2001
|
Wagner KD, Wagner N, Ghanbarian H, Grandjean V, Gounon P, Cuzin F, Rassoulzadegan M. RNA induction and inheritance of epigenetic cardiac hypertrophy in the mouse. Dev Cell 2008; 14:962-9. [PMID: 18539123 DOI: 10.1016/j.devcel.2008.03.009] [Citation(s) in RCA: 198] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 12/14/2007] [Accepted: 03/18/2008] [Indexed: 12/20/2022]
Abstract
Epigenetic regulation shapes normal and pathological mammalian development and physiology. Our previous work showed that Kit RNAs injected into fertilized mouse eggs can produce heritable epigenetic defects, or paramutations, with relevant loss-of-function pigmentation phenotypes, which affect adult phenotypes in multiple succeeding generations of mice. Here, we illustrate the relevance of paramutation to pathophysiology by injecting fertilized mouse eggs with RNAs targeting Cdk9, a key regulator of cardiac growth. Microinjecting fragments of either the coding region or the related microRNA miR-1 led to high levels of expression of homologous RNA, resulting in an epigenetic defect, cardiac hypertrophy, whose efficient hereditary transmission correlated with the presence of miR-1 in the sperm nucleus. In this case, paramutation increased rather than decreased expression of Cdk9. These results highlight the diversity of RNA-mediated epigenetic effects and may provide a paradigm for clinical cases of familial diseases whose inheritance is not fully explained in Mendelian terms.
Collapse
|
2002
|
Walker J, Harland R. Expression of microRNAs during embryonic development of Xenopus tropicalis. Gene Expr Patterns 2008; 8:452-456. [PMID: 18440877 PMCID: PMC2546408 DOI: 10.1016/j.gep.2008.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 03/15/2008] [Accepted: 03/25/2008] [Indexed: 11/16/2022]
Abstract
microRNAs (miRNAs) are short, non-coding RNAs that regulate gene expression and have prominent roles during early embryo development and organogenesis. We set out to determine the expression pattern of miRNAs in the developmental model system, Xenopus tropicalis. We made probes to predicted primary-miRNA transcripts and performed in situ hybridization. Our data show conserved and novel tissue-specific expression patterns during embryogenesis that suggest functional roles during development.
Collapse
Affiliation(s)
- James Walker
- Department of Molecular and Cell Biology, University of California, Berkeley CA 94720
| | - Richard Harland
- Department of Molecular and Cell Biology, University of California, Berkeley CA 94720
| |
Collapse
|
2003
|
Datta J, Kutay H, Nasser MW, Nuovo GJ, Wang B, Majumder S, Liu CG, Volinia S, Croce CM, Schmittgen TD, Ghoshal K, Jacob ST. Methylation mediated silencing of MicroRNA-1 gene and its role in hepatocellular carcinogenesis. Cancer Res 2008; 68:5049-58. [PMID: 18593903 PMCID: PMC2562630 DOI: 10.1158/0008-5472.can-07-6655] [Citation(s) in RCA: 374] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miR) are a class of small ( approximately 21 nucleotide) noncoding RNAs that, in general, negatively regulate gene expression. Some miRs harboring CGIs undergo methylation-mediated silencing, a characteristic of many tumor suppressor genes. To identify such miRs in liver cancer, the miRNA expression profile was analyzed in hepatocellular carcinoma (HCC) cell lines treated with 5-azacytidine (DNA hypomethylating agent) and/or trichostatin A (histone deacetylase inhibitor). The results showed that these epigenetic drugs differentially regulate expression of a few miRs, particularly miR-1-1, in HCC cells. The CGI spanning exon 1 and intron 1 of miR-1-1 was methylated in HCC cell lines and in primary human HCCs but not in matching liver tissues. The miR-1-1 gene was hypomethylated and activated in DNMT1-/- HCT 116 cells but not in DNMT3B null cells, indicating a key role for DNMT1 in its methylation. miR-1 expression was also markedly reduced in primary human hepatocellular carcinomas compared with matching normal liver tissues. Ectopic expression of miR-1 in HCC cells inhibited cell growth and reduced replication potential and clonogenic survival. The expression of FoxP1 and MET harboring three and two miR-1 cognate sites, respectively, in their respective 3'-untranslated regions, was markedly reduced by ectopic miR-1. Up-regulation of several miR-1 targets including FoxP1, MET, and HDAC4 in primary human HCCs and down-regulation of their expression in 5-AzaC-treated HCC cells suggest their role in hepatocarcinogenesis. The inhibition of cell cycle progression and induction of apoptosis after re-expression of miR-1 are some of the mechanisms by which DNA hypomethylating agents suppress hepatocarcinoma cell growth.
Collapse
Affiliation(s)
- Jharna Datta
- Deptartment of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Huban Kutay
- Deptartment of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Mohd W. Nasser
- Deptartment of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Gerard J. Nuovo
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| | - Bo Wang
- Deptartment of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Sarmila Majumder
- Deptartment of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Chang-Gong Liu
- Department of Molecular virology, Immunology and Medical Genetics, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Stefano Volinia
- Department of Molecular virology, Immunology and Medical Genetics, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Carlo M. Croce
- Department of Molecular virology, Immunology and Medical Genetics, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | | | - Kalpana Ghoshal
- Deptartment of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Samson T. Jacob
- Deptartment of Molecular and Cellular Biochemistry, Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
2004
|
Wang Z, Luo X, Lu Y, Yang B. miRNAs at the heart of the matter. J Mol Med (Berl) 2008; 86:771-783. [PMID: 18415070 PMCID: PMC2480593 DOI: 10.1007/s00109-008-0341-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 01/18/2008] [Accepted: 02/27/2008] [Indexed: 01/08/2023]
Abstract
Cardiovascular disease is among the main causes of morbidity and mortality in developed countries. The pathological process of the heart is associated with altered expression profile of genes that are important for cardiac function. MicroRNAs (miRNAs) have emerged as one of the central players of gene expression regulation. The implications of miRNAs in the pathological process of cardiovascular system have recently been recognized, representing the most rapidly evolving research field. Here, we summarize and analyze the currently available data from our own laboratory and other groups, providing a comprehensive overview of miRNA function in the heart, including a brief introduction of miRNA biology, expression profile of miRNAs in cardiac tissue, role of miRNAs in cardiac hypertrophy and heart failure, the arrhythmogenic potential of miRNAs, the involvement of miRNAs in vascular angiogenesis, and regulation of cardiomyocyte apoptosis by miRNAs. The target genes and signaling pathways linking the miRNAs to cardiovascular disease are highlighted. The applications of miRNA interference technologies for manipulating miRNA expression, stability, and function as new strategies for molecular therapy of human disease are evaluated. Finally, some specific issues related to future directions of the research on miRNAs relevant to cardiovascular disease are pinpointed and speculated.
