51
|
Ma J, Wang H, Liu R, Jin L, Tang Q, Wang X, Jiang A, Hu Y, Li Z, Zhu L, Li R, Li M, Li X. The miRNA Transcriptome Directly Reflects the Physiological and Biochemical Differences between Red, White, and Intermediate Muscle Fiber Types. Int J Mol Sci 2015; 16:9635-53. [PMID: 25938964 PMCID: PMC4463610 DOI: 10.3390/ijms16059635] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/24/2015] [Accepted: 04/13/2015] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that can regulate their target genes at the post-transcriptional level. Skeletal muscle comprises different fiber types that can be broadly classified as red, intermediate, and white. Recently, a set of miRNAs was found expressed in a fiber type-specific manner in red and white fiber types. However, an in-depth analysis of the miRNA transcriptome differences between all three fiber types has not been undertaken. Herein, we collected 15 porcine skeletal muscles from different anatomical locations, which were then clearly divided into red, white, and intermediate fiber type based on the ratios of myosin heavy chain isoforms. We further illustrated that three muscles, which typically represented each muscle fiber type (i.e., red: peroneal longus (PL), intermediate: psoas major muscle (PMM), white: longissimus dorsi muscle (LDM)), have distinct metabolic patterns of mitochondrial and glycolytic enzyme levels. Furthermore, we constructed small RNA libraries for PL, PMM, and LDM using a deep sequencing approach. Results showed that the differentially expressed miRNAs were mainly enriched in PL and played a vital role in myogenesis and energy metabolism. Overall, this comprehensive analysis will contribute to a better understanding of the miRNA regulatory mechanism that achieves the phenotypic diversity of skeletal muscles.
Collapse
Affiliation(s)
- Jideng Ma
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Hongmei Wang
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Rui Liu
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Long Jin
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Qianzi Tang
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Xun Wang
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Anan Jiang
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Yaodong Hu
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Zongwen Li
- Novogene Bioinformatics Institute, Beijing 100083, China.
| | - Li Zhu
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Ruiqiang Li
- Novogene Bioinformatics Institute, Beijing 100083, China.
| | - Mingzhou Li
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| | - Xuewei Li
- Institute of Animal Genetics & Breeding, College of Animal Science & Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, Sichuan, China.
| |
Collapse
|
52
|
Traeger LL, Volkening JD, Moffett H, Gallant JR, Chen PH, Novina CD, Phillips GN, Anand R, Wells GB, Pinch M, Güth R, Unguez GA, Albert JS, Zakon H, Sussman MR, Samanta MP. Unique patterns of transcript and miRNA expression in the South American strong voltage electric eel (Electrophorus electricus). BMC Genomics 2015; 16:243. [PMID: 25887781 PMCID: PMC4393597 DOI: 10.1186/s12864-015-1288-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/26/2015] [Indexed: 11/10/2022] Open
Abstract
Background With its unique ability to produce high-voltage electric discharges in excess of 600 volts, the South American strong voltage electric eel (Electrophorus electricus) has played an important role in the history of science. Remarkably little is understood about the molecular nature of its electric organs. Results We present an in-depth analysis of the genome of E. electricus, including the transcriptomes of eight mature tissues: brain, spinal cord, kidney, heart, skeletal muscle, Sachs’ electric organ, main electric organ, and Hunter’s electric organ. A gene set enrichment analysis based on gene ontology reveals enriched functions in all three electric organs related to transmembrane transport, androgen binding, and signaling. This study also represents the first analysis of miRNA in electric fish. It identified a number of miRNAs displaying electric organ-specific expression patterns, including one novel miRNA highly over-expressed in all three electric organs of E. electricus. All three electric organ tissues also express three conserved miRNAs that have been reported to inhibit muscle development in mammals, suggesting that miRNA-dependent regulation of gene expression might play an important role in specifying an electric organ identity from its muscle precursor. These miRNA data were supported using another complete miRNA profile from muscle and electric organ tissues of a second gymnotiform species. Conclusions Our work on the E. electricus genome and eight tissue-specific gene expression profiles will greatly facilitate future research on determining the coding and regulatory sequences that specify the function, development, and evolution of electric organs. Moreover, these data and future studies will be informed by the first comprehensive analysis of miRNA expression in an electric fish presented here. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1288-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lindsay L Traeger
- Department of Genetics, University of Wisconsin, Madison, WI, 53706, USA. .,Biotechnology Center, University of Wisconsin, Madison, WI, 53706, USA.
