201
|
Weng Y, Chen Y, Chen J, Liu Y, Bao T. RETRACTED ARTICLE: Common genetic variants in microRNA processing machinery genes are associated with risk and survival in patients with osteosarcoma. Mol Genet Genomics 2016. [DOI: 10.1007/s00438-015-1006-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
202
|
Basak I, Patil KS, Alves G, Larsen JP, Møller SG. microRNAs as neuroregulators, biomarkers and therapeutic agents in neurodegenerative diseases. Cell Mol Life Sci 2016; 73:811-27. [PMID: 26608596 PMCID: PMC11108480 DOI: 10.1007/s00018-015-2093-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/14/2015] [Accepted: 11/09/2015] [Indexed: 01/03/2023]
Abstract
The last decade has experienced the emergence of microRNAs as a key molecular tool for the diagnosis and prognosis of human diseases. Although the focus has mostly been on cancer, neurodegenerative diseases present an exciting, yet less explored, platform for microRNA research. Several studies have highlighted the significance of microRNAs in neurogenesis and neurodegeneration, and pre-clinical studies have shown the potential of microRNAs as biomarkers. Despite this, no bona fide microRNAs have been identified as true diagnostic or prognostic biomarkers for neurodegenerative disease. This is mainly due to the lack of precisely defined patient cohorts and the variability within and between individual cohorts. However, the discovery that microRNAs exist as stable molecules at detectable levels in body fluids has opened up new avenues for microRNAs as potential biomarker candidates. Furthermore, technological developments in microRNA biology have contributed to the possible design of microRNA-mediated disease intervention strategies. The combination of these advancements, with the availability of well-defined longitudinal patient cohort, promises to not only assist in developing invaluable diagnostic tools for clinicians, but also to increase our overall understanding of the underlying heterogeneity of neurodegenerative diseases. In this review, we present a comprehensive overview of the existing knowledge of microRNAs in neurodegeneration and provide a perspective of the applicability of microRNAs as a basis for future therapeutic intervention strategies.
Collapse
Affiliation(s)
- Indranil Basak
- Department of Biological Sciences, St. John's University, 8000 Utopia Parkway, New York, NY, 11439, USA
| | - Ketan S Patil
- Department of Biological Sciences, St. John's University, 8000 Utopia Parkway, New York, NY, 11439, USA
| | - Guido Alves
- Norwegian Center for Movement Disorders, Stavanger University Hospital, 4068, Stavanger, Norway
| | - Jan Petter Larsen
- Norwegian Center for Movement Disorders, Stavanger University Hospital, 4068, Stavanger, Norway
| | - Simon Geir Møller
- Department of Biological Sciences, St. John's University, 8000 Utopia Parkway, New York, NY, 11439, USA.
- Norwegian Center for Movement Disorders, Stavanger University Hospital, 4068, Stavanger, Norway.
| |
Collapse
|
203
|
Akhtar MM, Micolucci L, Islam MS, Olivieri F, Procopio AD. Bioinformatic tools for microRNA dissection. Nucleic Acids Res 2016; 44:24-44. [PMID: 26578605 PMCID: PMC4705652 DOI: 10.1093/nar/gkv1221] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 10/27/2015] [Accepted: 10/28/2015] [Indexed: 12/21/2022] Open
Abstract
Recently, microRNAs (miRNAs) have emerged as important elements of gene regulatory networks. MiRNAs are endogenous single-stranded non-coding RNAs (~22-nt long) that regulate gene expression at the post-transcriptional level. Through pairing with mRNA, miRNAs can down-regulate gene expression by inhibiting translation or stimulating mRNA degradation. In some cases they can also up-regulate the expression of a target gene. MiRNAs influence a variety of cellular pathways that range from development to carcinogenesis. The involvement of miRNAs in several human diseases, particularly cancer, makes them potential diagnostic and prognostic biomarkers. Recent technological advances, especially high-throughput sequencing, have led to an exponential growth in the generation of miRNA-related data. A number of bioinformatic tools and databases have been devised to manage this growing body of data. We analyze 129 miRNA tools that are being used in diverse areas of miRNA research, to assist investigators in choosing the most appropriate tools for their needs.
Collapse
Affiliation(s)
- Most Mauluda Akhtar
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona 60100, Italy Computational Pathology Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona 60100, Italy
| | - Luigina Micolucci
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona 60100, Italy Computational Pathology Unit, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona 60100, Italy
| | - Md Soriful Islam
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona 60100, Italy
| | - Fabiola Olivieri
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona 60100, Italy Center of Clinical Pathology and Innovative Therapies, Italian National Research Center on Aging (INRCA-IRCCS), Ancona 60121, Italy
| | - Antonio Domenico Procopio
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona 60100, Italy Center of Clinical Pathology and Innovative Therapies, Italian National Research Center on Aging (INRCA-IRCCS), Ancona 60121, Italy
| |
Collapse
|
204
|
Circulating cell-free microRNAs as biomarkers for colorectal cancer. Surg Today 2016; 46:13-24. [PMID: 25712224 DOI: 10.1007/s00595-015-1138-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/11/2015] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are a class of small, endogenous, non-coding, single-stranded RNAs that act as post-transcriptional regulators. Their discovery has provided new avenues for the diagnosis and treatment of cancer. The expression of both oncogenic and tumor suppressor miRNAs can be aberrantly either up- or down-regulated in cancer cells. These miRNAs target mRNAs of genes that either promote or inhibit tumor growth, and are one of several epigenetic factors that control the initiation and progression of colorectal cancer (CRC) and other cancers. Investigations of miRNAs as CRC biomarkers have employed the expression profiling of traditional tissue samples and, more recently, non-invasive samples, such as feces and body fluids, have been analyzed. MiRNAs may also be able to predict responses to chemo- and radiotherapy, and may be manipulated to modify CRC characteristics. We herein discuss the use of circulating miRNAs as possible non-invasive biomarkers of early CRC onset, relapse, or response to treatment. We also discuss the obstacles that currently limit the routine use of epigenetic biomarkers in clinical settings.
Collapse
|
205
|
Abstract
The transforming growth factor-β (TGF-β) is known to regulate a large number of biological processes and is involved in various aspects of tumor development. Recent studies have shown that the biogenesis of miRNAs can be regulated by TGF-β signaling directly via Smad-dependent mechanisms and/or other unknown mechanisms, which may induce autoregulatory feedback loops in response to the activation of TGF-β signaling, influencing the fate of tumor cells. In this chapter, we summarize the currently described mechanisms underlying TGF-β's regulation of miRNA biogenesis, and the functional role of TGF-β-regulated miRNAs in tumor initiation, epithelial-mesenchymal transition, and tumor microenvironment modulation. Finally, we introduce methods to study TGF-β-regulated miRNAs and their functions in tumor progression and metastasis using an example of publication from our lab demonstrating the presence of a TGF-β-miR-34a-CCL22 signaling axis, which serves as a potent etiological pathway for the development of hepatocellular carcinoma venous metastases.
Collapse
|
206
|
Nassar W, El-Ansary M, Fayyad T, Aziz MA. Extracellular Micro-RNAs in Health and Disease: Basic Science, Biogenesis and Release. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/ajmb.2016.61001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
207
|
Macias S, Cordiner RA, Gautier P, Plass M, Cáceres JF. DGCR8 Acts as an Adaptor for the Exosome Complex to Degrade Double-Stranded Structured RNAs. Mol Cell 2015; 60:873-85. [PMID: 26687677 PMCID: PMC4691244 DOI: 10.1016/j.molcel.2015.11.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 07/16/2015] [Accepted: 11/06/2015] [Indexed: 01/18/2023]
Abstract
The Microprocessor complex (DGCR8/Drosha) is required for microRNA (miRNA) biogenesis but also binds and regulates the stability of several types of cellular RNAs. Of particular interest, DGCR8 controls the stability of mature small nucleolar RNA (snoRNA) transcripts independently of Drosha, suggesting the existence of alternative DGCR8 complex(es) with other nucleases to process a variety of cellular RNAs. Here, we found that DGCR8 copurifies with subunits of the nuclear exosome, preferentially associating with its hRRP6-containing nucleolar form. Importantly, we demonstrate that DGCR8 is essential for the recruitment of the exosome to snoRNAs and to human telomerase RNA. In addition, we show that the DGCR8/exosome complex controls the stability of the human telomerase RNA component (hTR/TERC). Altogether, these data suggest that DGCR8 acts as an adaptor to recruit the exosome complex to structured RNAs and induce their degradation. DGCR8 forms an alternative complex with the hRRP6-containing form of the exosome DGCR8 acts as an adaptor to recruit the exosome to target structured RNAs The DGCR8/hRRP6 complex also controls the stability of human telomerase RNA
Collapse
Affiliation(s)
- Sara Macias
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Ross A Cordiner
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Philippe Gautier
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Mireya Plass
- Department of Biology, Center for Computational and Applied Transcriptomics, University of Copenhagen, Ole Maaloes Vej 5, 2200 Copenhagen, Denmark
| | - Javier F Cáceres
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK.
| |
Collapse
|
208
|
Joshi S, Wei J, Bishopric NH. A cardiac myocyte-restricted Lin28/let-7 regulatory axis promotes hypoxia-mediated apoptosis by inducing the AKT signaling suppressor PIK3IP1. Biochim Biophys Acta Mol Basis Dis 2015; 1862:240-51. [PMID: 26655604 DOI: 10.1016/j.bbadis.2015.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/02/2015] [Accepted: 12/01/2015] [Indexed: 12/14/2022]
Abstract
RATIONALE The let-7 family of microRNAs (miRs) regulates critical cell functions, including survival signaling, differentiation, metabolic control and glucose utilization. These functions may be important during myocardial ischemia. MiR-let-7 expression is under tight temporal and spatial control through multiple redundant mechanisms that may be stage-, isoform- and tissue-specific. OBJECTIVE To determine the mechanisms and functional consequences of miR-let-7 regulation by hypoxia in the heart. METHODS AND RESULTS MiR-let-7a, -7c and -7g were downregulated in the adult mouse heart early after coronary occlusion, and in neonatal rat ventricular myocytes subjected to hypoxia. Let-7 repression did not require glucose depletion, and occurred at a post-transcriptional level. Hypoxia also induced the RNA binding protein Lin28, a negative regulator of let-7. Hypoxia ineither induced Lin28 nor repressed miR-let-7 in cardiac fibroblasts. Both changes were abrogated by treatment with the histone deacetylase inhibitor trichostatin A. Restoration of let-7g to hypoxic myocytes and to ischemia-reperfused mouse hearts in vivo via lentiviral transduction potentiated the hypoxia-induced phosphorylation and activation of Akt, and prevented hypoxia-dependent caspase activation and death. Mechanistically, phosphatidyl inositol 3-kinase interacting protein 1 (Pik3ip1), a negative regulator of PI3K, was identified as a novel target of miR-let-7 by a crosslinking technique showing that miR-let-7g specifically targets Pik3ip1 to the cardiac myocyte Argonaute complex RISC. Finally, in non-failing and failing human myocardium, we found specific inverse relationships between Lin28 and miR-let-7g, and between miR-let-7g and PIK3IP1. CONCLUSION A conserved hypoxia-responsive Lin28-miR-let-7-Pik3ip1 regulatory axis is specific to cardiac myocytes and promotes apoptosis during myocardial ischemic injury.