Collapse
Affiliation(s)
- Zhiguo Wang
- Research Center, Montreal Heart Institute, Montreal, PQ H1T 1C8 Canada
- Department of Medicine, University of Montreal, Montreal, PQ H3C 3J7 Canada
- Cardiovascular Research Institute, Harbin Medical University, Harbin, Heilongjiang 150086 People’s Republic China
| | - Xioabin Luo
- Research Center, Montreal Heart Institute, Montreal, PQ H1T 1C8 Canada
- Department of Medicine, University of Montreal, Montreal, PQ H3C 3J7 Canada
| | - Yanjie Lu
- Department of Pharmacology (the State–Province Key Laboratories of Biomedicine–Pharmaceutics of China, Harbin Medical University, Harbin, Heilongjiang 150086 People’s Republic of China
- Cardiovascular Research Institute, Harbin Medical University, Harbin, Heilongjiang 150086 People’s Republic China
| | - Baofeng Yang
- Department of Pharmacology (the State–Province Key Laboratories of Biomedicine–Pharmaceutics of China, Harbin Medical University, Harbin, Heilongjiang 150086 People’s Republic of China
- Cardiovascular Research Institute, Harbin Medical University, Harbin, Heilongjiang 150086 People’s Republic China
| |
Collapse
|
2005
|
Hino K, Tsuchiya K, Fukao T, Kiga K, Okamoto R, Kanai T, Watanabe M. Inducible expression of microRNA-194 is regulated by HNF-1alpha during intestinal epithelial cell differentiation. RNA (NEW YORK, N.Y.) 2008; 14:1433-1442. [PMID: 18492795 PMCID: PMC2441992 DOI: 10.1261/rna.810208] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Accepted: 03/12/2008] [Indexed: 05/26/2023]
Abstract
Maintenance of the intestinal epithelium is based on well-balanced molecular mechanisms that confer the stable and continuous supply of specialized epithelial cell lineages from multipotent progenitors. Lineage commitment decisions in the intestinal epithelium system involve multiple regulatory systems that interplay with each other to establish the cellular identities. Here, we demonstrate that the microRNA system could be involved in intestinal epithelial cell differentiation, and that microRNA-194 (miR-194) is highly induced during this process. To investigate this inducible expression mechanism, we identified the genomic structure of the miR-194-2, -192 gene, one of the inducible class of miR-194 parental genes. Furthermore, we identified its transcriptional regulatory region that contains a consensus-binding motif for hepatocyte nuclear factor-1alpha (HNF-1alpha), which is well known as a transcription factor to regulate gene expression in intestinal epithelial cells. By chromatin immunoprecipitation assay and luciferase reporter analysis, we revealed that pri-miR-194-2 expression is controlled by HNF-1alpha, and its consensus binding region is required for the transcription of pri-miR-194-2 in vivo in an intestinal epithelial cell line, Caco-2. Our observations indicate that microRNA genes could be targets of lineage-specific transcription factors and that microRNAs are regulated by a tissue-specific manner in the intestinal epithelium. Therefore, our work suggests that induced expression of these microRNAs have important roles in intestinal epithelium maturation.
Collapse
Affiliation(s)
- Kimihiro Hino
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | | | | | | | | | | | | |
Collapse
|
2006
|
Abstract
MicroRNAs modulate target gene expression and are essential for normal development, but how does this pathway impact cell fate decisions? In this issue of Cell Stem Cell, Ivey et al. (2008) find that muscle-specific microRNAs repress nonmuscle genes to direct embryonic stem cell differentiation to mesoderm and muscle.
Collapse
Affiliation(s)
- Qintong Li
- Stem Cell Program, Children's Hospital Boston, Boston, MA 02115, USA
| | | |
Collapse
|
2007
|
A three-step pathway comprising PLZF/miR-146a/CXCR4 controls megakaryopoiesis. Nat Cell Biol 2008; 10:788-801. [PMID: 18568019 DOI: 10.1038/ncb1741] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 05/23/2008] [Indexed: 01/17/2023]
Abstract
MicroRNAs (miRNAs or miRs) regulate diverse normal and abnormal cell functions. We have identified a regulatory pathway in normal megakaryopoiesis, involving the PLZF transcription factor, miR-146a and the SDF-1 receptor CXCR4. In leukaemic cell lines PLZF overexpression downmodulated miR-146a and upregulated CXCR4 protein, whereas PLZF knockdown induced the opposite effects. In vitro assays showed that PLZF interacts with and inhibits the miR-146a promoter, and that miR-146a targets CXCR4 mRNA, impeding its translation. In megakaryopoietic cultures of CD34(+) progenitors, PLZF was upregulated, whereas miR-146a expression decreased and CXCR4 protein increased. MiR-146a overexpression and PLZF or CXCR4 silencing impaired megakaryocytic (Mk) proliferation, differentiation and maturation, as well as Mk colony formation. Mir-146a knockdown induced the opposite effects. Rescue experiments indicated that the effects of PLZF and miR-146a are mediated by miR-146a and CXCR4, respectively. Our data indicate that megakaryopoiesis is controlled by a cascade pathway, in which PLZF suppresses miR-146a transcription and thereby activates CXCR4 translation.
Collapse
|
2008
|
Wang CH, Lee DY, Deng Z, Jeyapalan Z, Lee SC, Kahai S, Lu WY, Zhang Y, Yang BB. MicroRNA miR-328 regulates zonation morphogenesis by targeting CD44 expression. PLoS One 2008; 3:e2420. [PMID: 18560585 PMCID: PMC2409976 DOI: 10.1371/journal.pone.0002420] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Accepted: 05/02/2008] [Indexed: 12/19/2022] Open
Abstract
Morphogenesis is crucial to initiate physiological development and tumor invasion. Here we show that a microRNA controls zonation morphogenesis by targeting hyaluronan receptor CD44. We have developed a novel system to study microRNA functions by generating constructs expressing pre-miRNAs and mature miRNAs. Using this system, we have demonstrated that expression of miR-328 reduced cell adhesion, aggregation, and migration, and regulated formation of capillary structure. Protein analysis indicated that miR-328 repressed CD44 expression. Activities of luciferase constructs harboring the target site in CD44, but not the one containing mutation, were repressed by miR-328. Zonation morphogenesis appeared in cells transfected by miR-328: miR-328-transfected cells were present on the surface of zonating structures while the control cells stayed in the middle. MiR-328-mediated CD44 actions was validated by anti-CD44 antibody, hyaluronidase, CD44 siRNA, and CD44 expression constructs. In vivo experiments showed that CD44-silencing cells appeared as layers on the surfaces of nodules or zonating structures. Immuno-histochemistry also exhibited CD44-negative cells on the surface layers of normal rat livers and the internal zones of Portal veins. Our results demonstrate that miR-328 targets CD44, which is essential in regulating zonation morphogenesis: silencing of CD44 expression is essential in sealing the zonation structures to facilitate their extension and to inhibit complex expansion.