| | - Jeremy D Volkening
- Biotechnology Center, University of Wisconsin, Madison, WI, 53706, USA. .,Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA.
| | - Howell Moffett
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, 02115, USA. .,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Jason R Gallant
- Department of Zoology, Michigan State University, East Lansing, MI, 48824, USA. .,BEACON Center for the Study of Evolution in Action, Lansing, USA.
| | - Po-Hao Chen
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, 02115, USA. .,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, 02141, USA.
| | - Carl D Novina
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA, 02115, USA. .,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, 02141, USA.
| | - George N Phillips
- BioSciences at Rice and Department of Chemistry, Rice University, Houston, TX, 77005, USA.
| | - Rene Anand
- Department of Pharmacology and Department of Neuroscience, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| | - Gregg B Wells
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, 77483, USA.
| | - Matthew Pinch
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA.
| | - Robert Güth
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA.
| | - Graciela A Unguez
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA.
| | - James S Albert
- Department of Biology, University of Louisiana, Lafayette, LA, 70503, USA.
| | - Harold Zakon
- BEACON Center for the Study of Evolution in Action, Lansing, USA. .,University of Texas, Austin, TX, 78712, USA. .,The Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, The Marine Biological Laboratory, Woods Hole, MA, 02543, USA.
| | - Michael R Sussman
- Biotechnology Center, University of Wisconsin, Madison, WI, 53706, USA. .,Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA.
| | | |
Collapse
|
53
|
Mione M, Bosserhoff A. MicroRNAs in melanocyte and melanoma biology. Pigment Cell Melanoma Res 2015; 28:340-54. [PMID: 25515738 DOI: 10.1111/pcmr.12346] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/15/2014] [Indexed: 12/18/2022]
Abstract
The importance of microRNAs as key molecular components of cellular processes is now being recognized. Recent reports have shown that microRNAs regulate processes as diverse as protein expression and nuclear functions inside cells and are able to signal extracellularly, delivered via exosomes, to influence cell fate at a distance. The versatility of microRNAs as molecular tools inspires the design of novel strategies to control gene expression, protein stability, DNA repair and chromatin accessibility that may prove very useful for therapeutic approaches due to the extensive manageability of these small molecules. However, we still lack a comprehensive understanding of the microRNA network and its interactions with the other layers of regulatory elements in cellular and extracellular functions. This knowledge may be necessary before we exploit microRNA versatility in therapeutic settings. To identify rules of interactions between microRNAs and other regulatory systems, we begin by reviewing microRNA activities in a single cell type: the melanocyte, from development to disease.
Collapse
Affiliation(s)
- Marina Mione
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggestein-Leopoldshafen, Germany
| | | |
Collapse
|
54
|
Santulli G. microRNAs Distinctively Regulate Vascular Smooth Muscle and Endothelial Cells: Functional Implications in Angiogenesis, Atherosclerosis, and In-Stent Restenosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 887:53-77. [PMID: 26662986 PMCID: PMC4871245 DOI: 10.1007/978-3-319-22380-3_4] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endothelial cells (EC) and vascular smooth muscle cells (VSMC) are the main cell types within the vasculature. We describe here how microRNAs (miRs)--noncoding RNAs that can regulate gene expression via translational repression and/or post-transcriptional degradation--distinctively modulate EC and VSMC function in physiology and disease. In particular, the specific roles of miR-126 and miR-143/145, master regulators of EC and VSMC function, respectively, are deeply explored. We also describe the mechanistic role of miRs in the regulation of the pathophysiology of key cardiovascular processes including angiogenesis, atherosclerosis, and in-stent restenosis post-angioplasty. Drawbacks of currently available therapeutic options are discussed, pointing at the challenges and potential clinical opportunities provided by miR-based treatments.
