both enzymes were found capable of operating in ammonium-based solutions that are preferred for high-resolution analysis by direct infusion electrospray ionization (ESI). Sequence analysis by tandem mass spectrometry (MS/MS) was performed to confirm mapping assignments and to resolve possible ambiguities arising from the concomitant formation of isobaric products with identical base composition and different sequences. The observed products grouped together into ladder-type series that facilitated their assignment to unique regions of the substrate, but revealed also a certain level of uncertainty in identifying the boundaries between paired and unpaired regions. Various experimental factors that are known to stabilize nucleic acid structure, such as higher ionic strength, presence of Mg(II), etc., increased the accuracy of cleavage information, but did not completely eliminate deviations from expected results. These observations suggest extreme caution in interpreting the results afforded by these types of reagents. Regardless of the analytical platform of choice, the results highlighted the need to repeat probing experiments under the most diverse possible conditions to recognize potential artifacts and to increase the level of confidence in the observed structural information.
Collapse
Affiliation(s)
- Matteo Scalabrin
- The RNA Institute, University at Albany-SUNY, Albany, NY, 12222, USA
| | | | | | | |
Collapse
|
521
|
Lee H, Kim C, Ku JL, Kim W, Yoon SK, Kuh HJ, Lee JH, Nam SW, Lee EK. A long non-coding RNA snaR contributes to 5-fluorouracil resistance in human colon cancer cells. Mol Cells 2014; 37:540-6. [PMID: 25078450 PMCID: PMC4132306 DOI: 10.14348/molcells.2014.0151] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 06/30/2014] [Accepted: 06/30/2014] [Indexed: 12/11/2022] Open
Abstract
Several types of genetic and epigenetic regulation have been implicated in the development of drug resistance, one significant challenge for cancer therapy. Although changes in the expression of non-coding RNA are also responsible for drug resistance, the specific identities and roles of them remain to be elucidated. Long non-coding RNAs (lncRNAs) are a type of ncRNA (> 200 nt) that influence the regulation of gene expression in various ways. In this study, we aimed to identify differentially expressed lncRNAs in 5-fluorouracil-resistant colon cancer cells. Using two pairs of 5-FU-resistant cells derived from the human colon cancer cell lines SNU-C4 and SNU-C5, we analyzed the expression of 90 lncRNAs by qPCR-based profiling and found that 19 and 23 lncRNAs were differentially expressed in SNU-C4R and SNU-C5R cells, respectively. We confirmed that snaR and BACE1AS were downregulated in resistant cells. To further investigate the effects of snaR on cell growth, cell viability and cell cycle were analyzed after transfection of siRNAs targeting snaR. Down-regulation of snaR decreased cell death after 5-FU treatment, which indicates that snaR loss decreases in vitro sensitivity to 5-FU. Our results provide an important insight into the involvement of lncRNAs in 5-FU resistance in colon cancer cells.
Collapse
Affiliation(s)
- Heejin Lee
- Department of Biochemistry, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Chongtae Kim
- Department of Biochemistry, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Ja-Lok Ku
- Cancer Research Institute and Cancer Research Center, Seoul National University, Seoul 110-744, Korea
| | - Wook Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| | - Sungjoo Kim Yoon
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
- Department of Biomedical Science, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Hyo-Jeong Kuh
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
- Department of Biomedical Science, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
| | - Suk Woo Nam
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
- Department of Pathology, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
| |
Collapse
|
522
|
Faulkner S, Elia G, O' Boyle P, Dunn M, Morris D. Composition of the bovine uterine proteome is associated with stage of cycle and concentration of systemic progesterone. Proteomics 2014; 13:3333-53. [PMID: 24115321 DOI: 10.1002/pmic.201300204] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/24/2013] [Accepted: 09/11/2013] [Indexed: 11/08/2022]
Abstract
Early embryonic loss accounts for over 70% of total embryonic and foetal loss in dairy cattle. Early embryonic development and survival is associated with the concentration of systemic progesterone. To determine if the uterine proteome is influenced by stage of cycle or systemic progesterone concentrations, uterine flushings were collected from the ipsi- and contralateral uterine horns of beef heifers on Days 7 (n = 10) and 15 (n = 10) of the oestrous cycle. Animals were separated into low or high progesterone groups based on plasma progesterone concentrations on Day 5 of the cycle. Samples were albumin depleted before iTRAQ R labeling and subsequent strong cation exchange-LC-MS/MS analyses. A total of 20 proteins were up to 5.9-fold higher (p<0.05) and 20 were up to 2.3-fold lower on Day 15 compared toDay 7. In addition, the expression of a number of proteins on Day 7 and/or 15 of the cycle was correlated with progesterone concentrations during Days 3–7 or the rate of change in progesterone between Days 3 and 7. This study highlights the dynamic changes occurring in the microenvironment surrounding the embryo during this period. The findings here also support the hypothesis that progesterone supports embryonic development by altering the maternal uterine environment.
Collapse
|
523
|
Comparisons of microRNA expression profiles in vitreous humor between eyes with macular hole and eyes with proliferative diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2014; 253:335-42. [PMID: 24970617 DOI: 10.1007/s00417-014-2692-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 06/02/2014] [Accepted: 06/03/2014] [Indexed: 10/25/2022] Open
Abstract
PURPOSE MicroRNAs (miRNAs) are small noncoding RNAs which regulate the activities of target mRNAs. We compared the expression profiles of the miRNAs in the vitreous of eyes with macular hole (MH) to that in eyes with proliferative diabetic retinopathy (PDR). METHODS Vitreous and whole blood samples were collected from four patients with MH and from four patients with PDR. We assayed for 168 miRNAs in the vitreous and serum samples by the microRNA PCR Panel method. RESULTS The mean number of miRNAs expressed in the vitreous was 63 (55-69) in eyes with MH and 86 (65-117) in eyes with PDR. The mean number of miRNAs expressed in the serum was 162 (159-167) in the MH patients and 142 (115-160) in the PDR patients. Twenty-six miRNAs were expressed in the vitreous of both MH and PDR eyes. Although there was no significant difference in the levels of 20 of the 26 (73 %) miRNAs expressed in both MH and PDR eyes, six of 26 miRNAs (24 %) (hsa-miR-15a, hsa-miR320a, hsa-miR-320b, hsa-miR-93, hsa-miR-29a, and hsa-miR-423-5p) were expressed significantly more highly in PDR eyes. In addition, the mean fold changes of three miRNAs, hsa-miR-23a, hsa-miR-320a, and hsa-miR-320b, in the vitreous to serum were significantly higher in the PDR group than in the MH group. CONCLUSIONS The expression of several miRNAs related to angiogenesis and fibrosis was expressed significantly higher in the vitreous of eyes with PDR. Further studies are needed to understand the role played by the miRNAs in the biological function of the eye.