Collapse
Affiliation(s)
- Chia-Hui Wang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Daniel Y. Lee
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Zhaoqun Deng
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Zina Jeyapalan
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Shao-Chen Lee
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Shireen Kahai
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Wei-Yang Lu
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Anaesthesia, University of Toronto, Toronto, Canada
| | - Yaou Zhang
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Burton B. Yang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- * E-mail:
| |
Collapse
|
2009
|
Fazi F, Nervi C. MicroRNA: basic mechanisms and transcriptional regulatory networks for cell fate determination. Cardiovasc Res 2008; 79:553-61. [PMID: 18539629 DOI: 10.1093/cvr/cvn151] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Characterization of regulatory mechanisms affecting microRNA (miRNA) expression and activity is providing novel clues for the identification of genes and complex regulatory circuits that determine cell and tissue specificity. Here, we review the molecular events leading to miRNA biogenesis and activity, focusing above all on endogenous and epigenetic transcriptional networks involving miRNA in early development, cellular lineage specification/differentiation of nervous, skeletal and cardiac muscle tissues and in haematopoiesis, as the de-regulation of such networks may be relevant to disease pathogenesis.
Collapse
Affiliation(s)
- Francesco Fazi
- Department of Histology and Medical Embryology, 'Sapienza' University of Rome, Italy
| | | |
Collapse
|
2010
|
Latronico MVG, Condorelli G. On the road to the definition of the cardiac miRNome in human disease states. J Mol Cell Cardiol 2008; 45:162-4. [PMID: 18619609 DOI: 10.1016/j.yjmcc.2008.05.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 05/27/2008] [Indexed: 11/15/2022]
|
2011
|
Abstract
Discovered less than a decade ago, micro-RNAs (miRNAs) have emerged as important regulators of gene expression in mammals. They consist of short nucleic acids, on average approximately 22 nucleotides in length. The miRNAs exert their effect by binding directly to target messenger RNAs (mRNAs) and inhibiting mRNA stability and translation. Each miRNA can bind to multiple targets and many miRNAs can bind to the same target mRNA, allowing for a complex pattern of regulation of gene expression. Once bound to their targets, miRNAs can suppress translation of the mRNA by either sequestration or degradation of the message. Thus, miRNAs function as powerful and sensitive posttranscriptional regulators of gene expression. This review will summarize what is known about miRNA biogenesis, expression, regulation, function, mode of action, and role in disease processes with an emphasis on miRNAs in mammals. We discuss some of the methodology employed in miRNA research and the potential of miRNAs as therapeutic targets. The role of miRNAs in signal transduction and cellular stress is reviewed. Lastly, we identify new exciting avenues of research on the role of miRNAs in toxicogenomics and the possibility of epigenetic effects on gene expression.
Collapse
Affiliation(s)
- Alice Hudder
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan 48201-2654
| | - Raymond F. Novak
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan 48201-2654
| |
Collapse
|
2012
|
Callis TE, Wang DZ. Taking microRNAs to heart. Trends Mol Med 2008; 14:254-60. [DOI: 10.1016/j.molmed.2008.03.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 03/19/2008] [Accepted: 03/19/2008] [Indexed: 01/04/2023]
|
2013
|
Abstract
MicroRNAs are a class of recently discovered small RNA molecules that regulate other genes in the human genome. Studies in human cells and model organisms have begun to reveal the mechanisms of microRNA activity, and the wide range of normal physiological functions they influence. Their alteration in pathologic states from cancer to cardiovascular disease is also increasingly clear. A review of current evidence for the role of these molecules in human health and disease will be helpful to pathologists and medical researchers as the fascinating story of these small regulators continues to unfold.
Collapse
Affiliation(s)
- Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-2297, USA.
| |
Collapse
|
2014
|
Thum T, Catalucci D, Bauersachs J. MicroRNAs: novel regulators in cardiac development and disease. Cardiovasc Res 2008; 79:562-70. [PMID: 18511432 DOI: 10.1093/cvr/cvn137] [Citation(s) in RCA: 276] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous, small ribonucleotides regulating the translation of target messenger RNAs that have been shown to be involved in orchestrating growth, development, function, and stress responses of various organs, including the heart. Muscle miRNAs are mainly controlled by a network of myogenic transcription factors, and throughout cardiac development they fine-tune regulatory protein levels in a spatiotemporal manner. Recent profiling studies revealed that miRNA expression patterns are derailed in both human cardiac disease and animal models of cardiac hypertrophy and failure. Modulation of miRNA expression in vitro as well as in vivo has revealed an important role of miRNAs in regulating heart function, particularly cardiac growth and conductance. Here, we overview the recent findings on miRNAs in cardiac development and disease and report the latest advances in the identification and validation of miRNA targets, which are important for a comprehensive understanding of cardiac miRNA function. Finally, we focus on the development and use of miRNA antagonists (antagomirs) to target miRNAs in vivo, which may translate into novel therapeutic strategies for heart disease in the future.
Collapse
Affiliation(s)
- Thomas Thum
- Medizinische Klinik und Poliklinik I, Universitätsklinikum, Julius-Maximilians-Universität, Josef-Schneider-Street 2, D-97080 Würzburg, Germany.
| | | | | |
Collapse
|
2015
|
Abstract
Adaptations that are the result of exercise require a multitude of changes at the level of gene expression. The mechanisms involved in regulating these changes are many, and can occur at various points in the pathways that affect gene expression. The completion of the human genome sequence, along with the genomes of related species, has provided an enormous amount of information to help dissect and understand these pathways. High-throughput methods, such as DNA microarrays, were the first on the scene to take advantage of this wealth of information. A new generation of microarrays has now taken the next step in revealing the mechanisms controlling gene expression. Analysis of the regulation of gene expression can now be profiled in a high-throughput fashion. However, the application of this technology has yet to be fully realized in the exercise physiology community. This review will highlight some of the latest advances in microarrays and briefly discuss some potential applications to the field of exercise physiology.
Collapse
Affiliation(s)
- Carl Virtanen
- Microarray Centre, University Health Network, MaRS Centre, Toronto Discovery Tower, 101 College St., Toronto, ON M5G 1L7, Canada
| | | |
Collapse
|
2016
|
Affiliation(s)
- Elly M Tanaka
- Center for Regenerative Therapies, 01307 Dresden, Germany
| | | |
Collapse
|
2017
|
Abstract
MicroRNAs are non-coding small RNAs that regulate gene expression by Watson-Crick base pairing to target messenger RNA (mRNA). They are involved in most biological and pathological processes, including tumorigenesis. The binding of microRNA to mRNA is critical for regulating the mRNA level and protein expression. However, this binding can be affected by single-nucleotide polymorphisms that can reside in the microRNA target site, which can either abolish existing binding sites or create illegitimate binding sites. Therefore, polymorphisms in microRNA can have a differing effect on gene and protein expression and represent another type of genetic variability that can influence the risk of certain human diseases. Different approaches have been used to predict and identify functional polymorphisms within microRNA-binding sites. The biological relevance of these polymorphisms in predicted microRNA-binding sites is beginning to be examined in large case-control studies.