Collapse
Affiliation(s)
- Shaurya Joshi
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jianqin Wei
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nanette H Bishopric
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, United States; Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States.
| |
Collapse
|
209
|
Abstract
MicroRNAs (miRNAs) are integral to the gene regulatory network. A single miRNA is capable of controlling the expression of hundreds of protein coding genes and modulate a wide spectrum of biological functions, such as proliferation, differentiation, stress responses, DNA repair, cell adhesion, motility, inflammation, cell survival, senescence and apoptosis, all of which are fundamental to tumorigenesis. Overexpression, genetic amplification, and gain-of-function mutation of oncogenic miRNAs ("onco-miRs") as well as genetic deletion and loss-of-function mutation of tumor suppressor miRNAs ("suppressor-miRs") are linked to human cancer. In addition to the dysregulation of a specific onco-miR or suppressor-miRs, changes in global miRNA levels resulting from a defective miRNA biogenesis pathway play a role in tumorigenesis. The function of individual onco-miRs and suppressor-miRs and their target genes in cancer has been described in many different articles elsewhere. In this review, we primarily focus on the recent development regarding the dysregulation of the miRNA biogenesis pathway and its contribution to cancer.
Collapse
Affiliation(s)
- Akiko Hata
- a Cardiovascular Research Institute, University of California , San Francisco , CA , USA
| | - Risa Kashima
- a Cardiovascular Research Institute, University of California , San Francisco , CA , USA
| |
Collapse
|
210
|
The human decapping scavenger enzyme DcpS modulates microRNA turnover. Sci Rep 2015; 5:16688. [PMID: 26584588 PMCID: PMC4653633 DOI: 10.1038/srep16688] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 10/19/2015] [Indexed: 11/16/2022] Open
Abstract
The decapping scavenger enzyme DcpS is known for its role in hydrolyzing the cap structure following mRNA degradation. Recently, we discovered a new function in miRNA degradation activation for the ortholog of DcpS in C. elegans. Here we show that human DcpS conserves its role in miRNA turnover. In human cells, DcpS is a nucleocytoplasmic shuttling protein that activates miRNA degradation independently of its scavenger decapping activity in the cytoplasmic compartment. We also demonstrate that this new function for DcpS requires the contribution of the 5′-3′ exonuclease Xrn2. Our findings support a conserved role of DcpS as a modulator of miRNA turnover in animals.
Collapse
|
211
|
Wang W, Liu W, Liu Q, Li B, An L, Hao R, Zhao J, Liu S, Song J. Coordinated microRNA and messenger RNA expression profiles for understanding sexual dimorphism of gonads and the potential roles of microRNA in the steroidogenesis pathway in Nile tilapia (Oreochromis niloticus). Theriogenology 2015; 85:970-978. [PMID: 26719037 DOI: 10.1016/j.theriogenology.2015.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 11/05/2015] [Accepted: 11/07/2015] [Indexed: 12/20/2022]
Abstract
Sexual dimorphism is a widespread phenomenon in animals. However, the potential role of microRNAs (miRNAs) in regulating this dimorphism is not fully understood. In our study, we used an integrated approach to identify functional targets of miRNA by combining the paired expression profiles of miRNAs and messenger RNAs (mRNAs) in ovaries and testes of young Nile tilapia, Oreochromis niloticus. The results revealed that 67 upregulated and nine downregulated miRNAs and 2299 upregulated and 3260 downregulated genes were identified in the ovary compared with those in the testis (P < 0.01). The target genes of differentially expressed miRNAs were predicted and overlapped with the differentially expressed mRNAs. Furthermore, Kyoto Encyclopedia of Genes and Genomes pathway analyses were conducted in these coincident genes. By correlating miRNA-mRNA and predicting computational target, two types of negatively regulatory miRNA-mRNA correlations (upregulated or downregulated miRNA and downregulated or upregulated mRNA) were obtained. Seven functional miRNA-target gene pairs, miR-17-5p/DMRT1, miR-20a/DMRT1, miR-138/CYP17A2, miR-338/CYP17A2, miR-200a/CYP17A2, miR-456/AMH, and miR-138/AMH, were predicted at the sequence level and further detected by real-time polymerase chain reaction on the basis of the significantly negative relationships. Our results suggest that the integrated analysis of miRNA and mRNA expression profiling can provide novel insights into the molecular mechanism of sexual dimorphism.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Aquiculture, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China
| | - Wenzhong Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China.
| | - Qing Liu
- Department of Aquiculture, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China; Key Laboratory of Freshwater Fish Germplasm Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Baojun Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China
| | - Lixia An
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China
| | - Ruirong Hao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China
| | - Jinliang Zhao
- Key Laboratory of Freshwater Fish Germplasm Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China.
| | - Shaozhen Liu
- Department of Aquiculture, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China
| | - Jing Song
- Department of Aquiculture, College of Animal Science and Technology, Shanxi Agricultural University, Shanxi, China
| |
Collapse
|
212
|
Roberts AF, Devos Y, Lemgo GNY, Zhou X. Biosafety research for non-target organism risk assessment of RNAi-based GE plants. FRONTIERS IN PLANT SCIENCE 2015; 6:958. [PMID: 26594220 PMCID: PMC4635219 DOI: 10.3389/fpls.2015.00958] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/20/2015] [Indexed: 05/02/2023]
Abstract
RNA interference, or RNAi, refers to a set of biological processes that make use of conserved cellular machinery to silence genes. Although there are several variations in the source and mechanism, they are all triggered by double stranded RNA (dsRNA) which is processed by a protein complex into small, single stranded RNA, referred to as small interfering RNAs (siRNA) with complementarity to sequences in genes targeted for silencing. The use of the RNAi mechanism to develop new traits in plants has fueled a discussion about the environmental safety of the technology for these applications, and this was the subject of a symposium session at the 13th ISBGMO in Cape Town, South Africa. This paper continues that discussion by proposing research areas that may be beneficial for future environmental risk assessments of RNAi-based genetically modified plants, with a particular focus on non-target organism assessment.
Collapse
Affiliation(s)
- Andrew F. Roberts
- ILSI Research Foundation, Center for Environmental Risk AssessmentWashington, DC, USA
| | - Yann Devos
- GMO Unit, European Food Safety AuthorityParma, Italy
| | - Godwin N. Y. Lemgo
- NEPAD Agency – African Biosafety Network of ExpertiseOuagadougou, Burkina Faso
| | - Xuguo Zhou
- Department of Entomology, College of Agriculture, Food and Environment, University of KentuckyLexington, KY, USA
| |
Collapse
|
213
|
Brunet-Vega A, Pericay C, Quílez ME, Ramírez-Lázaro MJ, Calvet X, Lario S. Variability in microRNA recovery from plasma: Comparison of five commercial kits. Anal Biochem 2015; 488:28-35. [DOI: 10.1016/j.ab.2015.07.018] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/27/2015] [Accepted: 07/31/2015] [Indexed: 02/06/2023]
|
214
|
KH domain protein RCF3 is a tissue-biased regulator of the plant miRNA biogenesis cofactor HYL1. Proc Natl Acad Sci U S A 2015; 112:14096-101. [PMID: 26512101 DOI: 10.1073/pnas.1512865112] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The biogenesis of microRNAs (miRNAs), which regulate mRNA abundance through posttranscriptional silencing, comprises multiple well-orchestrated processing steps. We have identified the Arabidopsis thaliana K homology (KH) domain protein REGULATOR OF CBF GENE EXPRESSION 3 (RCF3) as a cofactor affecting miRNA biogenesis in specific plant tissues. MiRNA and miRNA-target levels were reduced in apex-enriched samples of rcf3 mutants, but not in other tissues. Mechanistically, RCF3 affects miRNA biogenesis through nuclear interactions with the phosphatases C-TERMINAL DOMAIN PHOSPHATASE-LIKE1 and 2 (CPL1 and CPL2). These interactions are essential to regulate the phosphorylation status, and thus the activity, of the double-stranded RNA binding protein and DICER-LIKE1 (DCL1) cofactor HYPONASTIC LEAVES1 (HYL1).
Collapse
|
215
|
Gong Z, Yang Q, Zeng Z, Zhang W, Li X, Zu X, Deng H, Chen P, Liao Q, Xiang B, Zhou M, Li X, Li Y, Xiong W, Li G. An integrative transcriptomic analysis reveals p53 regulated miRNA, mRNA, and lncRNA networks in nasopharyngeal carcinoma. Tumour Biol 2015; 37:3683-95. [PMID: 26462838 DOI: 10.1007/s13277-015-4156-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022] Open
Abstract
It has been reported that p53 dysfunction is closely related to the carcinogenesis of nasopharyngeal carcinoma (NPC). Recently, an increasing body of evidence has indicated that microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) participate in p53-associated signaling pathways and, in addition to mRNAs, form a complex regulation network to promote tumor occurrence and progression. The aim of this study was to elucidate the p53-regulated miRNAs, mRNAs, and lncRNAs and their regulating networks in NPC. Firstly, we overexpressed p53 in the NPC cell line HNE2 and performed transcriptomic gene expression profiling (GEP) analysis, which included miRNAs, mRNAs, and lncRNAs, using microarray technology at 0, 12, 24, and 48 h after transfection. There were 38 miRNAs (33 upregulated and 5 downregulated), 2107 mRNAs (296 upregulated and 1811 downregulated), and 1190 lncRNAs (133 upregulated and 1057 downregulated) that were significantly dysregulated by p53. Some of the dysregulated molecules were confirmed by quantitative real-time polymerase chain reaction (qRT-PCR). Then, we integrated previously published miRNAs, mRNAs, and lncRNAs GEP datasets from NPC biopsies to investigate the expression of these p53 regulated molecules and found that 7 miRNAs, 218 mRNAs, and 101 lncRNAs regulated by p53 were also differentially expressed in NPC tissues. Finally, p53-regulated miRNA, mRNA, and lncRNA networks were constructed using bioinformatics methods. These miRNAs, mRNAs, and lncRNAs may participate in p53 downstream signaling pathways and play important roles in the carcinogenesis of NPC. Thorough investigations of their biological functions and regulating relationships will provide a novel view of the p53 signaling pathway, and the restoration of p53 functioning or its downstream gene regulating network is potentially of great value in treating NPC patients.