Collapse
|
524
|
Liu PY, Erriquez D, Marshall GM, Tee AE, Polly P, Wong M, Liu B, Bell JL, Zhang XD, Milazzo G, Cheung BB, Fox A, Swarbrick A, Hüttelmaier S, Kavallaris M, Perini G, Mattick JS, Dinger ME, Liu T. Effects of a novel long noncoding RNA, lncUSMycN, on N-Myc expression and neuroblastoma progression. J Natl Cancer Inst 2014; 106:dju113. [PMID: 24906397 DOI: 10.1093/jnci/dju113] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Patients with neuroblastoma due to the amplification of a 130-kb genomic DNA region containing the MYCN oncogene have poor prognoses. METHODS Bioinformatics data were used to discover a novel long noncoding RNA, lncUSMycN, at the 130-kb amplicon. RNA-protein pull-down assays were used to identify proteins bound to lncUSMycN RNA. Kaplan-Meier survival analysis, multivariable Cox regression, and two-sided log-rank test were used to examine the prognostic value of lncUSMycN and NonO expression in three cohorts of neuroblastoma patients (n = 47, 88, and 476, respectively). Neuroblastoma-bearing mice were treated with antisense oligonucleotides targeting lncUSMycN (n = 12) or mismatch sequence (n = 13), and results were analyzed by multiple comparison two-way analysis of variance. All statistical tests were two-sided. RESULTS Bioinformatics data predicted lncUSMycN gene and RNA, and reverse-transcription polymerase chain reaction confirmed its three exons and two introns. The lncUSMycN gene was coamplified with MYCN in 88 of 341 human neuroblastoma tissues. lncUSMycN RNA bound to the RNA-binding protein NonO, leading to N-Myc RNA upregulation and neuroblastoma cell proliferation. High levels of lncUSMycN and NonO expression in human neuroblastoma tissues independently predicted poor patient prognoses (lncUSMycN: hazard ratio [HR] = 1.87, 95% confidence interval [CI] = 1.06 to 3.28, P = .03; NonO: HR = 2.48, 95% CI = 1.34 to 4.57, P = .004). Treatment with antisense oligonucleotides targeting lncUSMycN in neuroblastoma-bearing mice statistically significantly hindered tumor progression (P < .001). CONCLUSIONS Our data demonstrate the important roles of lncUSMycN and NonO in regulating N-Myc expression and neuroblastoma oncogenesis and provide the first evidence that amplification of long noncoding RNA genes can contribute to tumorigenesis.
Collapse
Affiliation(s)
- Pei Y Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Daniela Erriquez
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Glenn M Marshall
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Andrew E Tee
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Patsie Polly
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Mathew Wong
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Bing Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Jessica L Bell
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Xu D Zhang
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Giorgio Milazzo
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Belamy B Cheung
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Archa Fox
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Alexander Swarbrick
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Stefan Hüttelmaier
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Maria Kavallaris
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Giovanni Perini
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - John S Mattick
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Marcel E Dinger
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Tao Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL).
| |
Collapse
|
525
|
Eliasson L, Esguerra JLS. Role of non-coding RNAs in pancreatic beta-cell development and physiology. Acta Physiol (Oxf) 2014; 211:273-84. [PMID: 24666639 DOI: 10.1111/apha.12285] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 02/25/2014] [Accepted: 03/17/2014] [Indexed: 12/15/2022]
Abstract
The progression of diabetes is accompanied by increasing demand to the beta-cells to produce and secrete more insulin, requiring complex beta-cell adaptations. Functionally active and ubiquitous non-coding RNAs (ncRNAs) have the capacity to take part in such adaptations as they have been shown to be key regulatory molecules in various biological processes. In the pancreatic islets, the function of ncRNAs and their contribution to disease development is beginning to be understood. Here, we review the different classes of ncRNAs, such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), and their potential contribution to insulin secretion. A special focus will be on miRNAs and their regulatory function in beta-cell physiology and insulin exocytosis. As important players in gene regulation, ncRNAs have huge potential in opening innovative therapeutic avenues against diabetes and associated complications.
Collapse
Affiliation(s)
- L. Eliasson
- Department of Clinical Sciences-Malmö; Islet Cell Exocytosis; Lund University Diabetes Centre; Lund University; Malmö Sweden
| | - J. L. S. Esguerra
- Department of Clinical Sciences-Malmö; Islet Cell Exocytosis; Lund University Diabetes Centre; Lund University; Malmö Sweden
| |
Collapse
|
526
|
Gottlieb B, Beitel LK, Trifiro M. Changing genetic paradigms: creating next-generation genetic databases as tools to understand the emerging complexities of genotype/phenotype relationships. Hum Genomics 2014; 8:9. [PMID: 24885908 PMCID: PMC4040485 DOI: 10.1186/1479-7364-8-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/25/2014] [Indexed: 12/18/2022] Open
Abstract
Understanding genotype/phenotype relationships has become more complicated as increasing amounts of inter- and intra-tissue genetic heterogeneity have been revealed through next-generation sequencing and evidence showing that factors such as epigenetic modifications, non-coding RNAs and RNA editing can play an important role in determining phenotype. Such findings have challenged a number of classic genetic assumptions including (i) analysis of genomic sequence obtained from blood is an accurate reflection of the genotype responsible for phenotype expression in an individual; (ii) that significant genetic alterations will be found only in diseased individuals, in germline tissues in inherited diseases, or in specific diseased tissues in somatic diseases such as cancer; and (iii) that mutation rates in putative disease-associated genes solely determine disease phenotypes. With the breakdown of our traditional understanding of genotype to phenotype relationships, it is becoming increasingly apparent that new analytical tools will be required to determine the relationship between genotype and phenotypic expression. To this end, we are proposing that next-generation genetic database (NGDB) platforms be created that include new bioinformatics tools based on algorithms that can evaluate genetic heterogeneity, as well as powerful systems biology analysis tools to actively process and evaluate the vast amounts of both genomic and genomic-modifying information required to reveal the true relationships between genotype and phenotype.