Collapse
|
2018
|
|
2019
|
Koch T, Rosenbohm C, Hansen HF, Hansen B, Marie Straarup E, Kauppinen S. Locked Nucleic Acid: Properties and Therapeutic Aspects. THERAPEUTIC OLIGONUCLEOTIDES 2008. [DOI: 10.1039/9781847558275-00103] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Troels Koch
- Santaris Pharma A/S Bøge Allé 3 DK-2970 Hørsholm Denmark
| | | | | | - Bo Hansen
- Santaris Pharma A/S Bøge Allé 3 DK-2970 Hørsholm Denmark
| | | | | |
Collapse
|
2020
|
Michalak P, Malone JH. Testis-derived microRNA profiles of African clawed frogs (Xenopus) and their sterile hybrids. Genomics 2008; 91:158-64. [PMID: 18079091 DOI: 10.1016/j.ygeno.2007.10.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 10/26/2007] [Accepted: 10/29/2007] [Indexed: 12/12/2022]
Abstract
Gene regulation was long predicted to play a vital role in speciation and species divergence. Only recently with the advent of new technologies, however, has it been possible to address the question of the relative contributions of different mechanisms of gene expression to the evolution of phenotypic diversity. Here we broaden the question and ask whether microRNAs, a large class of small regulatory RNAs, play a role in reproductive isolation between species by contributing to hybrid male sterility. MicroRNAs from the testes of clawed frogs (Xenopus) were extracted and the expression profiles of sterile hybrids were compared with males of a parental species. Hybrid testes were largely microRNA-depleted relative to those of nonhybrids, and this pattern was validated with quantitative RT-PCR. A number of candidate differential microRNAs from this study have previously been described as testis-specific in the mouse, suggesting that microRNA structural conservation may be associated with functional retention.
Collapse
Affiliation(s)
- Pawel Michalak
- Department of Biology, University of Texas Arlington, Box 19498, Arlington, TX 76019, USA.
| | | |
Collapse
|
2021
|
Abstract
The Nobel Prize in Medicine and Physiology was awarded to the RNA interference (RNAi) field in 2006 because of the huge therapeutic potential this technique harbours. However, the RNAi technology is based on a natural mechanism that utilizes short noncoding RNA molecules (microRNAs) to regulate gene expression. This paper reviews our current knowledge about microRNAs focusing on their involvement in cancer and their potential as diagnostics and therapeutics.
Collapse
Affiliation(s)
- T Dalmay
- School of Biological Sciences, University of East Anglia, Norwich, UK.
| |
Collapse
|
2022
|
MicroRNAs flex their muscles. Trends Genet 2008; 24:159-66. [DOI: 10.1016/j.tig.2008.01.007] [Citation(s) in RCA: 254] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 01/08/2008] [Accepted: 01/11/2008] [Indexed: 11/21/2022]
|
2023
|
|
2024
|
Abstract
MicroRNAs (miRNAs) are approximately 22-nucleotide-long noncoding RNAs that normally function by suppressing translation and destabilizing messenger RNAs bearing complementary target sequences. Some miRNAs are expressed in a cell- or tissue-specific manner and may contribute to the establishment and/or maintenance of cellular identity. Recent studies indicate that tissue-specific miRNAs may function at multiple hierarchical levels of gene regulatory networks, from targeting hundreds of effector genes incompatible with the differentiated state to controlling the levels of global regulators of transcription and alternative pre-mRNA splicing. This multilevel regulation may allow individual miRNAs to profoundly affect the gene expression program of differentiated cells.
Collapse
Affiliation(s)
- Eugene V. Makeyev
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Tom Maniatis
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
2025
|
Abstract
The completion of genome sequences and subsequent high-throughput mapping of molecular networks have allowed us to study biology from the network perspective. Experimental, statistical and mathematical modeling approaches have been employed to study the structure, function and dynamics of molecular networks, and begin to reveal important links of various network properties to the functions of the biological systems. In agreement with these functional links, evolutionary selection of a network is apparently based on the function, rather than directly on the structure of the network. Dynamic modularity is one of the prominent features of molecular networks. Taking advantage of such a feature may simplify network-based biological studies through construction of process-specific modular networks and provide functional and mechanistic insights linking genotypic variations to complex traits or diseases, which is likely to be a key approach in the next wave of understanding complex human diseases. With the development of ready-to-use network analysis and modeling tools the networks approaches will be infused into everyday biological research in the near future.
Collapse
Affiliation(s)
- Jing-Dong Jackie Han
- Chinese Academy of Sciences Key Laboratory of Molecular Developmental Biology and Center for Molecular Systems Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Datun Road, Beijing 100101, China.
| |
Collapse
|
2026
|
Sun Q, Zhang Y, Yang G, Chen X, Zhang Y, Cao G, Wang J, Sun Y, Zhang P, Fan M, Shao N, Yang X. Transforming growth factor-beta-regulated miR-24 promotes skeletal muscle differentiation. Nucleic Acids Res 2008; 36:2690-9. [PMID: 18353861 PMCID: PMC2377434 DOI: 10.1093/nar/gkn032] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) have recently been proposed as a versatile class of molecules involved in regulation of a variety of biological processes. However, the role of miRNAs in TGF-beta-regulated biological processes is poorly addressed. In this study, we found that miR-24 was upregulated during myoblast differentiation and could be inhibited by TGF-beta1. Using both a reporter assay and Northern blot analysis, we showed that TGF-beta1 repressed miR-24 transcription which was dependent on the presence of Smad3 and a Smads binding site in the promoter region of miR-24. TGF-beta1 was unable to inhibit miR-24 expression in Smad3-deficient myoblasts, which exhibited accelerated myogenesis. Knockdown of miR-24 led to reduced expression of myogenic differentiation markers in C2C12 cells, while ectopic expression of miR-24 enhanced differentiation, and partially rescued inhibited myogenesis by TGF-beta1. This is the first study demonstrating a critical role for miRNAs in modulating TGF-beta-dependent inhibition of myogenesis, and provides a novel mechanism of the genetic regulation of TGF-beta signaling during skeletal muscle differentiation.
Collapse
Affiliation(s)
- Qiang Sun
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, 20 Dongdajie, Beijing 100071, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2027
|
Catalucci D, Latronico MVG, Condorelli G. MicroRNAs Control Gene Expression. Ann N Y Acad Sci 2008; 1123:20-9. [DOI: 10.1196/annals.1420.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
2028
|
Microarray-based label-free detection of RNA using bispyrene-modified 2'-O-methyl oligoribonucleotide as capture and detection probe. Bioorg Med Chem Lett 2008; 18:2590-3. [PMID: 18394885 DOI: 10.1016/j.bmcl.2008.03.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 02/19/2008] [Accepted: 03/14/2008] [Indexed: 11/22/2022]
Abstract
A novel oligonucleotide microarray that can detect RNAs without fluorescent labeling of sample RNAs was developed. As a capture and detection probe, bispyrene-modified 2'-O-methyl oligoribonucleotide (OMUpy2), whose fluorescence was dramatically increased when hybridized with its complementary RNA, was adopted. Fluorescence of the OMUpy2 tethered on the glass surface was enhanced as much as 22-fold by the addition of complementary oligoribonucleotide.