Collapse
Affiliation(s)
- Zhaojian Gong
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qian Yang
- Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,School of Nursing, Hunan Polytechnic of Environment and Biology, Hengyang, Hunan, China
| | - Zhaoyang Zeng
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Wenling Zhang
- Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuyu Zu
- Clinical Research Institution, the First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Hao Deng
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pan Chen
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bo Xiang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Wei Xiong
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
216
|
Cammaerts S, Strazisar M, Dierckx J, Del Favero J, De Rijk P. miRVaS: a tool to predict the impact of genetic variants on miRNAs. Nucleic Acids Res 2015; 44:e23. [PMID: 26384425 PMCID: PMC4756848 DOI: 10.1093/nar/gkv921] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/04/2015] [Indexed: 01/10/2023] Open
Abstract
Genetic variants in or near miRNA genes can have profound effects on miRNA expression and targeting. As user-friendly software for the impact prediction of miRNA variants on a large scale is still lacking, we created a tool called miRVaS. miRVaS automates this prediction by annotating the location of the variant relative to functional regions within the miRNA hairpin (seed, mature, loop, hairpin arm, flanks) and by annotating all predicted structural changes within the miRNA due to the variant. In addition, the tool defines the most important region that is predicted to have structural changes and calculates a conservation score that is indicative of the reliability of the structure prediction. The output is presented in a tab-separated file, which enables fast screening, and in an html file, which allows visual comparison between wild-type and variant structures. All separate images are provided for downstream use. Finally, we tested two different approaches on a small test set of published functionally validated genetic variants for their capacity to predict the impact of variants on miRNA expression.
Collapse
Affiliation(s)
- Sophia Cammaerts
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, Antwerp, 2610, Belgium University of Antwerp, Antwerp, 2610, Belgium
| | - Mojca Strazisar
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, Antwerp, 2610, Belgium University of Antwerp, Antwerp, 2610, Belgium
| | | | - Jurgen Del Favero
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, Antwerp, 2610, Belgium University of Antwerp, Antwerp, 2610, Belgium Multiplicom N.V., Niel, 2845, Belgium
| | - Peter De Rijk
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, Antwerp, 2610, Belgium University of Antwerp, Antwerp, 2610, Belgium
| |
Collapse
|
217
|
Kang H, Kim C, Lee H, Rho JG, Seo JW, Nam JW, Song WK, Nam SW, Kim W, Lee EK. Downregulation of microRNA-362-3p and microRNA-329 promotes tumor progression in human breast cancer. Cell Death Differ 2015; 23:484-95. [PMID: 26337669 DOI: 10.1038/cdd.2015.116] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 06/15/2015] [Accepted: 06/26/2015] [Indexed: 12/31/2022] Open
Abstract
p130Cas regulates cancer progression by driving tyrosine receptor kinase signaling. Tight regulation of p130Cas expression is necessary for survival, apoptosis, and maintenance of cell motility in various cell types. Several studies revealed that transcriptional and post-translational control of p130Cas are important for maintenance of its expression and activity. To explore novel regulatory mechanisms of p130Cas expression, we studied the effect of microRNAs (miRs) on p130Cas expression in human breast cancer MCF7 cells. Here, we provide experimental evidence that miR-362-3p and miR-329 perform a tumor-suppressive function and their expression is downregulated in human breast cancer. miR-362-3p and miR-329 inhibited cellular proliferation, migration, and invasion, thereby suppressing tumor growth, by downregulating p130Cas. Ectopic expression of p130Cas attenuated the inhibitory effects of the two miRs on tumor progression. Relative expression levels of miR-362-3p/329 and p130Cas between normal and breast cancer correlated inversely; miR-362-3p/329 expression was decreased, whereas that of p130Cas increased in breast cancers. Furthermore, we showed that downregulation of miR-362-3p and miR-329 was caused by differential DNA methylation of miR genes. Enhanced DNA methylation (according to methylation-specific PCR) was responsible for downregulation of miR-362-3p and miR-329 in breast cancer. Taken together, these findings point to a novel role for miR-362-3p and miR-329 as tumor suppressors; the miR-362-3p/miR-329-p130Cas axis seemingly has a crucial role in breast cancer progression. Thus, modulation of miR-362-3p/miR-329 may be a novel therapeutic strategy against breast cancer.
Collapse
Affiliation(s)
- H Kang
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - C Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - H Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - J G Rho
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - J-W Seo
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea
| | - J-W Nam
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea
| | - W K Song
- Department of Life Science, Bio Imaging and Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - S W Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Cancer Evolution Center, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - W Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - E K Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Cancer Evolution Center, College of Medicine, Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
218
|
Khan IS, Park CY, Mavropoulos A, Shariat N, Pollack JL, Barczak AJ, Erle DJ, McManus MT, Anderson MS, Jeker LT. Identification of MiR-205 As a MicroRNA That Is Highly Expressed in Medullary Thymic Epithelial Cells. PLoS One 2015; 10:e0135440. [PMID: 26270036 PMCID: PMC4535774 DOI: 10.1371/journal.pone.0135440] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/22/2015] [Indexed: 11/30/2022] Open
Abstract
Thymic epithelial cells (TECs) support T cell development in the thymus. Cortical thymic epithelial cells (cTECs) facilitate positive selection of developing thymocytes whereas medullary thymic epithelial cells (mTECs) facilitate the deletion of self-reactive thymocytes in order to prevent autoimmunity. The mTEC compartment is highly dynamic with continuous maturation and turnover, but the genetic regulation of these processes remains poorly understood. MicroRNAs (miRNAs) are important regulators of TEC genetic programs since miRNA-deficient TECs are severely defective. However, the individual miRNAs important for TEC maintenance and function and their mechanisms of action remain unknown. Here, we demonstrate that miR-205 is highly and preferentially expressed in mTECs during both thymic ontogeny and in the postnatal thymus. This distinct expression is suggestive of functional importance for TEC biology. Genetic ablation of miR-205 in TECs, however, neither revealed a role for miR-205 in TEC function during homeostatic conditions nor during recovery from thymic stress conditions. Thus, despite its distinct expression, miR-205 on its own is largely dispensable for mTEC biology.
Collapse
Affiliation(s)
- Imran S. Khan
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Chong Y. Park
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
| | - Anastasia Mavropoulos
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Nikki Shariat
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Joshua L. Pollack
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Andrea J. Barczak
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - David J. Erle
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Michael T. McManus
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Mark S. Anderson
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MSA); (LTJ)
| | - Lukas T. Jeker
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MSA); (LTJ)
| |
Collapse
|
219
|
Zhao XD, Lu YY, Guo H, Xie HH, He LJ, Shen GF, Zhou JF, Li T, Hu SJ, Zhou L, Han YN, Liang SL, Wang X, Wu KC, Shi YQ, Nie YZ, Fan DM. MicroRNA-7/NF-κB signaling regulatory feedback circuit regulates gastric carcinogenesis. J Cell Biol 2015; 210:613-27. [PMID: 26261179 PMCID: PMC4539989 DOI: 10.1083/jcb.201501073] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/30/2015] [Indexed: 01/27/2023] Open
Abstract
RELA and FOS are targets of miR-7 in gastric cancer cells and the miR-7/IKKε/RELA reciprocal feedback loop is important for gastric cancer induced by H. pylori infection. MicroRNAs play essential roles in gene expression regulation during carcinogenesis. Here, we investigated the role of miR-7 and the mechanism by which it is dysregulated in gastric cancer (GC). We used genome-wide screenings and identified RELA and FOS as novel targets of miR-7. Overexpression of miR-7 repressed RELA and FOS expression and prevented GC cell proliferation and tumorigenesis. These effects were clinically relevant, as low miR-7 expression was correlated with high RELA and FOS expression and poor survival in GC patients. Intriguingly, we found that miR-7 indirectly regulated RELA activation by targeting the IκB kinase IKKε. Furthermore, IKKε and RELA can repress miR-7 transcription, which forms a feedback circuit between miR-7 and nuclear factor κB (NF-κB) signaling. Additionally, we demonstrate that down-regulation of miR-7 may occur as a result of the aberrant activation of NF-κB signaling by Helicobacter pylori infection. These findings suggest that miR-7 may serve as an important regulator in GC development and progression.
Collapse
Affiliation(s)
- Xiao-Di Zhao
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yuan-Yuan Lu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hao Guo
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hua-Hong Xie
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Li-Jie He
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Gao-Fei Shen
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jin-Feng Zhou
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ting Li
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Si-Jun Hu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Lin Zhou
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ya-Nan Han
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Shu-Li Liang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xin Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Kai-Chun Wu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yong-Quan Shi
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yong-Zhan Nie
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Dai-Ming Fan
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
220
|
Romero-Cordoba SL, Salido-Guadarrama I, Rodriguez-Dorantes M, Hidalgo-Miranda A. miRNA biogenesis: biological impact in the development of cancer. Cancer Biol Ther 2015; 15:1444-55. [PMID: 25482951 PMCID: PMC4622859 DOI: 10.4161/15384047.2014.955442] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
microRNAs (miRNAs) are non coding RNAs with different biological functions and pathological implications. Given their role as post-transcriptional gene expression regulators, they are involved in several important physiological processes like development, cell differentiation and cell signaling. miRNAs act as modulators of gene expression programs in different diseases, particularly in cancer, where they act through the repression of genes which are critical for carcinogenesis. The expression level of mature miRNAs is the result of a fine mechanism of biogenesis, carried out by different enzymatic complexes that exert their function at transcriptional and post-transcriptional levels. In this review, we will focus our discussion on the alterations in the miRNA biogenesis machinery, and its impact on the establishment and development of cancer programs.