Collapse
Affiliation(s)
- Bruce Gottlieb
- Lady Davis Institute for Medical Research, 3755 Côte Ste Catherine Road, Montreal, QC H3T 1E2, Canada
- Segal Cancer Centre, Jewish General Hospital, 3755 Côte Ste Catherine Road, Montreal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Lenore K Beitel
- Lady Davis Institute for Medical Research, 3755 Côte Ste Catherine Road, Montreal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Mark Trifiro
- Lady Davis Institute for Medical Research, 3755 Côte Ste Catherine Road, Montreal, QC H3T 1E2, Canada
- Segal Cancer Centre, Jewish General Hospital, 3755 Côte Ste Catherine Road, Montreal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
527
|
Dong L, Li Y, Han C, Wang X, She L, Zhang H. miRNA microarray reveals specific expression in the peripheral blood of glioblastoma patients. Int J Oncol 2014; 45:746-56. [PMID: 24858071 DOI: 10.3892/ijo.2014.2459] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/17/2014] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are frequently dysregulated in glioblastoma (GBM) patients. It has been discovered that highly stable extracellular miRNAs circulate in the blood of both healthy individuals and patients. miRNAs in serum of patients with GBM and normal controls were analyzed by microarray analysis. The relevant bioinformatic analysis of the predicted target genes (gene ontology, pathway, gene network analysis) were performed. The miRNA microarray reveals differentially expressed miRNAs in serum between the GBM and normal controls. Of the 752 miRNAs, 115 miRNAs were upregulated in the GBM group, and 24 miRNAs were downregulated (fold change ≥2.0, P<0.01). By further analysis, we found that miR-576-5p, miR-340 and miR-626 were significantly overexpressed, but miR-320, let-7g-5p and miR-7-5P showed significantly low expression in GBM patients. By further bioinformatic analysis, we found that they possibly play important roles in the regulation of glioma signaling pathways. In summary, the six miRNAs are significant distinct in the peripheral blood of patients with GBM pathologies. These data suggest that the miRNA profile of the peripheral blood may serve as a new biomarker for glioma diagnosis with high specificity and sensitivity.
Collapse
Affiliation(s)
- Lun Dong
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Yuping Li
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Chongxu Han
- Department of Central Laboratory, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Xiaodong Wang
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Lei She
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Hengzhu Zhang
- Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
528
|
Perry MM, Tsitsiou E, Austin PJ, Lindsay MA, Gibeon DS, Adcock IM, Chung KF. Role of non-coding RNAs in maintaining primary airway smooth muscle cells. Respir Res 2014; 15:58. [PMID: 24886442 PMCID: PMC4039655 DOI: 10.1186/1465-9921-15-58] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/28/2014] [Indexed: 12/16/2022] Open
Abstract
Background The airway smooth muscle (ASM) cell maintains its own proliferative rate and contributes to the inflammatory response in the airways, effects that are inhibited by corticosteroids, used in the treatment of airways diseases. Objective We determined the differential expression of mRNAs, microRNAs (miRNAs) and long noncoding RNA species (lncRNAs) in primary ASM cells following treatment with a corticosteroid, dexamethasone, and fetal calf serum (FCS). Methods mRNA, miRNA and lncRNA expression was measured by microarray and quantitative real-time PCR. Results A small number of miRNAs (including miR-150, −371-5p, −718, −940, −1181, −1207-5p, −1915, and −3663-3p) were decreased following exposure to dexamethasone and FCS. The mRNA targets of these miRNAs were increased in expression. The changes in mRNA expression were associated with regulation of ASM actin cytoskeleton. We also observed changes in expression of lncRNAs, including natural antisense, pseudogenes, intronic lncRNAs, and intergenic lncRNAs following dexamethasone and FCS. We confirmed the change in expression of three of these, LINC00882, LINC00883, PVT1, and its transcriptional activator, c-MYC. We propose that four of these lincRNAs (RP11-46A10.4, LINC00883, BCYRN1, and LINC00882) act as miRNA ‘sponges’ for 4 miRNAs (miR-150, −371-5p, −940, −1207-5p). Conclusion This in-vitro model of primary ASM cell phenotype was associated with the regulation of several ncRNAs. Their identification allows for in-vitro functional experimentation to establish causality with the primary ASM phenotype, and in airway diseases such as asthma and chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Mark M Perry
- Experimental Studies, National Heart and Lung Institute, Imperial College, London & Royal Brompton NIHR Biomedical Research Unit, Dovehouse Street, London SW3 6LY, UK.
| | | | | | | | | | | | | |
Collapse
|
529
|
Miranda KC, Bond DT, Levin JZ, Adiconis X, Sivachenko A, Russ C, Brown D, Nusbaum C, Russo LM. Massively parallel sequencing of human urinary exosome/microvesicle RNA reveals a predominance of non-coding RNA. PLoS One 2014; 9:e96094. [PMID: 24816817 PMCID: PMC4015934 DOI: 10.1371/journal.pone.0096094] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 04/03/2014] [Indexed: 11/22/2022] Open
Abstract
Intact RNA from exosomes/microvesicles (collectively referred to as microvesicles) has sparked much interest as potential biomarkers for the non-invasive analysis of disease. Here we use the Illumina Genome Analyzer to determine the comprehensive array of nucleic acid reads present in urinary microvesicles. Extraneous nucleic acids were digested using RNase and DNase treatment and the microvesicle inner nucleic acid cargo was analyzed with and without DNase digestion to examine both DNA and RNA sequences contained in microvesicles. Results revealed that a substantial proportion (∼87%) of reads aligned to ribosomal RNA. Of the non-ribosomal RNA sequences, ∼60% aligned to non-coding RNA and repeat sequences including LINE, SINE, satellite repeats, and RNA repeats (tRNA, snRNA, scRNA and srpRNA). The remaining ∼40% of non-ribosomal RNA reads aligned to protein coding genes and splice sites encompassing approximately 13,500 of the known 21,892 protein coding genes of the human genome. Analysis of protein coding genes specific to the renal and genitourinary tract revealed that complete segments of the renal nephron and collecting duct as well as genes indicative of the bladder and prostate could be identified. This study reveals that the entire genitourinary system may be mapped using microvesicle transcript analysis and that the majority of non-ribosomal RNA sequences contained in microvesicles is potentially functional non-coding RNA, which play an emerging role in cell regulation.