Collapse
|
2029
|
Yin VP, Thomson JM, Thummel R, Hyde DR, Hammond SM, Poss KD. Fgf-dependent depletion of microRNA-133 promotes appendage regeneration in zebrafish. Genes Dev 2008; 22:728-33. [PMID: 18347091 PMCID: PMC2275425 DOI: 10.1101/gad.1641808] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 01/21/2008] [Indexed: 12/26/2022]
Abstract
Appendage regeneration is defined by rapid changes in gene expression that achieve dramatic developmental effects, suggesting involvement of microRNAs (miRNAs). Here, we find dynamic regulation of many miRNAs during zebrafish fin regeneration. In particular, miR-133 levels are high in uninjured fins but low during regeneration. When regeneration was blocked by Fibroblast growth factor (Fgf) receptor inhibition, high miR-133 levels were quickly restored. Experimentally increasing amounts of miR-133 attenuated fin regeneration. Conversely, miR-133 antagonism during Fgf receptor inhibition accelerated regeneration through increased proliferation within the regeneration blastema. The Mps1 kinase, an established positive regulator of blastemal proliferation, is an in vivo target of miR-133. Our findings identify miRNA depletion as a new regulatory mechanism for complex tissue regeneration.
Collapse
Affiliation(s)
- Viravuth P. Yin
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - J. Michael Thomson
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Ryan Thummel
- Center for Zebrafish Research and Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - David R. Hyde
- Center for Zebrafish Research and Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Scott M. Hammond
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Kenneth D. Poss
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
2030
|
Abstract
microRNAs (miRNAs) are a class of highly conserved small non-coding RNAs that negatively regulate gene expression post-transcriptionally. The emerging field of miRNA biology has begun to unravel roles for these regulatory molecules in a range of biological functions, including cardiac and skeletal muscle development, as well as in muscle-related disease processes. In this paper, we review the role of miRNAs in muscle biology. Recent genetic studies have demonstrated that miRNAs are required for both proper muscle development and function, with crucial roles for miRNAs being identified in regulating muscle cell proliferation and differentiation. Furthermore, dysregulated expression of miRNAs has been correlated to certain muscle-related diseases, including cardiac hypertrophy, cardiac arrhythmias, and muscular dystrophy.
Collapse
Affiliation(s)
- Thomas E Callis
- Carolina Cardiovascular Biology Center, Department of Cell & Developmental Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
2031
|
Histone deacetylase 5 acquires calcium/calmodulin-dependent kinase II responsiveness by oligomerization with histone deacetylase 4. Mol Cell Biol 2008; 28:3437-45. [PMID: 18332106 DOI: 10.1128/mcb.01611-07] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) phosphorylates histone deacetylase 4 (HDAC4), a class IIa HDAC, resulting in the cytosolic accumulation of HDAC4 and the derepression of the transcription factor myocyte enhancer factor 2. Phosphorylation by CaMKII requires docking of the kinase to a specific domain of HDAC4 not present in other HDACs. Paradoxically, however, CaMKII signaling can also promote the nuclear export of other class IIa HDACs, such as HDAC5. Here, we show that HDAC4 and HDAC5 form homo- and hetero-oligomers via a conserved coiled-coil domain near their amino termini. Whereas HDAC5 alone is unresponsive to CaMKII, it becomes responsive to CaMKII in the presence of HDAC4. The acquisition of CaMKII responsiveness by HDAC5 is mediated by HDAC5's direct association with HDAC4 and can occur by phosphorylation of HDAC4 or by transphosphorylation by CaMKII bound to HDAC4. Thus, HDAC4 integrates upstream Ca(2+)-dependent signals via its association with CaMKII and transmits these signals to HDAC5 by protein-protein interactions. We conclude that HDAC4 represents a point of convergence for CaMKII signaling to downstream HDAC-regulated genes, and we suggest that modulation of the interaction of CaMKII and HDAC4 represents a means of regulating CaMKII-dependent gene programs.
Collapse
|
2032
|
Ivey KN, Muth A, Arnold J, King FW, Yeh RF, Fish JE, Hsiao EC, Schwartz RJ, Conklin BR, Bernstein HS, Srivastava D. MicroRNA regulation of cell lineages in mouse and human embryonic stem cells. Cell Stem Cell 2008; 2:219-29. [PMID: 18371447 PMCID: PMC2293325 DOI: 10.1016/j.stem.2008.01.016] [Citation(s) in RCA: 459] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Revised: 01/09/2008] [Accepted: 01/24/2008] [Indexed: 12/28/2022]
Abstract
Cell fate decisions of pluripotent embryonic stem (ES) cells are dictated by activation and repression of lineage-specific genes. Numerous signaling and transcriptional networks progressively narrow and specify the potential of ES cells. Whether specific microRNAs help refine and limit gene expression and, thereby, could be used to manipulate ES cell differentiation has largely been unexplored. Here, we show that two serum response factor (SRF)-dependent muscle-specific microRNAs, miR-1 and miR-133, promote mesoderm formation from ES cells but have opposing functions during further differentiation into cardiac muscle progenitors. Furthermore, miR-1 and miR-133 were potent repressors of nonmuscle gene expression and cell fate during mouse and human ES cell differentiation. miR-1's effects were in part mediated by translational repression of the Notch ligand Delta-like 1 (Dll-1). Our findings indicate that muscle-specific miRNAs reinforce the silencing of nonmuscle genes during cell lineage commitment and suggest that miRNAs may have general utility in regulating cell-fate decisions from pluripotent ES cells.
Collapse
Affiliation(s)
- Kathryn N. Ivey
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Alecia Muth
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Joshua Arnold
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Frank W. King
- Department of Pediatrics, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Ru-Fang Yeh
- Department of Epidemiology and Biostatistics, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Jason E. Fish
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Edward C. Hsiao
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
- Department of Medicine, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Robert J. Schwartz
- Institute of Biosciences and Technology, Texas A & M Health Science Center, Baylor College of Medicine, Houston, TX 77030
| | - Bruce R. Conklin
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
- Department of Medicine, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
- Department of Molecular and Cellular Pharmacology, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Harold S. Bernstein
- Department of Pediatrics, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
- Department of Pediatrics, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
- Department of Biochemistry and Biophysics, University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158
| |
Collapse
|
2033
|
Abstract
The modulation of gene expression by small non-coding RNAs is a recently discovered level of gene regulation in animals and plants. In particular, microRNAs (miRNAs) and Piwi-interacting RNAs (piRNAs) have been implicated in various aspects of animal development, such as neuronal, muscle and germline development. During the past year, an improved understanding of the biological functions of small non-coding RNAs has been fostered by the analysis of genetic deletions of individual miRNAs in mammals. These studies show that miRNAs are key regulators of animal development and are potential human disease loci.