Collapse
Key Words
- Ago2, Argonaute 2 protein
- Ars2, Arsenic Resistance protein 2
- DGCR8, DiGeorge syndrome Critical Region 8 protein
- EMT, epithelial–mesenchymal transition
- KSRP, KH-type splicing regulatory protein
- MK2, MAPK-activated protein kinase 2
- PABP, poly(A)-binding protein
- PACT, kinase R–activating protein
- PRC2, Polycomb repressor complex
- RISC, RNA-induced silencing complex
- TRBP, TAR RNA binding protein
- TUT4, terminal uridine transferase-4
- XPO5, exportin 5
- cancer
- cellular signaling
- circRNA, circular RNA
- hnRNPs, heterogeneous nuclear ribonucleoproteins
- miRNA biogenesis
- miRNAs, microRNAs
Collapse
|
221
|
Du P, Wang L, Sliz P, Gregory RI. A Biogenesis Step Upstream of Microprocessor Controls miR-17∼92 Expression. Cell 2015; 162:885-99. [PMID: 26255770 DOI: 10.1016/j.cell.2015.07.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 04/24/2015] [Accepted: 06/22/2015] [Indexed: 12/21/2022]
Abstract
The precise control of miR-17∼92 microRNA (miRNA) is essential for normal development, and overexpression of certain miRNAs from this cluster is oncogenic. Here, we find that the relative expression of the six miRNAs processed from the primary (pri-miR-17∼92) transcript is dynamically regulated during embryonic stem cell (ESC) differentiation. Pri-miR-17∼92 is processed to a biogenesis intermediate, termed "progenitor-miRNA" (pro-miRNA). Pro-miRNA is an efficient substrate for Microprocessor and is required to selectively license production of pre-miR-17, pre-miR-18a, pre-miR-19a, pre-miR-20a, and pre-miR-19b from this cluster. Two complementary cis-regulatory repression domains within pri-miR-17∼92 are required for the blockade of miRNA processing through the formation of an autoinhibitory RNA conformation. The endonuclease CPSF3 (CPSF73) and the spliceosome-associated ISY1 are responsible for pro-miRNA biogenesis and expression of all miRNAs within the cluster except miR-92. Thus, developmentally regulated pro-miRNA processing is a key step controlling miRNA expression and explains the posttranscriptional control of miR-17∼92 expression in development.
Collapse
Affiliation(s)
- Peng Du
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Longfei Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Piotr Sliz
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Richard I Gregory
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Boston, MA 02115, USA.
| |
Collapse
|
222
|
Zhang L, Wei P, Shen X, Zhang Y, Xu B, Zhou J, Fan S, Hao Z, Shi H, Zhang X, Kong R, Xu L, Gao J, Zou D, Liang C. MicroRNA Expression Profile in Penile Cancer Revealed by Next-Generation Small RNA Sequencing. PLoS One 2015; 10:e0131336. [PMID: 26158897 PMCID: PMC4497725 DOI: 10.1371/journal.pone.0131336] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/01/2015] [Indexed: 12/14/2022] Open
Abstract
Penile cancer (PeCa) is a relatively rare tumor entity but possesses higher morbidity and mortality rates especially in developing countries. To date, the concrete pathogenic signaling pathways and core machineries involved in tumorigenesis and progression of PeCa remain to be elucidated. Several studies suggested miRNAs, which modulate gene expression at posttranscriptional level, were frequently mis-regulated and aberrantly expressed in human cancers. However, the miRNA profile in human PeCa has not been reported before. In this present study, the miRNA profile was obtained from 10 fresh penile cancerous tissues and matched adjacent non-cancerous tissues via next-generation sequencing. As a result, a total of 751 and 806 annotated miRNAs were identified in normal and cancerous penile tissues, respectively. Among which, 56 miRNAs with significantly different expression levels between paired tissues were identified. Subsequently, several annotated miRNAs were selected randomly and validated using quantitative real-time PCR. Compared with the previous publications regarding to the altered miRNAs expression in various cancers and especially genitourinary (prostate, bladder, kidney, testis) cancers, the most majority of deregulated miRNAs showed the similar expression pattern in penile cancer. Moreover, the bioinformatics analyses suggested that the putative target genes of differentially expressed miRNAs between cancerous and matched normal penile tissues were tightly associated with cell junction, proliferation, growth as well as genomic instability and so on, by modulating Wnt, MAPK, p53, PI3K-Akt, Notch and TGF-β signaling pathways, which were all well-established to participate in cancer initiation and progression. Our work presents a global view of the differentially expressed miRNAs and potentially regulatory networks of their target genes for clarifying the pathogenic transformation of normal penis to PeCa, which research resource also provides new insights into future investigations aimed to explore the in-depth mechanisms of miRNAs and other small RNAs including piRNAs in penile carcinogenesis regulation and effective target-specific theragnosis.
Collapse
Affiliation(s)
- Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Pengfei Wei
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Xudong Shen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Yuanwei Zhang
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Bo Xu
- Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Song Fan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Haoqiang Shi
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Xiansheng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Rui Kong
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Lingfan Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Jingjing Gao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Duohong Zou
- Key Laboratory of Oral Diseases Research of Anhui Province, Stomatologic College, Anhui Medical University, Hefei, Anhui, China
- * E-mail: (CZL); (DHZ)
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- * E-mail: (CZL); (DHZ)
| |
Collapse
|
223
|
Cho SH, Ko JJ, Kim JO, Jeon YJ, Yoo JK, Oh J, Oh D, Kim JW, Kim NK. 3'-UTR Polymorphisms in the MiRNA Machinery Genes DROSHA, DICER1, RAN, and XPO5 Are Associated with Colorectal Cancer Risk in a Korean Population. PLoS One 2015; 10:e0131125. [PMID: 26147304 PMCID: PMC4492935 DOI: 10.1371/journal.pone.0131125] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs play an important role in cancer initiation and development. The aim of this study was to investigate whether polymorphisms in miRNA machinery genes are associated with the development of colorectal cancer (CRC). RAN rs14035 CT heterozygotes and T allele carriers (CT + TT) genotypes had lower risk of CRC, while the DICER1 rs3742330, DROSHA rs10719, and XPO5 rs11077 polymorphisms were not associated with CRC in the full study sample. Specifically, male RAN rs14035 CT heterozygotes and XPO5 rs11077 AA genotype (CT/AA) carriers experienced reduced CRC susceptibility (both colon and rectal). Subgroup analysis demonstrated that the combined RAN rs14035 CT + TT genotype was associated with rectal cancer, but not colon cancer. In addition, the DICER1 rs3742330 AG genotype was associated with a significantly increased risk of colon cancer. Stratified analysis revealed the RAN rs14035 combined CT+TT genotype was associated with decreased CRC risk in male patients without diabetes mellitus (DM) and in patients with rectal cancer. In addition, we found the RAN rs14035 CC genotype was related to a decreased risk of CRC with respect to tumor size and metabolism of homocysteine and folate. Furthermore, patients diagnosed with hypertension or DM who carried the DROSHA rs10719 CC genotype showed increased CRC risk, while the XPO5 rs11077 AC+CC genotype led to increased CRC risk in patients with hypertension only. Our results indicate variations in RAN rs14035, DICER1 rs3742330, XPO5 rs11077, and DROSHA rs10719 of Korean patients are significantly associated with their risk of CRC.
Collapse
Affiliation(s)
- Sung Hwan Cho
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jung Jae Ko
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Jung Oh Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Young Joo Jeon
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jung Ki Yoo
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Jisu Oh
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Doyeun Oh
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jong Woo Kim
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
- * E-mail: (NKK); (JWK)
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
- Institute for Clinical Research, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
- * E-mail: (NKK); (JWK)
| |
Collapse
|
224
|
Elton TS, Yalowich JC. Experimental procedures to identify and validate specific mRNA targets of miRNAs. EXCLI JOURNAL 2015; 14:758-90. [PMID: 27047316 PMCID: PMC4817421 DOI: 10.17179/excli2015-319] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 05/20/2015] [Indexed: 12/14/2022]
Abstract
Functionally matured microRNAs (miRNAs) are small single-stranded non-coding RNA molecules which are emerging as important post-transcriptional regulators of gene expression and consequently are central players in many physiological and pathological processes. Since the biological roles of individual miRNAs will be dictated by the mRNAs that they regulate, the identification and validation of miRNA/mRNA target interactions is critical for our understanding of the regulatory networks governing biological processes. We promulgate the combined use of prediction algorithms, the examination of curated databases of experimentally supported miRNA/mRNA interactions, manual sequence inspection of cataloged miRNA binding sites in specific target mRNAs, and review of the published literature as a reliable practice for identifying and prioritizing biologically important miRNA/mRNA target pairs. Once a preferred miRNA/mRNA target pair has been selected, we propose that the authenticity of a functional miRNA/mRNA target pair be validated by fulfilling four well-defined experimental criteria. This review summarizes our current knowledge of miRNA biology, miRNA/mRNA target prediction algorithms, validated miRNA/mRNA target data bases, and outlines several experimental methods by which miRNA/mRNA targets can be authenticated. In addition, a case study of human endoglin is presented as an example of the utilization of these methodologies.
Collapse
Affiliation(s)
- Terry S Elton
- College of Pharmacy, Division of Pharmacology, The Ohio State University, Columbus, OH, USA
| | - Jack C Yalowich
- College of Pharmacy, Division of Pharmacology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
225
|
MicroRNA-138 regulates the balance of Th1/Th2 via targeting RUNX3 in psoriasis. Immunol Lett 2015; 166:55-62. [DOI: 10.1016/j.imlet.2015.05.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 05/13/2015] [Accepted: 05/25/2015] [Indexed: 11/18/2022]
|
226
|
Suzuki HI, Katsura A, Yasuda T, Ueno T, Mano H, Sugimoto K, Miyazono K. Small-RNA asymmetry is directly driven by mammalian Argonautes. Nat Struct Mol Biol 2015; 22:512-21. [PMID: 26098316 DOI: 10.1038/nsmb.3050] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/26/2015] [Indexed: 12/17/2022]
Abstract
Asymmetric selection of single-stranded guide RNAs from double-stranded RNA precursors is crucial in RNA silencing-mediated gene regulation. However, the precise mechanisms of small-RNA asymmetry are unclear, especially because asymmetric selection can still occur when the putative asymmetry sensors Drosophila R2D2 and mammalian Dicer are depleted. Here we report a direct contribution of mammalian Argonaute 2 (Ago2) to microRNA (miRNA) asymmetry. Ago2 selects strands with 5'-uridine or 5'-adenosine and thermodynamically unstable 5' ends in parallel through its two sensor regions, which contact the 5' nucleobases and 5'-phosphates of prospective guide strands. Hence, miRNA asymmetry shows superposed patterns reflecting 5'-end nucleotide identity ('digital' pattern) and thermodynamic stability ('analog' pattern). Furthermore, we demonstrate that cancer-associated miRNA variations reprogram asymmetric selection. Finally, our study presents a model of this universal principle, to aid in comprehensive understanding of miRNA function and development of new RNA-silencing therapies in precision medicine.