Collapse
Affiliation(s)
- Kevin C. Miranda
- Program in Membrane Biology, Division of Nephrology & Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel T. Bond
- Program in Membrane Biology, Division of Nephrology & Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joshua Z. Levin
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
| | - Xian Adiconis
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
| | - Andrey Sivachenko
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
| | - Carsten Russ
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
| | - Dennis Brown
- Program in Membrane Biology, Division of Nephrology & Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Chad Nusbaum
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
| | - Leileata M. Russo
- Program in Membrane Biology, Division of Nephrology & Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
530
|
Differential expression of long noncoding RNA in primary and recurrent nasopharyngeal carcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:404567. [PMID: 24822202 PMCID: PMC4009106 DOI: 10.1155/2014/404567] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/20/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Recent studies suggested that non-protein-coding genes are implicated in the tumorigenic process of nasopharyngeal carcinoma (NPC). In the present study, we aimed to identify the differentially expressed long noncoding RNA (lncRNA) using data available in the public domain. METHODS Microarray data set GSE12452 was reannotated with ncFANs. Real-time quantitative PCR was used to quantify and validate the identified lncRNAs in NPC. RESULTS In primary NPC, upregulation of lnc-C22orf32-1, lnc-AL355149.1-1, and lnc-ZNF674-1 was observed. High levels of lnc-C22orf32-1 and lnc-AL355149.1-1 were significantly associated with the male patients. In addition, increased expression of lnc-C22orf32-1 and lnc-ZNF674-1 was associated with advanced tumor stages. Recurrent NPC displayed a distinctive lncRNA expression pattern. lnc-BCL2L11-3 was significantly increased in the recurrent NPC tissues. In addition, significant reduction of lnc-AL355149.1-1 and lnc-ZNF674-1 was observed in the recurrent NPC tissues. CONCLUSIONS Our results demonstrated that it is feasible to identify the differentially expressed lncRNA in the microarray dataset by functional reannotation. The association of lncRNA with gender and tumor size implicated that lncRNA possibly plays a part in the pathogenesis of primary NPC. Further, the distinctive lncRNA identified in the recurrent NPC may reveal a distinctive development mechanism underlying tumor recurrence.
Collapse
|
531
|
Pogue AI, Hill JM, Lukiw WJ. MicroRNA (miRNA): sequence and stability, viroid-like properties, and disease association in the CNS. Brain Res 2014; 1584:73-9. [PMID: 24709119 DOI: 10.1016/j.brainres.2014.03.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 03/06/2014] [Accepted: 03/28/2014] [Indexed: 12/29/2022]
Abstract
MicroRNAs (miRNAs) constitute a relatively recently-discovered class of small non-coding RNAs (sncRNAs) that are gaining considerable attention in the molecular-genetic regulatory mechanisms that contribute to human health and disease. As highly soluble and mobile entities, emerging evidence indicates that miRNAs posess a highly selected ribonucleotide sequence structure, are part of an evolutionary ancient genetic signaling system, resemble the plant pathogens known as viroids in their structure, mode of generation and function, and are very abundant in the physiological fluids that surround cells and tissues. Persistence and altered abundance of miRNAs in the extracellular fluid (ECF) or cerebrospinal fluid (CSF) may play a role in the intercellular spreading of disease systemically, and throughout functionally-linked cellular and tissue systems such as the central nervous system (CNS). This short communication will review some of the more fascinating features of these highly structured single stranded RNAs (ssRNAs) with emphasis on their presence and function in the human CNS, with particular reference to Alzheimer׳s disease (AD) wherever possible.
Collapse
Affiliation(s)
| | - James M Hill
- Departments of Neurology, Neuroscience and Ophthalmology, LSU Neuroscience Center, School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA 70112, USA
| | - Walter J Lukiw
- Alchem Biotek, Toronto, ON, Canada, M5S 1A8; Departments of Neurology, Neuroscience and Ophthalmology, LSU Neuroscience Center, School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA 70112, USA.
| |
Collapse
|
532
|
Yin KJ, Hamblin M, Chen YE. Non-coding RNAs in cerebral endothelial pathophysiology: emerging roles in stroke. Neurochem Int 2014; 77:9-16. [PMID: 24704794 DOI: 10.1016/j.neuint.2014.03.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/21/2014] [Accepted: 03/24/2014] [Indexed: 12/20/2022]
Abstract
Cerebral vascular endothelial cells form the major element of the blood-brain barrier (BBB) and constitute the primary interface between circulating blood and brain parenchyma. The structural and functional changes in cerebral endothelium during cerebral ischemia are well known to result in BBB disruption, vascular inflammation, edema, and angiogenesis. These complex pathological processes directly contribute to brain infarction, neurological deficits, and post-stroke neurovascular remodeling. Ischemic endothelial dysfunction appears to be tightly controlled by multiple gene signaling networks. Non-coding RNAs (ncRNAs) are functional RNA molecules that are generally not translated into proteins but can actively regulate the expression and function of many thousands of protein-coding genes by different mechanisms. Various classes of ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), small nucleolar RNAs (snoRNAs) and piwi-interacting RNAs (piRNAs), are highly expressed in the cerebrovascular endothelium where they serve as critical mediators to maintain normal cerebral vascular functions. Dysregulation of ncRNA activities has been closely linked to the pathophysiology of cerebral vascular endothelium and neurologic functional disorders in the brain's response to ischemic stimuli. In this review, we summarize recent advancements of these ncRNA mediators in the brain vasculature, highlighting the specific roles of endothelial miRNAs in stroke.