Collapse
|
2034
|
The effect of central loops in miRNA:MRE duplexes on the efficiency of miRNA-mediated gene regulation. PLoS One 2008; 3:e1719. [PMID: 18320040 PMCID: PMC2248708 DOI: 10.1371/journal.pone.0001719] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2007] [Accepted: 01/28/2008] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) guide posttranscriptional repression of mRNAs. Hundreds of miRNAs have been identified but the target identification of mammalian mRNAs is still a difficult task due to a poor understanding of the interaction between miRNAs and the miRNA recognizing element (MRE). In recent research, the importance of the 5' end of the miRNA:MRE duplex has been emphasized and the effect of the tail region addressed, but the role of the central loop has largely remained unexplored. Here we examined the effect of the loop region in miRNA:MRE duplexes and found that the location of the central loop is one of the important factors affecting the efficiency of gene regulation mediated by miRNAs. It was further determined that the addition of a loop score combining both location and size as a new criterion for predicting MREs and their cognate miRNAs significantly decreased the false positive rates and increased the specificity of MRE prediction.
Collapse
|
2035
|
Abstract
Wnt regulation of muscle development is thought to be mediated by the beta-catenin-TCF/LEF-dependent canonical pathway. Here we demonstrate that beta-catenin, not TCF/LEF, is required for muscle differentiation. We showed that beta-catenin interacts directly with MyoD, a basic helix-loop-helix transcription factor essential for muscle differentiation and enhances its binding to E box elements and transcriptional activity. MyoD-mediated transactivation is inhibited in muscle cells when beta-catenin is deficient or the interaction between MyoD and beta-catenin is disrupted. These results demonstrate that beta-catenin is necessary for MyoD function, identifying MyoD as an effector in the Wnt canonical pathway.
Collapse
|
2036
|
Yang XJ, Seto E. The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men. Nat Rev Mol Cell Biol 2008; 9:206-18. [PMID: 18292778 PMCID: PMC2667380 DOI: 10.1038/nrm2346] [Citation(s) in RCA: 947] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein lysine deacetylases have a pivotal role in numerous biological processes and can be divided into the Rpd3/Hda1 and sirtuin families, each having members in diverse organisms including prokaryotes. In vertebrates, the Rpd3/Hda1 family contains 11 members, traditionally referred to as histone deacetylases (HDAC) 1-11, which are further grouped into classes I, II and IV. Whereas most class I HDACs are subunits of multiprotein nuclear complexes that are crucial for transcriptional repression and epigenetic landscaping, class II members regulate cytoplasmic processes or function as signal transducers that shuttle between the cytoplasm and the nucleus. Little is known about class IV HDAC11, although its evolutionary conservation implies a fundamental role in various organisms.
Collapse
Affiliation(s)
- Xiang-Jiao Yang
- Molecular Oncology Group, Department of Medicine, McGill University Health Center, Montréal, Québec, H3A 1A1, Canada.
| | | |
Collapse
|
2037
|
Barry SP, Davidson SM, Townsend PA. Molecular regulation of cardiac hypertrophy. Int J Biochem Cell Biol 2008; 40:2023-39. [PMID: 18407781 DOI: 10.1016/j.biocel.2008.02.020] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 02/13/2008] [Accepted: 02/15/2008] [Indexed: 01/05/2023]
Abstract
Heart failure is one of the leading causes of mortality in the western world and encompasses a wide spectrum of cardiac pathologies. When the heart experiences extended periods of elevated workload, it undergoes hypertrophic enlargement in response to the increased demand. Cardiovascular disease, such as that caused by myocardial infarction, obesity or drug abuse promotes cardiac myocyte hypertrophy and subsequent heart failure. A number of signalling modulators in the vasculature milieu are known to regulate heart mass including those that influence gene expression, apoptosis, cytokine release and growth factor signalling. Recent evidence using genetic and cellular models of cardiac hypertrophy suggests that pathological hypertrophy can be prevented or reversed and has promoted an enormous drive in drug discovery research aiming to identify novel and specific regulators of hypertrophy. In this review we describe the molecular characteristics of cardiac hypertrophy such as the aberrant re-expression of the fetal gene program. We discuss the various molecular pathways responsible for the co-ordinated control of the hypertrophic program including: natriuretic peptides, the adrenergic system, adhesion and cytoskeletal proteins, IL-6 cytokine family, MEK-ERK1/2 signalling, histone acetylation, calcium-mediated modulation and the exciting recent discovery of the role of microRNAs in controlling cardiac hypertrophy. Characterisation of the signalling pathways leading to cardiac hypertrophy has led to a wealth of knowledge about this condition both physiological and pathological. The challenge will be translating this knowledge into potential pharmacological therapies for the treatment of cardiac pathologies.
Collapse
Affiliation(s)
- Sean P Barry
- Medical Molecular Biology Unit, Institute of Child Health, University College London, 30 Guilford Street, London WC1N IEH, United Kingdom.
| | | | | |
Collapse
|
2038
|
Abstract
Reprogramming of somatic cells to a pluripotent embryonic stem cell-like state has been achieved by nuclear transplantation of a somatic nucleus into an enucleated egg and most recently by introducing defined transcription factors into somatic cells. Nuclear reprogramming is of great medical interest, as it has the potential to generate a source of patient-specific cells. Here, we review strategies to reprogram somatic cells to a pluripotent embryonic state and discuss our understanding of the molecular mechanisms of reprogramming based on recent insights into the regulatory circuitry of the pluripotent state.
Collapse
Affiliation(s)
- Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA.
| | | |
Collapse
|
2039
|
Wong CF, Tellam RL. MicroRNA-26a targets the histone methyltransferase Enhancer of Zeste homolog 2 during myogenesis. J Biol Chem 2008; 283:9836-43. [PMID: 18281287 DOI: 10.1074/jbc.m709614200] [Citation(s) in RCA: 249] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MicroRNA (miRNA) are important regulators of many biological processes, but the targets for most miRNA are still poorly defined. In this study, we profiled the expression of miRNA during myogenesis, from proliferating myoblasts through to terminally differentiated myotubes. Microarray results identified six significantly differentially expressed miRNA that were more than 2-fold different in myotubes. From this list, miRNA-26a (miR-26a), an up-regulated miRNA, was further examined. Overexpression of miR-26a in murine myogenic C2C12 cells induced creatine kinase activity, an enzyme that markedly increases during myogenesis. Further, myoD and myogenin mRNA expression levels were also up-regulated. These results suggest that increased expression of miR-26a promotes myogenesis. Through a bioinformatics approach, we identified the histone methyltransferase, Enhancer of Zeste homolog 2 (Ezh2), as a potential target of miR-26a. Overexpression of miR-26a suppressed the activity of a luciferase reporter construct fused with the 3'-untranslated region of Ezh2. In addition, miR-26a overexpression decreased Ezh2 mRNA expression. These results reveal a model of regulation during myogenesis whereby the up-regulation of miR-26a acts to post-transcriptionally repress Ezh2, a known suppressor of skeletal muscle cell differentiation.