Collapse
Affiliation(s)
- Hiroshi I Suzuki
- 1] David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA. [2] Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Katsura
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiko Yasuda
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihide Ueno
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Mano
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Koichi Sugimoto
- Department of Hematology and Oncology, JR Tokyo General Hospital, Tokyo, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
227
|
Chawla JPS, Iyer N, Soodan KS, Sharma A, Khurana SK, Priyadarshni P. Role of miRNA in cancer diagnosis, prognosis, therapy and regulation of its expression by Epstein-Barr virus and human papillomaviruses: With special reference to oral cancer. Oral Oncol 2015; 51:731-7. [PMID: 26093389 DOI: 10.1016/j.oraloncology.2015.05.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/24/2015] [Accepted: 05/26/2015] [Indexed: 01/15/2023]
Abstract
MicroRNAs (miRNAs) belong to class of small non-coding RNAs that regulate numerous biological processes by targeting broad set of messenger RNAs. Research on miRNA-based biomarkers has witnessed phenomenal growth, owing to non-invasive nature of miRNA based screening assays and their sensitivity and specificity in detecting cancers. Their discovery in humans in 2000 has led to an explosion in research in terms of their role as biomarker, therapeutic target and trying to elucidate their function. This review aims to summarize the function of microRNAs as well as to examine how dysregulation at any step in their biogenesis or functional pathway can play a role in development of cancer, together with its possible involvement in oral cancer. Overexpression of oncogenic miRNA may reduce protein products of tumor-suppressor genes but loss of tumor-suppressor miRNA expression may cause elevated levels of oncogenic protein. One or both of these alterations could represent new targets for cancer diagnosis and treatment in future. Many researchers have focused on genetic and epigenetic alterations in OSCC cells. The genetic susceptibility, endemic environment factors, and Epstein-Barr virus (EBV) infection are believed to be the major etiologic factors of OSCC. Once metastasis occurs, prognosis is very poor. It is urgently needed to develop biomarkers for early clinical diagnosis/prognosis, and novel effective therapies for oral carcinoma. High-risk HPV infection leads to aberrant expression of cellular oncogenic and tumor suppressive miRNAs. The emergence of miRNA knowledge, and its potential interactive action with such alterations, therefore creates new understanding of cell transformation.
Collapse
Affiliation(s)
- Jatinder Pal Singh Chawla
- Department of Oral and Maxillofacial Surgery, M.M. College of Dental Sciences & Research, MMU, Mullana, Ambala, Haryana, India.
| | - Nageshwar Iyer
- Department of Oral and Maxillofacial Surgery, M.M. College of Dental Sciences & Research, MMU, Mullana, Ambala, Haryana, India
| | - Kanwaldeep Singh Soodan
- Department of Oral and Maxillofacial Surgery, M.M. College of Dental Sciences & Research, MMU, Mullana, Ambala, Haryana, India
| | - Atul Sharma
- Department of Oral and Maxillofacial Surgery, M.M. College of Dental Sciences & Research, MMU, Mullana, Ambala, Haryana, India
| | - Sunpreet Kaur Khurana
- Department of Endodontics and Conservative Dentistry, Swami Devi Dyal Dental College and Hospital, Panchkula, Haryana, India
| | - Pratiksha Priyadarshni
- Department of Oral and Maxillofacial Surgery, M.M. College of Dental Sciences & Research, MMU, Mullana, Ambala, Haryana, India
| |
Collapse
|
228
|
Bobbin ML, Burnett JC, Rossi JJ. RNA interference approaches for treatment of HIV-1 infection. Genome Med 2015; 7:50. [PMID: 26019725 PMCID: PMC4445287 DOI: 10.1186/s13073-015-0174-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 05/13/2015] [Indexed: 01/05/2023] Open
Abstract
HIV/AIDS is a chronic and debilitating disease that cannot be cured with current antiretroviral drugs. While combinatorial antiretroviral therapy (cART) can potently suppress HIV-1 replication and delay the onset of AIDS, viral mutagenesis often leads to viral escape from multiple drugs. In addition to the pharmacological agents that comprise cART drug cocktails, new biological therapeutics are reaching the clinic. These include gene-based therapies that utilize RNA interference (RNAi) to silence the expression of viral or host mRNA targets that are required for HIV-1 infection and/or replication. RNAi allows sequence-specific design to compensate for viral mutants and natural variants, thereby drastically expanding the number of therapeutic targets beyond the capabilities of cART. Recent advances in clinical and preclinical studies have demonstrated the promise of RNAi therapeutics, reinforcing the concept that RNAi-based agents might offer a safe, effective, and more durable approach for the treatment of HIV/AIDS. Nevertheless, there are challenges that must be overcome in order for RNAi therapeutics to reach their clinical potential. These include the refinement of strategies for delivery and to reduce the risk of mutational escape. In this review, we provide an overview of RNAi-based therapies for HIV-1, examine a variety of combinatorial RNAi strategies, and discuss approaches for ex vivo delivery and in vivo delivery.
Collapse
Affiliation(s)
- Maggie L Bobbin
- Irell & Manella School of Biological Sciences, Beckman Research Institute of City of Hope, East Duarte Road, Duarte, CA 91010 USA
| | - John C Burnett
- Irell & Manella School of Biological Sciences, Beckman Research Institute of City of Hope, East Duarte Road, Duarte, CA 91010 USA ; Department of Molecular and Cell Biology, Beckman Research Institute of City of Hope, East Duarte Road, Duarte, CA 9101 USA
| | - John J Rossi
- Irell & Manella School of Biological Sciences, Beckman Research Institute of City of Hope, East Duarte Road, Duarte, CA 91010 USA ; Department of Molecular and Cell Biology, Beckman Research Institute of City of Hope, East Duarte Road, Duarte, CA 9101 USA
| |
Collapse
|
229
|
Cammaerts S, Strazisar M, De Rijk P, Del Favero J. Genetic variants in microRNA genes: impact on microRNA expression, function, and disease. Front Genet 2015; 6:186. [PMID: 26052338 PMCID: PMC4439572 DOI: 10.3389/fgene.2015.00186] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/05/2015] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression and like any other gene, their coding sequences are subject to genetic variation. Variants in miRNA genes can have profound effects on miRNA functionality at all levels, including miRNA transcription, maturation, and target specificity, and as such they can also contribute to disease. The impact of variants in miRNA genes is the focus of the present review. To put these effects into context, we first discuss the requirements of miRNA transcripts for maturation. In the last part an overview of available databases and tools and experimental approaches to investigate miRNA variants related to human disease is presented.
Collapse
Affiliation(s)
- Sophia Cammaerts
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, University of Antwerp Antwerp, Belgium
| | - Mojca Strazisar
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, University of Antwerp Antwerp, Belgium
| | - Peter De Rijk
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, University of Antwerp Antwerp, Belgium
| | - Jurgen Del Favero
- Applied Molecular Genomics Unit, Department of Molecular Genetics, VIB, University of Antwerp Antwerp, Belgium ; Multiplicom N.V., Niel Belgium
| |
Collapse
|
230
|
Aksoy-Aksel A, Zampa F, Schratt G. MicroRNAs and synaptic plasticity--a mutual relationship. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0515. [PMID: 25135976 DOI: 10.1098/rstb.2013.0515] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs (miRNAs) are rapidly emerging as central regulators of gene expression in the postnatal mammalian brain. Initial studies mostly focused on the function of specific miRNAs during the development of neuronal connectivity in culture, using classical gain- and loss-of-function approaches. More recently, first examples have documented important roles of miRNAs in plastic processes in intact neural circuits in the rodent brain related to higher cognitive abilities and neuropsychiatric disease. At the same time, evidence is accumulating that miRNA function itself is subjected to sophisticated control mechanisms engaged by the activity of neural circuits. In this review, we attempt to pay tribute to this mutual relationship between miRNAs and synaptic plasticity. In particular, in the first part, we summarize how neuronal activity influences each step in the lifetime of miRNAs, including the regulation of transcription, maturation, gene regulatory function and turnover in mammals. In the second part, we discuss recent examples of miRNA function in synaptic plasticity in rodent models and their implications for higher cognitive function and neurological disorders, with a special emphasis on epilepsy as a disorder of abnormal nerve cell activity.
Collapse
Affiliation(s)
- Ayla Aksoy-Aksel
- Institut für Physiologische Chemie, Biochemisch-Pharmakologisches Centrum Marburg, Philipps-Universität Marburg, 35032 Marburg, Germany
| | - Federico Zampa
- Institut für Physiologische Chemie, Biochemisch-Pharmakologisches Centrum Marburg, Philipps-Universität Marburg, 35032 Marburg, Germany
| | - Gerhard Schratt
- Institut für Physiologische Chemie, Biochemisch-Pharmakologisches Centrum Marburg, Philipps-Universität Marburg, 35032 Marburg, Germany
| |
Collapse
|
231
|
Fu X, Dong B, Tian Y, Lefebvre P, Meng Z, Wang X, Pattou F, Han W, Wang X, Lou F, Jove R, Staels B, Moore DD, Huang W. MicroRNA-26a regulates insulin sensitivity and metabolism of glucose and lipids. J Clin Invest 2015; 125:2497-509. [PMID: 25961460 DOI: 10.1172/jci75438] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/07/2015] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes (T2D) is characterized by insulin resistance and increased hepatic glucose production, yet the molecular mechanisms underlying these abnormalities are poorly understood. MicroRNAs (miRs) are a class of small, noncoding RNAs that have been implicated in the regulation of human diseases, including T2D. miR-26a is known to play a critical role in tumorigenesis; however, its function in cellular metabolism remains unknown. Here, we determined that miR-26a regulates insulin signaling and metabolism of glucose and lipids. Compared with lean individuals, overweight humans had decreased expression of miR-26a in the liver. Moreover, miR-26 was downregulated in 2 obese mouse models compared with control animals. Global or liver-specific overexpression of miR-26a in mice fed a high-fat diet improved insulin sensitivity, decreased hepatic glucose production, and decreased fatty acid synthesis, thereby preventing obesity-induced metabolic complications. Conversely, silencing of endogenous miR-26a in conventional diet-fed mice impaired insulin sensitivity, enhanced glucose production, and increased fatty acid synthesis. miR-26a targeted several key regulators of hepatic metabolism and insulin signaling. These findings reveal miR-26a as a regulator of liver metabolism and suggest miR-26a should be further explored as a potential target for the treatment of T2D.
Collapse
|
232
|
Segalla S, Pivetti S, Todoerti K, Chudzik MA, Giuliani EC, Lazzaro F, Volta V, Lazarevic D, Musco G, Muzi-Falconi M, Neri A, Biffo S, Tonon G. The ribonuclease DIS3 promotes let-7 miRNA maturation by degrading the pluripotency factor LIN28B mRNA. Nucleic Acids Res 2015; 43:5182-93. [PMID: 25925570 PMCID: PMC4446438 DOI: 10.1093/nar/gkv387] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 04/12/2015] [Indexed: 12/03/2022] Open
Abstract
Multiple myeloma, the second most frequent hematologic tumor after lymphomas, is an incurable cancer. Recent sequencing efforts have identified the ribonuclease DIS3 as one of the most frequently mutated genes in this disease. DIS3 represents the catalytic subunit of the exosome, a macromolecular complex central to the processing, maturation and surveillance of various RNAs. miRNAs are an evolutionarily conserved class of small noncoding RNAs, regulating gene expression at post-transcriptional level. Ribonucleases, including Drosha, Dicer and XRN2, are involved in the processing and stability of miRNAs. However, the role of DIS3 on the regulation of miRNAs remains largely unknown. Here we found that DIS3 regulates the levels of the tumor suppressor let-7 miRNAs without affecting other miRNA families. DIS3 facilitates the maturation of let-7 miRNAs by reducing in the cytoplasm the RNA stability of the pluripotency factor LIN28B, a inhibitor of let-7 processing. DIS3 inactivation, through the increase of LIN28B and the reduction of mature let-7, enhances the translation of let-7 targets such as MYC and RAS leading to enhanced tumorigenesis. Our study establishes that the ribonuclease DIS3, targeting LIN28B, sustains the maturation of let-7 miRNAs and suggests the increased translation of critical oncogenes as one of the biological outcomes of DIS3 inactivation.