Collapse
Affiliation(s)
- Ke-Jie Yin
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| | - Milton Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue SL83, New Orleans, LA 70112, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| |
Collapse
|
533
|
Barry G. Integrating the roles of long and small non-coding RNA in brain function and disease. Mol Psychiatry 2014; 19:410-6. [PMID: 24468823 DOI: 10.1038/mp.2013.196] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/12/2013] [Accepted: 12/16/2013] [Indexed: 12/20/2022]
Abstract
Regulatory RNA is emerging as the major architect of cognitive evolution and innovation in the mammalian brain. While the protein machinery has remained largely constant throughout animal evolution, the non protein-coding transcriptome has expanded considerably to provide essential and widespread cellular regulation, partly through directing generic protein function. Both long (long non-coding RNA) and small non-coding RNAs (for example, microRNA) have been demonstrated to be essential for brain development and higher cognitive abilities, and to be involved in psychiatric disease. Long non-coding RNAs, highly expressed in the brain and expanded in mammalian genomes, provide tissue- and activity-specific epigenetic and transcriptional regulation, partly through functional control of evolutionary conserved effector small RNA activity. However, increased cognitive sophistication has likely introduced concomitant psychiatric vulnerabilities, predisposing to conditions such as autism and schizophrenia, and cooperation between regulatory and effector RNAs may underlie neural complexity and concomitant fragility in the human brain.
Collapse
Affiliation(s)
- G Barry
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| |
Collapse
|
534
|
Xue M, Li X, Wu W, Zhang S, Wu S, Li Z, Chen W. Upregulation of long non-coding RNA urothelial carcinoma associated 1 by CCAAT/enhancer binding protein α contributes to bladder cancer cell growth and reduced apoptosis. Oncol Rep 2014; 31:1993-2000. [PMID: 24648007 PMCID: PMC4020618 DOI: 10.3892/or.2014.3092] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/06/2014] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNA urothelial carcinoma associated 1 (lncRNA-UCA1) is upregulated in bladder cancer and plays a pivotal role in bladder cancer progression and metastasis. Recent studies and our research found that lncRNA-UCA1 may be an important biomarker and therapeutic target for bladder cancer. However, the molecular mechanism involved in the upregulation of lncRNA-UCA1 in bladder cancer is largely unknown. In the present study, we showed that lncRNA-UCA1 expression in bladder cancer cells was upregulated by transcription factor CCAAT/enhancer binding protein α (C/EBPα), which was the only candidate transcription factor simultaneously predicted by a total of five bioinformatical software programs. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay indicated that C/EBPα bound to the lncRNA-UCA1 core promoter region in vitro and in vivo. The luciferase assays further showed that there was a point mutation (A231G) in the C/EBPα binding site of the lncRNA-UCA1 core promoter in various bladder cancer cell lines, which in turn significantly increased the transcriptional activity of lncRNA-UCA1. We also demonstrated that C/EBPα siRNA treatment contributed to the downregulation of lncRNA-UCA1 expression, whereas overexpression of C/EBPα enhanced lncRNA-UCA1 expression. Furthermore, lncRNA-UCA1 transcriptional repression by C/EBPα siRNA sharply reduced cell viability and induced cell apoptosis in vitro. Collectively, our results provide a novel therapeutic strategy for bladder cancer by effectively interrupting the binding of the lncRNA-UCA1 promoter and certain transcription factors, so as to reverse the upregulation of lncRNA-UCA1 and prevent bladder cancer progression.
Collapse
Affiliation(s)
- Mei Xue
- Center for Translational Medicine, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Xu Li
- Center for Translational Medicine, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Wenjing Wu
- Clinical Laboratory, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Shuwan Zhang
- Center for Translational Medicine, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Shouzhen Wu
- Center for Translational Medicine, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Zhengkun Li
- Center for Translational Medicine, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Wei Chen
- Clinical Laboratory, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|
535
|
Hill JM, Lukiw WJ. Comparing miRNAs and viroids; highly conserved molecular mechanisms for the transmission of genetic information. Front Cell Neurosci 2014; 8:45. [PMID: 24600348 PMCID: PMC3929837 DOI: 10.3389/fncel.2014.00045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 02/01/2014] [Indexed: 12/29/2022] Open
Affiliation(s)
- James M Hill
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, LA, USA ; Department of Microbiology, Louisiana State University Health Sciences Center New Orleans, LA, USA ; Department of Ophthalmology, Louisiana State University Health Sciences Center New Orleans, LA, USA ; Department of Pharmacology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center New Orleans, LA, USA ; Department of Microbiology, Louisiana State University Health Sciences Center New Orleans, LA, USA ; Department of Neurology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| |
Collapse
|
536
|
Pang Q, Ge J, Shao Y, Sun W, Song H, Xia T, Xiao B, Guo J. Increased expression of long intergenic non-coding RNA LINC00152 in gastric cancer and its clinical significance. Tumour Biol 2014; 35:5441-7. [PMID: 24523021 DOI: 10.1007/s13277-014-1709-3] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 01/29/2014] [Indexed: 12/12/2022] Open
Abstract
It has been known that differential expression of long non-coding RNA (lncRNA) plays critical roles in carcinogenesis. However, the significance of lncRNA, especially long intergenic ncRNA (lincRNA, the main type of lncRNA family), in the diagnosis of gastric cancer is largely unknown. The aim of this study was to determine the expression level of LINC00152, a newfound lincRNA, in gastric carcinoma and its clinical association. The expression of LINC00152 in 71 pairs of tumorous and adjacent normal tissues from patients with gastric cancer was detected by quantitative real-time reverse transcription-polymerase chain reaction. And then, the potential associations between its level in gastric cancer tissue and the clinicopathological features were analyzed. Finally, a receiver operating characteristic (ROC) curve was constructed for differentiating patients with gastric cancer from patients with benign gastric diseases. The results showed that the expression level of LINC00152 in gastric carcinoma was significantly increased, compared with matched normal tissue (P=0.045) and normal mucosa from health control (P=0.004), respectively. Levels of LINC00152 in gastric cancer cell lines, BGC-823, MGC-803, and SGC-7901, were significantly higher than those in human normal gastric epithelial cell line GES-1. In addition, high expression of LINC00152 was correlated with invasion (P=0.042). LINC00152 levels in gastric juice from patients with gastric cancer were further found significantly higher than those from normal controls (P=0.002). Moreover, the area under the ROC curve (AUC) was up to 0.645 (95 % CI=0.559-0.740, P=0.003). This study highlights that lincRNA LINC00152 might be a novel biomarker for predicting gastric cancer.