Collapse
MESH Headings
- Angioplasty
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Endothelial Cells/cytology
- Endothelial Cells/metabolism
- Gene Expression Regulation
- Graft Occlusion, Vascular/genetics
- Graft Occlusion, Vascular/metabolism
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Physiologic
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Stents/adverse effects
- Vascular Remodeling
Collapse
|
55
|
Johar D, Siragam V, Mahood TH, Keijzer R. New insights into lung development and diseases: the role of microRNAs. Biochem Cell Biol 2014; 93:139-48. [PMID: 25563747 DOI: 10.1139/bcb-2014-0103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) are short endogenous noncoding RNA molecules (∼ 22 nucleotides) that can regulate gene expression at the post-transcription level. Research interest in the role of miRNAs in lung biology is emerging. MiRNAs have been implicated in a range of processes such as development, homeostasis, and inflammatory diseases in lung tissues and are capable of inducing differentiation, morphogenesis, and apoptosis. In recent years, several studies have reported that miRNAs are differentially regulated in lung development and lung diseases in response to epigenetic changes, providing new insights for their versatile role in various physiological and pathological processes in the lung. In this review, we discuss the contribution of miRNAs to lung development and diseases and possible future implications in the field of lung biology.
Collapse
Affiliation(s)
- Dina Johar
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology (adjunct), University of Manitoba and Biology of Breathing Theme, Manitoba Institute of Child Health, Winnipeg, Manitoba R3E 3P4, Canada
| | | | | | | |
Collapse
|
56
|
WANG XINGSHAN, QIAN WENWEI, WU ZHIHONG, BIAN YANYAN, WENG XISHENG. Preliminary screening of differentially expressed circulating microRNAs in patients with steroid-induced osteonecrosis of the femoral head. Mol Med Rep 2014; 10:3118-24. [DOI: 10.3892/mmr.2014.2660] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 08/07/2014] [Indexed: 11/06/2022] Open
|
57
|
miR-10a regulates proliferation of human cardiomyocyte progenitor cells by targeting GATA6. PLoS One 2014; 9:e103097. [PMID: 25068583 PMCID: PMC4113387 DOI: 10.1371/journal.pone.0103097] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 06/27/2014] [Indexed: 11/19/2022] Open
Abstract
microRNAs (miRNAs) play essential roles in cardiogenesis. The altered expression of miRNAs can result in cardiac malformations by inducing abnormalities in the behavior of cardiac cells. However, the role of miR-10a in the regulation of cardiomyocyte progenitor cells (CMPCs) remains undetermined. In the present study, we found that up- or down-regulation of miR-10a inhibited or promoted the proliferation of human CMPCs, respectively, without affecting their differentiation toward cardiomyocytes. miR-10a bound to GATA6 directly and reduced GATA6 expression. Over-expression of GATA6 greatly attenuated the miR-10a-mediated inhibitory effect on the proliferation of human CMPCs. Thus, our results indicate that miR-10a could effectively modulate the proliferation of human CMPCs by targeting GATA6. The finding provides novel insights into the potency of miR-10a during heart development.
Collapse
|
58
|
Smirnova L, Block K, Sittka A, Oelgeschläger M, Seiler AEM, Luch A. MicroRNA profiling as tool for in vitro developmental neurotoxicity testing: the case of sodium valproate. PLoS One 2014; 9:e98892. [PMID: 24896083 PMCID: PMC4045889 DOI: 10.1371/journal.pone.0098892] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/08/2014] [Indexed: 01/10/2023] Open
Abstract
Studying chemical disturbances during neural differentiation of murine embryonic stem cells (mESCs) has been established as an alternative in vitro testing approach for the identification of developmental neurotoxicants. miRNAs represent a class of small non-coding RNA molecules involved in the regulation of neural development and ESC differentiation and specification. Thus, neural differentiation of mESCs in vitro allows investigating the role of miRNAs in chemical-mediated developmental toxicity. We analyzed changes in miRNome and transcriptome during neural differentiation of mESCs exposed to the developmental neurotoxicant sodium valproate (VPA). A total of 110 miRNAs and 377 mRNAs were identified differently expressed in neurally differentiating mESCs upon VPA treatment. Based on miRNA profiling we observed that VPA shifts the lineage specification from neural to myogenic differentiation (upregulation of muscle-abundant miRNAs, mir-206, mir-133a and mir-10a, and downregulation of neural-specific mir-124a, mir-128 and mir-137). These findings were confirmed on the mRNA level and via immunochemistry. Particularly, the expression of myogenic regulatory factors (MRFs) as well as muscle-specific genes (Actc1, calponin, myosin light chain, asporin, decorin) were found elevated, while genes involved in neurogenesis (e.g. Otx1, 2, and Zic3, 4, 5) were repressed. These results were specific for valproate treatment and―based on the following two observations―most likely due to the inhibition of histone deacetylase (HDAC) activity: (i) we did not observe any induction of muscle-specific miRNAs in neurally differentiating mESCs exposed to the unrelated developmental neurotoxicant sodium arsenite; and (ii) the expression of muscle-abundant mir-206 and mir-10a was similarly increased in cells exposed to the structurally different HDAC inhibitor trichostatin A (TSA). Based on our results we conclude that miRNA expression profiling is a suitable molecular endpoint for developmental neurotoxicity. The observed lineage shift into myogenesis, where miRNAs may play an important role, could be one of the developmental neurotoxic mechanisms of VPA.