Collapse
Affiliation(s)
- Chung Fai Wong
- CSIRO Livestock Industries, Queensland Bioscience Precinct, 306 Carmody Road, St. Lucia 4067, QLD, Australia
| | | |
Collapse
|
2040
|
Stenvang J, Kauppinen S. MicroRNAs as targets for antisense-based therapeutics. Expert Opin Biol Ther 2008; 8:59-81. [PMID: 18081537 DOI: 10.1517/14712598.8.1.59] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are a novel class of endogenous non-coding single-stranded RNAs, which regulate gene expression post-transcriptionally by base pairing with their target mRNAs. So far > 5000 miRNA entries have been registered and miRNAs have been implicated in most, if not all, central cellular processes and several diseases. As the mechanism of action for miRNA regulation of target mRNAs is mediated by Watson-Crick base pairing, antisense oligonucleotides targeting the miRNAs appear as an obvious choice to specifically inhibit miRNA function. Indeed, miRNAs can be antagonized in vivo by oligonucleotides composed of high-affinity nucleotide mimics. Lessons learned from traditional antisense strategies and small-interfering RNA approaches, that is from potent nucleotide mimics, design rules, pharmacokinetics, administration and safety issues, are likely to pave the way for future clinical trials of miRNA-antagonizing oligonucleotides.
Collapse
Affiliation(s)
- Jan Stenvang
- University of Copenhagen, Wilhelm Johannsen Centre for Functional Genome Research, Department of Cellular and Molecular Medicine, Blegdamsvej 3, DK-2200 Copenhagen, Denmark.
| | | |
Collapse
|
2041
|
Kobayashi T, Lu J, Cobb BS, Rodda SJ, McMahon AP, Schipani E, Merkenschlager M, Kronenberg HM. Dicer-dependent pathways regulate chondrocyte proliferation and differentiation. Proc Natl Acad Sci U S A 2008; 105:1949-54. [PMID: 18238902 PMCID: PMC2538863 DOI: 10.1073/pnas.0707900105] [Citation(s) in RCA: 284] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Indexed: 12/21/2022] Open
Abstract
Small noncoding RNAs, microRNAs (miRNAs), bind to messenger RNAs through base pairing to suppress gene expression. Despite accumulating evidence that miRNAs play critical roles in various biological processes across diverse organisms, their roles in mammalian skeletal development have not been demonstrated. Here, we show that Dicer, an essential component for biogenesis of miRNAs, is essential for normal skeletal development. Dicer-null growth plates show a progressive reduction in the proliferating pool of chondrocytes, leading to severe skeletal growth defects and premature death of mice. The reduction of proliferating chondrocytes in Dicer-null growth plates is caused by two distinct mechanisms: decreased chondrocyte proliferation and accelerated differentiation into postmitotic hypertrophic chondrocytes. These defects appear to be caused by mechanisms downstream or independent of the Ihh-PTHrP signaling pathway, a pivotal signaling system that regulates chondrocyte proliferation and differentiation. Microarray analysis of Dicer-null chondrocytes showed limited expression changes in miRNA-target genes, suggesting that, in the majority of cases, chondrocytic miRNAs do not directly regulate target RNA abundance. Our results demonstrate the critical role of the Dicer-dependent pathway in the regulation of chondrocyte proliferation and differentiation during skeletal development.
Collapse
Affiliation(s)
- Tatsuya Kobayashi
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | | | | | | | | | | | | | | |
Collapse
|
2042
|
Targeted deletion of Dicer in the heart leads to dilated cardiomyopathy and heart failure. Proc Natl Acad Sci U S A 2008; 105:2111-6. [PMID: 18256189 DOI: 10.1073/pnas.0710228105] [Citation(s) in RCA: 446] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease is the leading cause of human morbidity and mortality. Dilated cardiomyopathy (DCM) is the most common form of cardiomyopathy associated with heart failure. Here, we report that cardiac-specific knockout of Dicer, a gene encoding a RNase III endonuclease essential for microRNA (miRNA) processing, leads to rapidly progressive DCM, heart failure, and postnatal lethality. Dicer mutant mice show misexpression of cardiac contractile proteins and profound sarcomere disarray. Functional analyses indicate significantly reduced heart rates and decreased fractional shortening of Dicer mutant hearts. Consistent with the role of Dicer in animal hearts, Dicer expression was decreased in end-stage human DCM and failing hearts and, most importantly, a significant increase of Dicer expression was observed in those hearts after left ventricle assist devices were inserted to improve cardiac function. Together, our studies demonstrate essential roles for Dicer in cardiac contraction and indicate that miRNAs play critical roles in normal cardiac function and under pathological conditions.
Collapse
|
2043
|
Abstract
PURPOSE OF REVIEW In skeletal muscle, environmental demands activate signal transduction pathways that ultimately promote adaptive changes in myofiber cytoarchitecture and protein composition. Recent advances in determining the factors involved in these signal transduction pathways provide insight into possible therapeutic methods to remodel skeletal muscle. RECENT FINDINGS Advances in genetic engineering have allowed the introduction or depletion of factors within the myofiber, facilitating the evaluation of signaling factors during muscle remodeling. Using transgenic mouse models, activation of specific signaling pathways promoted type I oxidative myofibers, increased the fatigue resistance of muscle, increased skeletal muscle mass and ameliorated muscle injury in myopathic mouse models. Moreover, new technologies are being used to generate global gene and protein expression profiles to identify new factors involved in skeletal muscle remodeling. Finally, small RNAs, microRNAs, are emerging as powerful regulators of gene expression in most tissues, including skeletal muscle. Recent findings predict that targeted delivery of miRNAs will specifically manipulate genes and if used therapeutically will revolutionize clinical medicine. SUMMARY Developing drugs to target signaling pathways associated with remodeling myofibers provides a possible therapeutic approach to combat skeletal muscle disease. In addition, genome-wide technologies can identify new biomarkers capable of diagnosing myopathies and determine a patient's response to therapy. Furthermore, therapeutic strategies are being designed to target microRNAs in anticipation of blocking gene repression correlated with muscle pathology.