Collapse
Affiliation(s)
- Qianqian Pang
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, 315211, China
| | | | | | | | | | | | | | | |
Collapse
|
537
|
Hill JM, Zhao Y, Bhattacharjee S, Lukiw WJ. miRNAs and viroids utilize common strategies in genetic signal transfer. Front Mol Neurosci 2014; 7:10. [PMID: 24574963 PMCID: PMC3918644 DOI: 10.3389/fnmol.2014.00010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/22/2014] [Indexed: 12/29/2022] Open
Affiliation(s)
- James M Hill
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, Louisiana State University New Orleans, LA, USA ; Department of Ophthalmology, Louisiana State University Health Sciences Center, Louisiana State University New Orleans, LA, USA ; Department of Microbiology, Louisiana State University Health Sciences Center, Louisiana State University New Orleans, LA, USA
| | - Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, Louisiana State University New Orleans, LA, USA
| | - Surjyadipta Bhattacharjee
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, Louisiana State University New Orleans, LA, USA
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, Louisiana State University New Orleans, LA, USA ; Department of Ophthalmology, Louisiana State University Health Sciences Center, Louisiana State University New Orleans, LA, USA ; Department of Neurology, Louisiana State University Health Sciences Center, Louisiana State University New Orleans, LA, USA
| |
Collapse
|
538
|
Beach A, Zhang HG, Ratajczak MZ, Kakar SS. Exosomes: an overview of biogenesis, composition and role in ovarian cancer. J Ovarian Res 2014; 7:14. [PMID: 24460816 PMCID: PMC3932023 DOI: 10.1186/1757-2215-7-14] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 01/22/2014] [Indexed: 12/12/2022] Open
Abstract
Exosomes are tiny membrane-bound vesicles that are over produced by most proliferating cell types during normal and pathological states. Their levels are up-regulated during pregnancy and disease states such as cancer. Exosomes contain a wide variety of proteins, lipids, RNAs, non-transcribed RNAs, microRNAs and small RNAs that are representative to their cellular origin and shuttle from a donor cell to a recipient cell. From intercellular communication to tumor proliferation, exosomes carry out a diverse range of functions, both helpful and harmful. Useful as biomarkers, exosomes may be applicable in diagnostic assessments as well as cell-free anti-tumor vaccines. Exosomes of ovarian cancer contain different set of proteins and miRNAs compared to exosomes of normal, cancer-free individuals. These molecules may be used as multiple “barcode” for the development of a diagnostic tool for early detection of ovarian cancer.
Collapse
Affiliation(s)
| | | | | | - Sham S Kakar
- Department of Medicine, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
539
|
A computational framework to infer human disease-associated long noncoding RNAs. PLoS One 2014; 9:e84408. [PMID: 24392133 PMCID: PMC3879311 DOI: 10.1371/journal.pone.0084408] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 11/13/2013] [Indexed: 12/11/2022] Open
Abstract
As a major class of noncoding RNAs, long noncoding RNAs (lncRNAs) have been implicated in various critical biological processes. Accumulating researches have linked dysregulations and mutations of lncRNAs to a variety of human disorders and diseases. However, to date, only a few human lncRNAs have been associated with diseases. Therefore, it is very important to develop a computational method to globally predict potential associated diseases for human lncRNAs. In this paper, we developed a computational framework to accomplish this by combining human lncRNA expression profiles, gene expression profiles, and human disease-associated gene data. Applying this framework to available human long intergenic noncoding RNAs (lincRNAs) expression data, we showed that the framework has reliable accuracy. As a result, for non-tissue-specific lincRNAs, the AUC of our algorithm is 0.7645, and the prediction accuracy is about 89%. This study will be helpful for identifying novel lncRNAs for human diseases, which will help in understanding the roles of lncRNAs in human diseases and facilitate treatment. The corresponding codes for our method and the predicted results are all available at http://asdcd.amss.ac.cn/MingXiLiu/lncRNA-disease.html.
Collapse
|
540
|
Halley P, Kadakkuzha BM, Faghihi MA, Magistri M, Zeier Z, Khorkova O, Coito C, Hsiao J, Lawrence M, Wahlestedt C. Regulation of the apolipoprotein gene cluster by a long noncoding RNA. Cell Rep 2014; 6:222-30. [PMID: 24388749 DOI: 10.1016/j.celrep.2013.12.015] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 04/03/2013] [Accepted: 12/10/2013] [Indexed: 01/22/2023] Open
Abstract
Apolipoprotein A1 (APOA1) is the major protein component of high-density lipoprotein (HDL) in plasma. We have identified an endogenously expressed long noncoding natural antisense transcript, APOA1-AS, which acts as a negative transcriptional regulator of APOA1 both in vitro and in vivo. Inhibition of APOA1-AS in cultured cells resulted in the increased expression of APOA1 and two neighboring genes in the APO cluster. Chromatin immunoprecipitation (ChIP) analyses of a ∼50 kb chromatin region flanking the APOA1 gene demonstrated that APOA1-AS can modulate distinct histone methylation patterns that mark active and/or inactive gene expression through the recruitment of histone-modifying enzymes. Targeting APOA1-AS with short antisense oligonucleotides also enhanced APOA1 expression in both human and monkey liver cells and induced an increase in hepatic RNA and protein expression in African green monkeys. Furthermore, the results presented here highlight the significant local modulatory effects of long noncoding antisense RNAs and demonstrate the therapeutic potential of manipulating the expression of these transcripts both in vitro and in vivo.
Collapse
Affiliation(s)
- Paul Halley
- Center for Therapeutic Innovation, University of Miami, Miller School of Medicine, NW 10(th) Avenue, Miami, FL 33136, USA; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, NW 10(th) Avenue, Miami, FL 33136, USA
| | - Beena M Kadakkuzha
- Center for Therapeutic Innovation, University of Miami, Miller School of Medicine, NW 10(th) Avenue, Miami, FL 33136, USA; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, NW 10(th) Avenue, Miami, FL 33136, USA
| | - Mohammad Ali Faghihi
- Center for Therapeutic Innovation, University of Miami, Miller School of Medicine, NW 10(th) Avenue, Miami, FL 33136, USA; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, NW 10(th) Avenue, Miami, FL 33136, USA
| | - Marco Magistri
- Center for Therapeutic Innovation, University of Miami, Miller School of Medicine, NW 10(th) Avenue, Miami, FL 33136, USA; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, NW 10(th) Avenue, Miami, FL 33136, USA
| | - Zane Zeier
- Center for Therapeutic Innovation, University of Miami, Miller School of Medicine, NW 10(th) Avenue, Miami, FL 33136, USA; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, NW 10(th) Avenue, Miami, FL 33136, USA
| | - Olga Khorkova
- OPKO-CURNA, 10320 USA Today Way, Miramar, FL 33025, USA
| | - Carlos Coito
- OPKO-CURNA, 10320 USA Today Way, Miramar, FL 33025, USA
| | - Jane Hsiao
- OPKO-CURNA, 10320 USA Today Way, Miramar, FL 33025, USA
| | | | - Claes Wahlestedt
- Center for Therapeutic Innovation, University of Miami, Miller School of Medicine, NW 10(th) Avenue, Miami, FL 33136, USA; Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, NW 10(th) Avenue, Miami, FL 33136, USA.