Collapse
Affiliation(s)
- Lena Smirnova
- Federal Institute for Risk Assessment (BfR), Berlin, Germany
- * E-mail:
| | - Katharina Block
- Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | | | | | | | - Andreas Luch
- Federal Institute for Risk Assessment (BfR), Berlin, Germany
| |
Collapse
|
59
|
Pérez-Rivas LG, Jerez JM, Carmona R, de Luque V, Vicioso L, Claros MG, Viguera E, Pajares B, Sánchez A, Ribelles N, Alba E, Lozano J. A microRNA signature associated with early recurrence in breast cancer. PLoS One 2014; 9:e91884. [PMID: 24632820 PMCID: PMC3954835 DOI: 10.1371/journal.pone.0091884] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/14/2014] [Indexed: 12/20/2022] Open
Abstract
Recurrent breast cancer occurring after the initial treatment is associated with poor outcome. A bimodal relapse pattern after surgery for primary tumor has been described with peaks of early and late recurrence occurring at about 2 and 5 years, respectively. Although several clinical and pathological features have been used to discriminate between low- and high-risk patients, the identification of molecular biomarkers with prognostic value remains an unmet need in the current management of breast cancer. Using microarray-based technology, we have performed a microRNA expression analysis in 71 primary breast tumors from patients that either remained disease-free at 5 years post-surgery (group A) or developed early (group B) or late (group C) recurrence. Unsupervised hierarchical clustering of microRNA expression data segregated tumors in two groups, mainly corresponding to patients with early recurrence and those with no recurrence. Microarray data analysis and RT-qPCR validation led to the identification of a set of 5 microRNAs (the 5-miRNA signature) differentially expressed between these two groups: miR-149, miR-10a, miR-20b, miR-30a-3p and miR-342-5p. All five microRNAs were down-regulated in tumors from patients with early recurrence. We show here that the 5-miRNA signature defines a high-risk group of patients with shorter relapse-free survival and has predictive value to discriminate non-relapsing versus early-relapsing patients (AUC = 0.993, p-value<0.05). Network analysis based on miRNA-target interactions curated by public databases suggests that down-regulation of the 5-miRNA signature in the subset of early-relapsing tumors would result in an overall increased proliferative and angiogenic capacity. In summary, we have identified a set of recurrence-related microRNAs with potential prognostic value to identify patients who will likely develop metastasis early after primary breast surgery.