Collapse
Affiliation(s)
- Simona Segalla
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20133 Milan, Italy
| | - Silvia Pivetti
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20133 Milan, Italy
| | - Katia Todoerti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Malgorzata Agata Chudzik
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20133 Milan, Italy
| | - Erica Claudia Giuliani
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20133 Milan, Italy
| | - Federico Lazzaro
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20132 Milan, Italy
| | - Viviana Volta
- Molecular Histology and Cell Growth Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Science Institute, 20132 Milan, Italy
| | - Dejan Lazarevic
- Center for Translational Genomics and Bioinformatics, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giovanna Musco
- Dulbecco Telethon Institute, S. Raffaele Hospital, 20132 Milan, Italy
| | - Marco Muzi-Falconi
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20132 Milan, Italy
| | - Antonino Neri
- Department of Clinical Sciences and Community Health, University of Milan, Hematology1 CTMO, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefano Biffo
- Molecular Histology and Cell Growth Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Science Institute, 20132 Milan, Italy Dipartimento di Scienze e Innovazione Tecnologica, University of Piemonte Orientale, 15100 Alessandria, Italy
| | - Giovanni Tonon
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20133 Milan, Italy Università Vita-Salute San Raffaele, Milan, 20132, Italy
| |
Collapse
|
233
|
Xu J, Zhang W, Lv Q, Zhu D. Overexpression of miR-21 promotes the proliferation and migration of cervical cancer cells via the inhibition of PTEN. Oncol Rep 2015; 33:3108-16. [PMID: 25963606 DOI: 10.3892/or.2015.3931] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/04/2015] [Indexed: 11/06/2022] Open
Abstract
The oncogenic miR-21 has been widely recognized to promote the development and progression of various types of malignant tumors, but not cervical cancers. The aim of this study was to examine the expression of miR-21 and PTEN in cervical cancer specimens using quantitative PCR. The miR-21 level was then manipulated in the cervical cancer lines and the regulation of miR-21 on the proliferation, migration and invasion of cervical cancer cells was determined. Additionally, we determined the role of PTEN in the miR-21-regulated proliferation, migration and invasion of cervical cancer cells. miR-21 was upregulated in the cervical cancer specimens, negatively correlating with the PTEN mRNA level. Transfection of the miR-21 mimics was markedly promoted, whereas the miR-21 inhibitor suppressed the proliferation, migration and invasion of cervical cancer cells, with a significant inhibition of PTEN expression. In addition, the overexpression of PTEN markedly inhibited the proliferation and migration of the cervical cancer cells. The present study showed the upregulation of miR-21 in invasive cervical cancers, and confirmed the promotion of miR-21 with regard to the proliferation, migration and invasion in cervical cancer cells via inhibiting the PTEN expression. To the best of our knowledge, this is the first study to confirm that the miR-21/PTEN pathway promotes cervical cancer.
Collapse
Affiliation(s)
- Jingjie Xu
- The First Department of Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Zhang
- The First Department of Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qiongying Lv
- The First Department of Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Dingjun Zhu
- The First Department of Gynaecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
234
|
MicroRNA and Breast Cancer: Understanding Pathogenesis, Improving Management. Noncoding RNA 2015; 1:17-43. [PMID: 29861413 PMCID: PMC5932537 DOI: 10.3390/ncrna1010017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 04/10/2015] [Accepted: 04/14/2015] [Indexed: 12/14/2022] Open
Abstract
The advent of the microRNAs in the early 1990s has proven to be a tremendously significant development within the purview of gene regulation. They participate in the regulation of a broad assembly of processes vital to proper cell function and the perturbation of these pathways following alteration of miRNA expression is strongly believed to contribute to the pathogenesis of cancer. This review provides a comprehensive overview of the miRNAs that have to date been well-characterized in the context of human breast neoplasia. Detailed discussion will center around their role in tumor initiation and progression, control of epithelial-mesenchymal transition (EMT), cancer stem cell formation, use as biomarkers in tissues and circulation, as well as their role in cancer treatment. In addition, attention will be given to topics which remain underexplored, such as miRNA control of cancer cell metabolism and the genomic/epigenetic origins underlying the preliminary disruption of miRNA expression in disease. This review will also address and attempt to resolve instances where discordant, inter-study findings have been reported (examples of which are replete in the literature) while also identifying bottlenecks hampering progress in miRNA research and other challenges that confront this fledgling but promising field of biomedical research.
Collapse
|
235
|
García R, Nistal JF, Merino D, Price NL, Fernández-Hernando C, Beaumont J, González A, Hurlé MA, Villar AV. p-SMAD2/3 and DICER promote pre-miR-21 processing during pressure overload-associated myocardial remodeling. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1520-30. [PMID: 25887159 DOI: 10.1016/j.bbadis.2015.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/23/2015] [Accepted: 04/07/2015] [Indexed: 12/21/2022]
Abstract
Transforming growth factor-β (TGF-β) induces miR-21 expression which contributes to fibrotic events in the left ventricle (LV) under pressure overload. SMAD effectors of TGF-β signaling interact with DROSHA to promote primary miR-21 processing into precursor miR-21 (pre-miR-21). We hypothesize that p-SMAD-2 and -3 also interact with DICER1 to regulate the processing of pre-miR-21 to mature miR-21 in cardiac fibroblasts under experimental and clinical pressure overload. The subjects of the study were mice undergoing transverse aortic constriction (TAC) and patients with aortic stenosis (AS). In vitro, NIH-3T3 fibroblasts transfected with pre-miR-21 responded to TGF-β1 stimulation by overexpressing miR-21. Overexpression and silencing of SMAD2/3 resulted in higher and lower production of mature miR-21, respectively. DICER1 co-precipitated along with SMAD2/3 and both proteins were up-regulated in the LV from TAC-mice. Pre-miR-21 was isolated bound to the DICER1 maturation complex. Immunofluorescence analysis revealed co-localization of p-SMAD2/3 and DICER1 in NIH-3T3 and mouse cardiac fibroblasts. DICER1-p-SMAD2/3 protein-protein interaction was confirmed by in situ proximity ligation assay. Myocardial up-regulation of DICER1 constituted a response to pressure overload in TAC-mice. DICER mRNA levels correlated directly with those of TGF-β1, SMAD2 and SMAD3. In the LV from AS patients, DICER mRNA was up-regulated and its transcript levels correlated directly with TGF-β1, SMAD2, and SMAD3. Our results support that p-SMAD2/3 interacts with DICER1 to promote pre-miR-21 processing to mature miR-21. This new TGFβ-dependent regulatory mechanism is involved in miR-21 overexpression in cultured fibroblasts, and in the pressure overloaded LV of mice and human patients.
Collapse
Affiliation(s)
- Raquel García
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - J Francisco Nistal
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain; Servicio de Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - David Merino
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Nathan L Price
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Javier Beaumont
- Programa de Enfermedades Cardiovasculares, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Arantxa González
- Programa de Enfermedades Cardiovasculares, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - María A Hurlé
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain.
| | - Ana V Villar
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, Santander, Spain; Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| |
Collapse
|
236
|
Li X, Zhao Z. MicroRNA biomarkers for early detection of embryonic malformations in pregnancy. JOURNAL OF BIOMOLECULAR RESEARCH & THERAPEUTICS 2015; 3. [PMID: 25859419 DOI: 10.4172/2167-7956.1000119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Congenital birth defects, manifested in newborn infants, are formed during early embryogenesis. Targeted and individualized interventions to prevent birth defects require early detection of risk and signs of developmental abnormalities. Current diagnosis of structural anomalies largely relies on ultrasonography, which can only detect abnormities after their formation in fetuses. Biomolecules, mainly proteins, in maternal blood have been used as indicators of fetal anomalies; however, they lack adequate sensitivity for detecting embryonic malformations. Recently, cell-free microRNAs (miRNAs) have been found in blood and evaluated as biomarkers for diseases. Expression of certain miRNAs in maternal plasma has been shown to be correlated with birth defects in infants. Although their reliability and sensitivity remain to be validated, miRNAs, which can be amplified and sequenced, are potentially sensitive and specific biomarkers for early embryonic dysmorphogenesis.
Collapse
Affiliation(s)
- Xuezheng Li
- Department of Pharmacy, Affiliated Hospital of Yanbian University, Yanji, Jilin, China
| | - Zhiyong Zhao
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
237
|
Song C, Chen H, Wang T, Zhang W, Ru G, Lang J. Expression profile analysis of microRNAs in prostate cancer by next-generation sequencing. Prostate 2015; 75:500-16. [PMID: 25597612 DOI: 10.1002/pros.22936] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/03/2014] [Indexed: 12/29/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is the second leading cause of tumor mortality among males in western societies. In China, the diagnostic and fatality rate of PCa is increasing yearly. METHODS To characterize underlying molecular mechanisms, the microRNA (miRNA) profile of high-grade PCa, low-grade PCa, and benign prostate hyperplasia (BPH) were compared using high-throughput Illumina sequencing and quantitative real-time PCR (qRT-PCR) methods. Moreover, a variety of biological information softwares and databases were applied to predict the target genes of miRNA, molecular functions, and signal pathways. RESULTS Eighteen miRNAs were differentially expressed (fold change ≥ 2, P < 0.05), of which thirteen were upregulated and five were downregulated by sequencing. This was confirmed by qRT-PCR in more clinical tissue samples. In the tumors, miRNAs (miR-125b-5p, miR-126-5p, miR-151a-5p, miR-221-3p, and miR-222-3p) were significantly upregulated with downregulation of miR-486-5p. In addition, 13 novel miRNAs were identified from three prostate tissue libraries, with 12 of them assayed in 21 human normal tissues by qRT-PCR. Multiple databases indicated target genes for these differentially expressed miRNAs. Function annotation of target genes indicated that most of them tend to target genes involved in signal transduction and cell communication, especially cancer-related PI3K-Akt and p53 signaling pathway. CONCLUSIONS The small RNA transcriptomes obtained in this study uncovers six differentially expressed miRNAs and 12 novel miRNAs, and provides a better understanding of the expression and function of miRNAs in the development of PCa and reveals several miRNAs in PCa that may have biomarker and therapeutic potentials.