| |
Collapse
|
541
|
Guennewig B, Cooper AA. The Central Role of Noncoding RNA in the Brain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 116:153-94. [DOI: 10.1016/b978-0-12-801105-8.00007-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
542
|
Pogue AI, Clement C, Hill JM, Lukiw WJ. Evolution of microRNA (miRNA) Structure and Function in Plants and Animals: Relevance to Aging and Disease. ACTA ACUST UNITED AC 2014; 2. [PMID: 26146648 PMCID: PMC4489142 DOI: 10.4172/2329-8847.1000119] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
| | | | - James M Hill
- Departments of Ophthalmology, LSU Neuroscience Center, USA ; Departments of Microbiology, LSU Neuroscience Center, USA ; Departments of Pharmacology, LSU Neuroscience Center, USA ; Departments of Neurology, LSU Neuroscience Center, USA
| | - Walter J Lukiw
- Alchem Biotek, Toronto ON, M5S 1A8, Canada ; Departments of Ophthalmology, LSU Neuroscience Center, USA ; Departments of Neurology, LSU Neuroscience Center, USA
| |
Collapse
|
543
|
Abstract
Stress response is considered to have adaptive value for organisms faced with stressful condition. Chronic stress however adversely affects the physiology and may lead to neuropsychiatric disorders. Repeated stressful events in animal models have been shown to cause long-lasting changes in neural circuitries at molecular, cellular, and physiological level, leading to disorders of mood as well as cognition. Molecular studies in recent years have implicated diverse epigenetic mechanisms, including histone modifications, DNA methylation, and noncoding RNAs, that underlie dysregulation of genes in the affected neural circuitries in chronic stress-induced pathophysiology. A review of the myriad epigenetic regulatory mechanisms associated with neural and behavioral responses in animal models of stress-induced neuropsychiatric disorders is presented here. The review also deals with clinical evidence of the epigenetic dysregulation of genes in psychiatric disorders where chronic stress appears to underlie the etiopathology.
Collapse
|
544
|
Serafini G, Pompili M, Hansen KF, Obrietan K, Dwivedi Y, Shomron N, Girardi P. The involvement of microRNAs in major depression, suicidal behavior, and related disorders: a focus on miR-185 and miR-491-3p. Cell Mol Neurobiol 2014; 34:17-30. [PMID: 24213247 PMCID: PMC11488878 DOI: 10.1007/s10571-013-9997-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/12/2013] [Indexed: 01/08/2023]
Abstract
Major depressive disorders are common and disabling conditions associated with significant psychosocial impairment and suicide risk. At least 3-4 % of all depressive individuals die by suicide. Evidence suggests that small non-coding RNAs, in particular microRNAs (miRNAs), play a critical role in major affective disorders as well as suicide. We performed a detailed review of the current literature on miRNAs and their targets in major depression and related disorders as well as suicidal behavior, with a specific focus on miR-185 and miR-491-3p, which have been suggested to participate in the pathogenesis of major depression and/or suicide. miRNAs play a fundamental role in the development of the brain. Several miRNAs are reported to influence neuronal and circuit formation by negatively regulating gene expression. Global miRNA reduced expression was found in the prefrontal cortex of depressed suicide completers when compared to that of nonpsychiatric controls who died of other causes. One particular miRNA, miR-185, was reported to regulate TrkB-T1, which has been associated with suicidal behavior upon truncation. Furthermore, cAMP response element-binding protein-brain-derived neurotrophic factor pathways may regulate, through miRNAs, the homeostasis of neural and synaptic pathways playing a crucial role in major depression. miRNAs have gained attention as key players involved in nervous system development, physiology, and disease. Further evidence is needed to clarify the exact role that miRNAs play in major depression and related disorders and suicidal behavior.
Collapse
Affiliation(s)
- Gianluca Serafini
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1037, 00189, Rome, Italy,
| | | | | | | | | | | | | |
Collapse
|
545
|
Kumar A, Upadhyaya KC. Perspectives on the Human Genome. Anim Biotechnol 2014. [DOI: 10.1016/b978-0-12-416002-6.00031-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
546
|
Kaushik K, Leonard VE, KV S, Lalwani MK, Jalali S, Patowary A, Joshi A, Scaria V, Sivasubbu S. Dynamic expression of long non-coding RNAs (lncRNAs) in adult zebrafish. PLoS One 2013; 8:e83616. [PMID: 24391796 PMCID: PMC3877055 DOI: 10.1371/journal.pone.0083616] [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: 10/10/2013] [Accepted: 11/05/2013] [Indexed: 01/27/2023] Open
Abstract
Long non-coding RNAs (lncRNA) represent an assorted class of transcripts having little or no protein coding capacity and have recently gained importance for their function as regulators of gene expression. Molecular studies on lncRNA have uncovered multifaceted interactions with protein coding genes. It has been suggested that lncRNAs are an additional layer of regulatory switches involved in gene regulation during development and disease. LncRNAs expressing in specific tissues or cell types during adult stages can have potential roles in form, function, maintenance and repair of tissues and organs. We used RNA sequencing followed by computational analysis to identify tissue restricted lncRNA transcript signatures from five different tissues of adult zebrafish. The present study reports 442 predicted lncRNA transcripts from adult zebrafish tissues out of which 419 were novel lncRNA transcripts. Of these, 77 lncRNAs show predominant tissue restricted expression across the five major tissues investigated. Adult zebrafish brain expressed the largest number of tissue restricted lncRNA transcripts followed by cardiovascular tissue. We also validated the tissue restricted expression of a subset of lncRNAs using independent methods. Our data constitute a useful genomic resource towards understanding the expression of lncRNAs in various tissues in adult zebrafish. Our study is thus a starting point and opens a way towards discovering new molecular interactions of gene expression within the specific adult tissues in the context of maintenance of organ form and function.