Collapse
|
2044
|
Tili E, Michaille JJ, Gandhi V, Plunkett W, Sampath D, Calin GA. miRNAs and their potential for use against cancer and other diseases. Future Oncol 2008; 3:521-37. [PMID: 17927518 DOI: 10.2217/14796694.3.5.521] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
miRNAs are 19-24 nucleotide long noncoding RNAs found in almost all genetically dissected species, including viruses, plants, nematodes, flies, fish, mice and humans. Rapid advances have been made in understanding their physiological functions, while abnormal patterns of miRNA expression have been found in many disease states, most notably human cancer. It is now clear that miRNAs represent a class of genes with a great potential for use in diagnosis, prognosis and therapy. In this review we will focus on the discoveries that elucidate their crucial role in mammalian diseases, particularly in cancer, and propose that miRNA-based gene therapy might become the potential technology of choice in a wide range of human diseases including cancer.
Collapse
Affiliation(s)
- Esmerina Tili
- Ohio State University, Department of Molecular Virology, Immunology & Medical Genetics and Comprehensive Cancer Center, Columbus, OH 43210, USA.
| | | | | | | | | | | |
Collapse
|
2045
|
Mizuno Y, Yagi K, Tokuzawa Y, Kanesaki-Yatsuka Y, Suda T, Katagiri T, Fukuda T, Maruyama M, Okuda A, Amemiya T, Kondoh Y, Tashiro H, Okazaki Y. miR-125b inhibits osteoblastic differentiation by down-regulation of cell proliferation. Biochem Biophys Res Commun 2008; 368:267-72. [PMID: 18230348 DOI: 10.1016/j.bbrc.2008.01.073] [Citation(s) in RCA: 238] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2007] [Accepted: 01/13/2008] [Indexed: 12/22/2022]
Abstract
Although various microRNAs regulate cell differentiation and proliferation, no miRNA has been reported so far to play an important role in the regulation of osteoblast differentiation. Here we describe the role of miR-125b in osteoblastic differentiation in mouse mesenchymal stem cells, ST2, by regulating cell proliferation. The expression of miR-125b was time-dependently increased in ST2 cells, and the increase in miR-125b expression was attenuated in osteoblastic-differentiated ST2 cells induced by BMP-4. The transfection of exogenous miR-125b inhibited proliferation of ST2 cells and caused inhibition of osteoblastic differentiation. In contrast, when the endogenous miR-125b was blocked by transfection of its antisense RNA molecule, alkaline phosphatase activity after BMP-4 treatment was elevated. These results strongly suggest that miR-125b is involved in osteoblastic differentiation through the regulation of cell proliferation.
Collapse
Affiliation(s)
- Yosuke Mizuno
- Division of Functional Genomics & Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka City, Saitama 350-1241, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2046
|
Marcon E, Babak T, Chua G, Hughes T, Moens PB. miRNA and piRNA localization in the male mammalian meiotic nucleus. Chromosome Res 2008; 16:243-60. [DOI: 10.1007/s10577-007-1190-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2007] [Revised: 11/20/2007] [Accepted: 11/20/2007] [Indexed: 10/22/2022]
|
2047
|
MicroRNAs in the miR-106b family regulate p21/CDKN1A and promote cell cycle progression. Mol Cell Biol 2008; 28:2167-74. [PMID: 18212054 DOI: 10.1128/mcb.01977-07] [Citation(s) in RCA: 447] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
microRNAs in the miR-106b family are overexpressed in multiple tumor types and are correlated with the expression of genes that regulate the cell cycle. Consistent with these observations, miR-106b family gain of function promotes cell cycle progression, whereas loss of function reverses this phenotype. Microarray profiling uncovers multiple targets of the family, including the cyclin-dependent kinase inhibitor p21/CDKN1A. We show that p21 is a direct target of miR-106b and that its silencing plays a key role in miR-106b-induced cell cycle phenotypes. We also show that miR-106b overrides a doxorubicin-induced DNA damage checkpoint. Thus, miR-106b family members contribute to tumor cell proliferation in part by regulating cell cycle progression and by modulating checkpoint functions.
Collapse
|
2048
|
The emerging biology of satellite cells and their therapeutic potential. Trends Mol Med 2008; 14:82-91. [PMID: 18218339 DOI: 10.1016/j.molmed.2007.12.004] [Citation(s) in RCA: 250] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Revised: 12/03/2007] [Accepted: 12/03/2007] [Indexed: 12/12/2022]
Abstract
Adult skeletal muscle contains an abundant and highly accessible population of muscle stem and progenitor cells called satellite cells. The primary function of satellite cells is to mediate postnatal muscle growth and repair. Owing to their availability and remarkable capacity to regenerate damaged muscle, satellite cells and their descendent myoblasts have been considered as powerful candidates for cell-based therapies to treat muscular dystrophies and other neuromuscular diseases. However, regenerative medicine in muscle repair requires a thorough understanding of, and the ability to manipulate, the molecular mechanisms that control the proliferation, self-renewal and myogenic differentiation of satellite cells. Here, we review the latest advances in our current understanding of the quiescence, activation, proliferation and self-renewal of satellite cells and the challenges in the development of satellite cell-based regenerative medicine.
Collapse
|
2049
|
Abstract
The heart is among the most conserved organs of the body and is susceptible to defects more than any other organ. Heart malformations, in fact, occur in roughly 1% of newborns. Moreover, cardiovascular disease arising during adult life is among the main causes of morbidity and mortality in developed countries. It is not surprising, therefore, that much effort is being channeled into understanding the development, physiology, and pathology of the cardiovascular system. MicroRNAs, a newly discovered class of small ribonucleotide-based regulators of gene expression, are being implicated in an increasing number of biological processes, and the study of their role in cardiovascular biology is just beginning. Here, we briefly overview microRNAs in general and report on the recent findings regarding their importance for the heart and vasculature, in particular. The new insights that are being gained will permit not only a greater understanding of cardiovascular pathologies but also, hopefully, the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Michael V G Latronico
- Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, Scientific and Technology Pole, Milan, Italy
| | | | | |
Collapse
|
2050
|
Dykxhoorn DM, Chowdhury D, Lieberman J. RNA interference and cancer: endogenous pathways and therapeutic approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 615:299-329. [PMID: 18437900 DOI: 10.1007/978-1-4020-6554-5_14] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The endogenous RNA interference (RNAi) pathway regulates cellular differentiation and development using small noncoding hairpin RNAs, called microRNAs. This chapter will review the link between mammalian microRNAs and genes involved in cellular proliferation, differentiation, and apoptosis. Some microRNAs act as oncogenes or tumor suppressor genes, but the target gene networks they regulate are just beginning to be described. Cancer cells have altered atterns of microRNA expression, which can be used to identify the cell of origin and to subtype cancers. RNAi has also been used to identify novel genes involved in cellular transformation using forward genetic screening methods previously only possible in invertebrates. Possible strategies and obstacles to harnessing RNAi for cancer therapy will also be discussed.
Collapse
Affiliation(s)
- Derek M Dykxhoorn
- Institute for Biomedical Research and Department of Pediatrics, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | | | |
Collapse
|