Collapse
Affiliation(s)
- Chunjiao Song
- Medical Research Center, Shaoxing people's Hospital, Shaoxing, China
| | | | | | | | | | | |
Collapse
|
238
|
Song K, Han C, Dash S, Balart LA, Wu T. MiR-122 in hepatitis B virus and hepatitis C virus dual infection. World J Hepatol 2015; 7:498-506. [PMID: 25848473 PMCID: PMC4381172 DOI: 10.4254/wjh.v7.i3.498] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/06/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) and hepatitis C virus (HCV) infections are the most common causes of chronic liver diseases and hepatocelluar carcinomas. Over the past few years, the liver-enriched microRNA-122 (miR-122) has been shown to differentially regulate viral replication of HBV and HCV. It is notable that the level of miR-122 is positively and negatively regulated by HCV and HBV, respectively. Consistent with the well-documented phenomenon that miR-122 promotes HCV accumulation, inhibition of miR-122 has been shown as an effective therapy for the treatment of HCV infection in both chimpanzees and humans. On the other hand, miR-122 is also known to block HBV replication, and HBV has recently been shown to inhibit miR-122 expression; such a reciprocal inhibition between miR-122 and HBV suggests an intriguing possibility that miR-122 replacement may represent a potential therapy for treatment of HBV infection. As HBV and HCV have shared transmission routes, dual infection is not an uncommon scenario, which is associated with more advanced liver disease than either HBV or HCV mono-infection. Thus, there is a clear need to further understand the interaction between HBV and HCV and to delineate the role of miR-122 in HBV/HCV dual infection in order to devise effective therapy. This review summarizes the current understanding of HBV/HCV dual infection, focusing on the pathobiological role and therapeutic potential of miR-122.
Collapse
|
239
|
Liu L, Ouyang M, Rao JN, Zou T, Xiao L, Chung HK, Wu J, Donahue JM, Gorospe M, Wang JY. Competition between RNA-binding proteins CELF1 and HuR modulates MYC translation and intestinal epithelium renewal. Mol Biol Cell 2015; 26:1797-810. [PMID: 25808495 PMCID: PMC4436827 DOI: 10.1091/mbc.e14-11-1500] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/16/2015] [Indexed: 12/17/2022] Open
Abstract
The mammalian intestinal epithelium is one of the most rapidly self-renewing tissues in the body, and its integrity is preserved through strict regulation. The RNA-binding protein (RBP) ELAV-like family member 1 (CELF1), also referred to as CUG-binding protein 1 (CUGBP1), regulates the stability and translation of target mRNAs and is implicated in many aspects of cellular physiology. We show that CELF1 competes with the RBP HuR to modulate MYC translation and regulates intestinal epithelial homeostasis. Growth inhibition of the small intestinal mucosa by fasting in mice was associated with increased CELF1/Myc mRNA association and decreased MYC expression. At the molecular level, CELF1 was found to bind the 3'-untranslated region (UTR) of Myc mRNA and repressed MYC translation without affecting total Myc mRNA levels. HuR interacted with the same Myc 3'-UTR element, and increasing the levels of HuR decreased CELF1 binding to Myc mRNA. In contrast, increasing the concentrations of CELF1 inhibited formation of the [HuR/Myc mRNA] complex. Depletion of cellular polyamines also increased CELF1 and enhanced CELF1 association with Myc mRNA, thus suppressing MYC translation. Moreover, ectopic CELF1 overexpression caused G1-phase growth arrest, whereas CELF1 silencing promoted cell proliferation. These results indicate that CELF1 represses MYC translation by decreasing Myc mRNA association with HuR and provide new insight into the molecular functions of RBPs in the regulation of intestinal mucosal growth.
Collapse
Affiliation(s)
- Lan Liu
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Miao Ouyang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Jaladanki N Rao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Tongtong Zou
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Hee Kyoung Chung
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Jing Wu
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - James M Donahue
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201 Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201 Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
240
|
|
241
|
Chung SH, Gillies M, Sugiyama Y, Zhu L, Lee SR, Shen W. Profiling of microRNAs involved in retinal degeneration caused by selective Müller cell ablation. PLoS One 2015; 10:e0118949. [PMID: 25741709 PMCID: PMC4351074 DOI: 10.1371/journal.pone.0118949] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 01/26/2015] [Indexed: 12/21/2022] Open
Abstract
Dysfunction of Müller cells has been implicated in the pathogenesis of several retinal diseases. In order to understand the potential contribution of Müller cells to retinal disease better, we have developed a transgenic model in which foci of Müller cell ablation can be selectively induced. MicroRNAs (miRNAs), small non-coding RNAs that are involved in post-transcriptional modulation, have critical functions in various biological processes. The aim of this study was to profile differential expression of miRNAs and to examine changes in their target genes 2 weeks after Müller cell ablation. We identified 20 miRNAs using the miScript HC PCR array. Data analysis using two target gene prediction databases (TargetScan and mirTarBase) revealed 78 overlapping target genes. DAVID and KEGG pathway analysis suggested that the target genes were generally involved in cell apoptosis, p53, neurotrophin, calcium, chemokine and Jak-STAT signalling pathways. Changes in seven target genes including Cyclin D2, Caspase 9, insulin-like growth factor 1, IL-1 receptor-associated kinase (IRAK), calmodulin (CALM) and Janus kinase 2 (Jak2), were validated with qRT-PCR and western blots. The cellular localisation of cleaved-caspase 9, Cyclin D2, Jak2 and CALM was examined by immunofluorescence studies. We found that the transcription of some miRNAs was positively, rather than negatively, correlated with their target genes. After confirming that overexpressed miR-133a-3p was localised to the outer nuclear layer in the damaged retina, we validated the correlation between miR-133a-3p and one of its predicted target genes, cyclin D2, with a luciferase assay in 661 photoreceptor cells. Results revealed by miRNA profiling, target gene analysis and validation were generally consistent with our previous findings that selective Müller cell ablation causes photoreceptor degeneration and neuroinflammation. Our data on alterations of miRNAs and their target gene expression after Müller cell ablation provide further insights into the potential role of Müller cell dysfunction in retinal disease.
Collapse
Affiliation(s)
- Sook Hyun Chung
- Macular Research Group, Clinical Ophthalmology and Eye Health, Save Sight Institute, University of Sydney, Sydney, Australia
- * E-mail:
| | - Mark Gillies
- Macular Research Group, Clinical Ophthalmology and Eye Health, Save Sight Institute, University of Sydney, Sydney, Australia
| | - Yuki Sugiyama
- Lens Research Group, Clinical Ophthalmology and Eye Health, Save Sight Institute, University of Sydney, Sydney, Australia
| | - Ling Zhu
- Macular Research Group, Clinical Ophthalmology and Eye Health, Save Sight Institute, University of Sydney, Sydney, Australia
| | - So-Ra Lee
- Macular Research Group, Clinical Ophthalmology and Eye Health, Save Sight Institute, University of Sydney, Sydney, Australia
| | - Weiyong Shen
- Macular Research Group, Clinical Ophthalmology and Eye Health, Save Sight Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
242
|
Baril P, Ezzine S, Pichon C. Monitoring the spatiotemporal activities of miRNAs in small animal models using molecular imaging modalities. Int J Mol Sci 2015; 16:4947-72. [PMID: 25749473 PMCID: PMC4394458 DOI: 10.3390/ijms16034947] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/17/2015] [Accepted: 02/17/2015] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate gene expression by binding mRNA targets via sequence complementary inducing translational repression and/or mRNA degradation. A current challenge in the field of miRNA biology is to understand the functionality of miRNAs under physiopathological conditions. Recent evidence indicates that miRNA expression is more complex than simple regulation at the transcriptional level. MiRNAs undergo complex post-transcriptional regulations such miRNA processing, editing, accumulation and re-cycling within P-bodies. They are dynamically regulated and have a well-orchestrated spatiotemporal localization pattern. Real-time and spatio-temporal analyses of miRNA expression are difficult to evaluate and often underestimated. Therefore, important information connecting miRNA expression and function can be lost. Conventional miRNA profiling methods such as Northern blot, real-time PCR, microarray, in situ hybridization and deep sequencing continue to contribute to our knowledge of miRNA biology. However, these methods can seldom shed light on the spatiotemporal organization and function of miRNAs in real-time. Non-invasive molecular imaging methods have the potential to address these issues and are thus attracting increasing attention. This paper reviews the state-of-the-art of methods used to detect miRNAs and discusses their contribution in the emerging field of miRNA biology and therapy.
Collapse
Affiliation(s)
- Patrick Baril
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, 45071 Orléans, France.
| | - Safia Ezzine
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, 45071 Orléans, France.
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Université d'Orléans, 45071 Orléans, France.
| |
Collapse
|
243
|
Zhang Y, Zeng CJ, He L, Ding L, Tang KY, Peng WP. Selection of endogenous reference microRNA genes for quantitative reverse transcription polymerase chain reaction studies of boar spermatozoa cryopreservation. Theriogenology 2015; 83:634-41. [DOI: 10.1016/j.theriogenology.2014.10.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 10/28/2014] [Accepted: 10/28/2014] [Indexed: 10/24/2022]
|
244
|
Kong X, Yu J, Bi J, Qi H, Di W, Wu L, Wang L, Zha J, Lv S, Zhang F, Li Y, Hu F, Liu F, Zhou H, Liu J, Ding G. Glucocorticoids transcriptionally regulate miR-27b expression promoting body fat accumulation via suppressing the browning of white adipose tissue. Diabetes 2015; 64:393-404. [PMID: 25187367 PMCID: PMC4876791 DOI: 10.2337/db14-0395] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Long-term glucocorticoid (GC) treatment induces central fat accumulation and metabolic dysfunction. We demonstrate that microRNA-27b (miR-27b) plays a central role in the pathogenesis of GC-induced central fat accumulation. Overexpression of miR-27b had the same effects as dexamethasone (DEX) treatment on the inhibition of brown adipose differentiation and the energy expenditure of primary adipocytes. Conversely, antagonizing miR-27b function prevented DEX suppression of the expression of brown adipose tissue-specific genes. GCs transcriptionally regulate miR-27b expression through a GC receptor-mediated direct DNA-binding mechanism, and miR-27b suppresses browning of white adipose tissue (WAT) by targeting the three prime untranslated region of Prdm16. In vivo, antagonizing miR-27b function in DEX-treated mice resulted in the efficient induction of brown adipocytes within WAT and improved GC-induced central fat accumulation. Collectively, these results indicate that miR-27b functions as a central target of GC and as an upstream regulator of Prdm16 to control browning of WAT and, consequently, may represent a potential target in preventing obesity.