Collapse
Affiliation(s)
- Kriti Kaushik
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhavan, New Delhi, India
| | - Vincent Elvin Leonard
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Shamsudheen KV
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Mukesh Kumar Lalwani
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Saakshi Jalali
- G.N. Ramachandran Knowledge Center for Genome Informatics, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhavan, New Delhi, India
| | - Ashok Patowary
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Adita Joshi
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Vinod Scaria
- G.N. Ramachandran Knowledge Center for Genome Informatics, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhavan, New Delhi, India
- * E-mail: (VS); (SS)
| | - Sridhar Sivasubbu
- Genomics and Molecular Medicine, CSIR Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Anusandhan Bhavan, New Delhi, India
- * E-mail: (VS); (SS)
| |
Collapse
|
547
|
Winterling C, Koch M, Koeppel M, Garcia-Alcalde F, Karlas A, Meyer TF. Evidence for a crucial role of a host non-coding RNA in influenza A virus replication. RNA Biol 2013; 11:66-75. [PMID: 24440876 DOI: 10.4161/rna.27504] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence suggests the non-protein coding human genome is of vital importance for human cell function. Besides small RNAs, the diverse class of long non-coding RNAs (lncRNAs) recently came into focus. However, their relevance for infection, a major evolutionary driving force, remains elusive. Using two commercially available microarray systems, namely NCode™ and Sureprint™ G3, we identified differential expression of 42 ncRNAs during influenza A virus (IAV) infection in human lung epithelial cells. This included several classes of lncRNAs, including large intergenic ncRNAs (lincRNAs). As analyzed by qRT-PCR, expression of one lincRNA, which we termed virus inducible lincRNA (VIN), is induced by several IAV strains (H1N1, H3N2, H7N7) as well as vesicular stomatitis virus. However, we did not observe an induction of VIN by influenza B virus, treatment with RNA mimics, or IFNβ. Thus, VIN expression seems to be a specific response to certain viral infections. RNA fractionation and RNA-FISH experiments revealed that VIN is localized to the host cell nucleus. Most importantly, we show that abolition of VIN by RNA interference restricts IAV replication and viral protein synthesis, highlighting the relevance of this lincRNA for productive IAV infection. Our observations suggest that viral pathogens interfere with the non-coding portion of the human genome, thereby guaranteeing their successful propagation, and that the expression of VIN correlates with their virulence. Consequently, our study provides a novel approach for understanding virus pathogenesis in greater detail, which will enable future design of new antiviral strategies targeting the host's non-protein coding genome.
Collapse
Affiliation(s)
- Carla Winterling
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Manuel Koch
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Max Koeppel
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Fernando Garcia-Alcalde
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Alexander Karlas
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology; Max Planck Institute for Infection Biology; Berlin, Germany
| |
Collapse
|
548
|
Li L, Sun R, Liang Y, Pan X, Li Z, Bai P, Zeng X, Zhang D, Zhang L, Gao L. Association between polymorphisms in long non-coding RNA PRNCR1 in 8q24 and risk of colorectal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:104. [PMID: 24330491 PMCID: PMC4029281 DOI: 10.1186/1756-9966-32-104] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 12/05/2013] [Indexed: 01/12/2023]
Abstract
BACKGROUND Genome-wide association studies have identified that genetic variants in 8q24 confer susceptibility to colorectal cancer (CRC). Recently, a novel lncRNA (PRNCR1) that located in the 8q24 was discovered. Single nucleotide polymorphisms (SNPs) in the lncRNAs may influence the process of splicing and stability of mRNA conformation, resulting in the modification of its interacting partners. We hypothesized that SNPs in the lncRNA PRNCR1 may be related to the risk of CRC. METHODS We conducted a case-control study and genotyped five tag SNPs in the lncRNA PRNCR1 in 908 subjects including 313 cases with CRC and 595 control subjects using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assay. RESULTS In overall analyses, we found that the rs13252298 and rs1456315 were associated with significantly decreased risks of CRC. In stratification analyses, we found that CRC patients carrying the rs1456315G were likely to have a tumor size of greater than 5 cm (G vs. A: adjusted OR = 1.56, 95% CI: 1.10-2.23). Additionally, patients with the rs7007694C and rs16901946G had decreased risks to develop poorly differentiated CRC, whereas patients with the rs1456315G had an increased risk to develop poorly differentiated CRC. CONCLUSION These findings suggest that SNPs in the lncRNA PRNCR1 may contribute to susceptibility to CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lin Zhang
- Laboratory of Molecular and Translational Medicine, West China Institute of Women and Children's Health; Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P,R, China.
| | | |
Collapse
|
549
|
|
550
|
Josset L, Tisoncik-Go J, Katze MG. Moving H5N1 studies into the era of systems biology. Virus Res 2013; 178:151-67. [PMID: 23499671 PMCID: PMC3834220 DOI: 10.1016/j.virusres.2013.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 02/24/2013] [Indexed: 12/20/2022]
Abstract
The dynamics of H5N1 influenza virus pathogenesis are multifaceted and can be seen as an emergent property that cannot be comprehended without looking at the system as a whole. In past years, most of the high-throughput studies on H5N1-host interactions have focused on the host transcriptomic response, at the cellular or the lung tissue level. These studies pointed out that the dynamics and magnitude of the innate immune response and immune cell infiltration is critical to H5N1 pathogenesis. However, viral-host interactions are multidimensional and advances in technologies are creating new possibilities to systematically measure additional levels of 'omic data (e.g. proteomic, metabolomic, and RNA profiling) at each temporal and spatial scale (from the single cell to the organism) of the host response. Natural host genetic variation represents another dimension of the host response that determines pathogenesis. Systems biology models of H5N1 disease aim at understanding and predicting pathogenesis through integration of these different dimensions by using intensive computational modeling. In this review, we describe the importance of 'omic studies for providing a more comprehensive view of infection and mathematical models that are being developed to integrate these data. This review provides a roadmap for what needs to be done in the future and what computational strategies should be used to build a global model of H5N1 pathogenesis. It is time for systems biology of H5N1 pathogenesis to take center stage as the field moves toward a more comprehensive view of virus-host interactions.
Collapse
Affiliation(s)
- Laurence Josset
- Department of Microbiology, School of Medicine, University of Washington, Seattle, WA 98195, United States
| | | | | |
Collapse
|