Collapse
Affiliation(s)
- Xiaocen Kong
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Jing Yu
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Jianhua Bi
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Hanmei Qi
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Wenjuan Di
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Lin Wu
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Long Wang
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Juanmin Zha
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Shan Lv
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Feng Zhang
- Department of General Surgery, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Yan Li
- Metabolic Syndrome Research Center of Central South University, Institute of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Fang Hu
- Metabolic Syndrome Research Center of Central South University, Institute of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Feng Liu
- Metabolic Syndrome Research Center of Central South University, Institute of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Juan Liu
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Guoxian Ding
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
245
|
Chang YL, Ho BC, Sher S, Yu SL, Yang PC. miR-146a and miR-370 coordinate enterovirus 71-induced cell apoptosis through targeting SOS1 and GADD45β. Cell Microbiol 2015; 17:802-18. [PMID: 25469565 DOI: 10.1111/cmi.12401] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 11/20/2014] [Accepted: 11/24/2014] [Indexed: 12/22/2022]
Abstract
Enterovirus 71 (EV71) is an emerging life-threatening pathogen particularly in the Asia-Pacific region. The major pathogenic feature in EV71-infected cells is apoptosis. However, which molecular mechanism mainly contributes to EV71-induced apoptosis is not investigated thoroughly. MicroRNAs (MiRNAs), the newly discovered molecules, govern a wide range of biological functions through post-transcriptional regulation including viral pathogenesis. Whether miRNAs and messenger RNAs (mRNAs) coordinate to trigger host cell apoptosis in EV71 infection was investigated in this study. We conducted an apoptosis-oriented approach using both mRNA and miRNA profiling and bioinformatic analysis. We identified two major apoptosis-associated signalling pathways, Bcl2 antagonist of cell death (BAD) phosphorylation and p53-dependent apoptosis pathways, in which Son of sevenless homolog 1 (SOS1) and Growth arrest and DNA damage-inducible protein 45β (GADD45β) were predicted as targets of miR-146a and miR-370 respectively. Luciferase reporter assays and Western blots demonstrated the negative regulation between miR-146a and SOS1 and between miR-370 and GADD45β. Silencing of miR-146a restored SOS1 expression and partially attenuated EV71 infection-induced apoptosis. Conversely, ectopic expression of miR-370 decreased virus infection-induced GADD45β expression and also diminished apoptosis. Finally, the transfection of antagomiR-146a and miR-370 contributed to attenuating EV71 infection-induced apoptosis. Herein we clearly demonstrate that EV71-induced cell apoptosis is partly governed by altered miRNAs.
Collapse
Affiliation(s)
- Ya-Ling Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bing-Ching Ho
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.,NTU Center for Genomic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Singh Sher
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.,NTU Center for Genomic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pathology and Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Center for Optoelectronic Biomedicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pan-Chyr Yang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,NTU Center for Genomic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
246
|
Telomerase reverse transcriptase regulates microRNAs. Int J Mol Sci 2015; 16:1192-208. [PMID: 25569094 PMCID: PMC4307298 DOI: 10.3390/ijms16011192] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 12/26/2014] [Indexed: 12/30/2022] Open
Abstract
MicroRNAs are small non-coding RNAs that inhibit the translation of target mRNAs. In humans, most microRNAs are transcribed by RNA polymerase II as long primary transcripts and processed by sequential cleavage of the two RNase III enzymes, DROSHA and DICER, into precursor and mature microRNAs, respectively. Although the fundamental functions of microRNAs in RNA silencing have been gradually uncovered, less is known about the regulatory mechanisms of microRNA expression. Here, we report that telomerase reverse transcriptase (TERT) extensively affects the expression levels of mature microRNAs. Deep sequencing-based screens of short RNA populations revealed that the suppression of TERT resulted in the downregulation of microRNAs expressed in THP-1 cells and HeLa cells. Primary and precursor microRNA levels were also reduced under the suppression of TERT. Similar results were obtained with the suppression of either BRG1 (also called SMARCA4) or nucleostemin, which are proteins interacting with TERT and functioning beyond telomeres. These results suggest that TERT regulates microRNAs at the very early phases in their biogenesis, presumably through non-telomerase mechanism(s).
Collapse
|
247
|
Franzoni E, Booker SA, Parthasarathy S, Rehfeld F, Grosser S, Srivatsa S, Fuchs HR, Tarabykin V, Vida I, Wulczyn FG. miR-128 regulates neuronal migration, outgrowth and intrinsic excitability via the intellectual disability gene Phf6. eLife 2015; 4. [PMID: 25556700 PMCID: PMC4337614 DOI: 10.7554/elife.04263] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 12/31/2014] [Indexed: 12/13/2022] Open
Abstract
miR-128, a brain-enriched microRNA, has been implicated in the control of neurogenesis and synaptogenesis but its potential roles in intervening processes have not been addressed. We show that post-transcriptional mechanisms restrict miR-128 accumulation to post-mitotic neurons during mouse corticogenesis and in adult stem cell niches. Whereas premature miR-128 expression in progenitors for upper layer neurons leads to impaired neuronal migration and inappropriate branching, sponge-mediated inhibition results in overmigration. Within the upper layers, premature miR-128 expression reduces the complexity of dendritic arborization, associated with altered electrophysiological properties. We show that Phf6, a gene mutated in the cognitive disorder Börjeson-Forssman-Lehmann syndrome, is an important regulatory target for miR-128. Restoring PHF6 expression counteracts the deleterious effect of miR-128 on neuronal migration, outgrowth and intrinsic physiological properties. Our results place miR-128 upstream of PHF6 in a pathway vital for cortical lamination as well as for the development of neuronal morphology and intrinsic excitability. DOI:http://dx.doi.org/10.7554/eLife.04263.001 The unique capabilities of the mammalian brain depend on the patterns formed by spatial arrangements and connections between millions (sometimes billions) of electrically active cells called neurons, and on the connections between these neurons. During the development of the cortex, the largest part of the brain, neurons are born in stem cell areas that lie deep inside the brain, and these newly made neurons then migrate outwards to their final positions close to the surface of the adult brain. Franzoni et al. have examined how two molecules, a small RNA called miR-128 and a protein called PHF6, control when and how neurons migrate through the cortex and then grow to form connections with other neurons as they mature. Mutations that disrupt PHF6 can cause intellectual disabilities, and one possible reason for this is that PHF6 is needed to ensure that the neurons migrate to the correction location. Franzoni et al. now show that miR-128 can reduce the production of PHF6 and is therefore responsible for controlling when and where PHF6 is active. Studying miR-128 in detail, they show that although an inactive precursor form of miR-128 is present in stem cells and migrating neurons, the active form of miR-128 is only found in neurons that have already reached their final position in the cortex. Franzoni et al. used genetic methods to override the switch that controls when miR-128 becomes active. When the amount of miR-128 was artificially reduced, the neurons migrated too far. Artificially increasing the amount of miR-128 had the opposite effect: both the movement of the neurons and, later, their growth were defective. PHF6 was the key to these effects: if PHF6 levels were kept close to normal, miR-128 could no longer interfere with the movement and growth of the neurons. Further work will be required to better understand how miR-128 is turned off and on, and how PHF6 acts to control neuronal movement and growth. DOI:http://dx.doi.org/10.7554/eLife.04263.002
Collapse
Affiliation(s)
- Eleonora Franzoni
- Institute for Cell and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sam A Booker
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Srinivas Parthasarathy
- Institute for Cell and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Frederick Rehfeld
- Institute for Cell and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sabine Grosser
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Swathi Srivatsa
- Institute for Cell and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heiko R Fuchs
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University, Düsseldorf, Germany
| | - Victor Tarabykin
- Institute for Cell and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Imre Vida
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - F Gregory Wulczyn
- Institute for Cell and Neurobiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
248
|
Duran-Figueroa N, Badillo-Corona JA, Naisbitt DJ, Castrejon-Flores JL. Towards the development of mechanism-based biomarkers to diagnose drug hypersensitivity. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00238e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
T-cells are activated by different mechanisms in the presence of drugs, metabolites or haptens, and they release several molecules that can be used in the diagnosis of drug hypersensitivity.
Collapse
Affiliation(s)
- N. Duran-Figueroa
- Instituto Politécnico Nacional
- Unidad Profesional Interdisciplinaria de Biotecnología
- Mexico City
- Mexico
| | - J. A. Badillo-Corona
- Instituto Politécnico Nacional
- Unidad Profesional Interdisciplinaria de Biotecnología
- Mexico City
- Mexico
| | - D. J. Naisbitt
- MRC Centre for Drug Safety Science
- Department of Pharmacology
- University of Liverpool
- Liverpool
- UK
| | - J. L. Castrejon-Flores
- Instituto Politécnico Nacional
- Unidad Profesional Interdisciplinaria de Biotecnología
- Mexico City
- Mexico
| |
Collapse
|
249
|
Aghanoori MR, Mirzaei B, Tavallaei M. MiRNA Molecular Profiles in Human Medical Conditions: Connecting Lung Cancer and Lung Development Phenomena. Asian Pac J Cancer Prev 2014; 15:9557-65. [DOI: 10.7314/apjcp.2014.15.22.9557] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
250
|
Lawless N, Vegh P, O'Farrelly C, Lynn DJ. The Role of microRNAs in Bovine Infection and Immunity. Front Immunol 2014; 5:611. [PMID: 25505900 PMCID: PMC4245999 DOI: 10.3389/fimmu.2014.00611] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/13/2014] [Indexed: 11/13/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small, non-coding RNAs that are recognized as critical regulators of immune gene expression during infection. Many immunologically significant human miRNAs have been found to be conserved in agriculturally important species, including cattle. Discovering how bovine miRNAs mediate the immune defense during infection is critical to understanding the etiology of the most prevalent bovine diseases. Here, we review current knowledge of miRNAs in the bovine genome, and discuss the advances in understanding of miRNAs as regulators of immune cell function, and bovine immune response activation, regulation, and resolution. Finally, we consider the future perspectives on miRNAs in bovine viral disease, their role as potential biomarkers and in therapy.
Collapse
Affiliation(s)
- Nathan Lawless
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany , Meath , Ireland ; School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland
| | - Peter Vegh
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany , Meath , Ireland ; School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College Dublin , Dublin , Ireland
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity College Dublin , Dublin , Ireland
| | - David J Lynn
- South Australian Health and Medical Research Institute, North Terrace , Adelaide, SA , Australia ; School of Medicine, Flinders University , Bedford Park, SA , Australia
| |
Collapse
|