Collapse
Affiliation(s)
- Luis G Pérez-Rivas
- Laboratorio de Oncología Molecular, Servicio de Oncología Médica, Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - José M Jerez
- Departamento de Lenguajes y Ciencias de la Computación, Universidad de Málaga, Málaga, Spain
| | - Rosario Carmona
- Plataforma Andaluza de Bioinformática, Universidad de Málaga, Málaga, Spain
| | - Vanessa de Luque
- Laboratorio de Oncología Molecular, Servicio de Oncología Médica, Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Luis Vicioso
- Servicio de Anatomía Patológica, Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - M Gonzalo Claros
- Plataforma Andaluza de Bioinformática, Universidad de Málaga, Málaga, Spain; Departmento de Biología Molecular y Bioquímica, Universidad de Málaga, Málaga, Spain
| | - Enrique Viguera
- Departmento of Biología Celular, Genética y Fisiología Animal, Universidad de Málaga, Málaga, Spain
| | - Bella Pajares
- Laboratorio de Oncología Molecular, Servicio de Oncología Médica, Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Alfonso Sánchez
- Laboratorio de Oncología Molecular, Servicio de Oncología Médica, Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Nuria Ribelles
- Laboratorio de Oncología Molecular, Servicio de Oncología Médica, Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Emilio Alba
- Laboratorio de Oncología Molecular, Servicio de Oncología Médica, Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - José Lozano
- Laboratorio de Oncología Molecular, Servicio de Oncología Médica, Instituto de Biomedicina de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain; Departmento de Biología Molecular y Bioquímica, Universidad de Málaga, Málaga, Spain
| |
Collapse
|
60
|
Hoss AG, Kartha VK, Dong X, Latourelle JC, Dumitriu A, Hadzi TC, MacDonald ME, Gusella JF, Akbarian S, Chen JF, Weng Z, Myers RH. MicroRNAs located in the Hox gene clusters are implicated in huntington's disease pathogenesis. PLoS Genet 2014; 10:e1004188. [PMID: 24586208 PMCID: PMC3937267 DOI: 10.1371/journal.pgen.1004188] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/06/2014] [Indexed: 12/12/2022] Open
Abstract
Transcriptional dysregulation has long been recognized as central to the pathogenesis of Huntington's disease (HD). MicroRNAs (miRNAs) represent a major system of post-transcriptional regulation, by either preventing translational initiation or by targeting transcripts for storage or for degradation. Using next-generation miRNA sequencing in prefrontal cortex (Brodmann Area 9) of twelve HD and nine controls, we identified five miRNAs (miR-10b-5p, miR-196a-5p, miR-196b-5p, miR-615-3p and miR-1247-5p) up-regulated in HD at genome-wide significance (FDR q-value<0.05). Three of these, miR-196a-5p, miR-196b-5p and miR-615-3p, were expressed at near zero levels in control brains. Expression was verified for all five miRNAs using reverse transcription quantitative PCR and all but miR-1247-5p were replicated in an independent sample (8HD/8C). Ectopic miR-10b-5p expression in PC12 HTT-Q73 cells increased survival by MTT assay and cell viability staining suggesting increased expression may be a protective response. All of the miRNAs but miR-1247-5p are located in intergenic regions of Hox clusters. Total mRNA sequencing in the same samples identified fifteen of 55 genes within the Hox cluster gene regions as differentially expressed in HD, and the Hox genes immediately adjacent to the four Hox cluster miRNAs as up-regulated. Pathway analysis of mRNA targets of these miRNAs implicated functions for neuronal differentiation, neurite outgrowth, cell death and survival. In regression models among the HD brains, huntingtin CAG repeat size, onset age and age at death were independently found to be inversely related to miR-10b-5p levels. CAG repeat size and onset age were independently inversely related to miR-196a-5p, onset age was inversely related to miR-196b-5p and age at death was inversely related to miR-615-3p expression. These results suggest these Hox-related miRNAs may be involved in neuroprotective response in HD. Recently, miRNAs have shown promise as biomarkers for human diseases and given their relationship to disease expression, these miRNAs are biomarker candidates in HD.
Collapse
Affiliation(s)
- Andrew G. Hoss
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Graduate Program in Genetics and Genomics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Vinay K. Kartha
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Xianjun Dong
- Program in Bioinformatics and Integrative Biology, and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jeanne C. Latourelle
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Alexandra Dumitriu
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Tiffany C. Hadzi
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Marcy E. MacDonald
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - James F. Gusella
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Schahram Akbarian
- Friedman Brain Institute, Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Richard H. Myers
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Genome Science Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
61
|
Hu R, Pan W, Fedulov AV, Jester W, Jones MR, Weiss ST, Panettieri RA, Tantisira K, Lu Q. MicroRNA-10a controls airway smooth muscle cell proliferation via direct targeting of the PI3 kinase pathway. FASEB J 2014; 28:2347-57. [PMID: 24522205 DOI: 10.1096/fj.13-247247] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Airway smooth muscle (ASM) cells play important physiological roles in the lung, and abnormal proliferation of ASM directly contributes to the airway remodeling during development of lung diseases such as asthma. MicroRNAs are small yet versatile gene tuners that regulate a variety of cellular processes, including cell growth and proliferation; however, little is known about the precise role of microRNAs in the proliferation of the ASM. Here we report that a specific microRNA (miR-10a) controls ASM proliferation through directly inhibiting the phosphoinositide 3-kinase (PI3K) pathway. Next-generation sequencing identified miR-10a as the most abundant microRNA expressed in primary human airway smooth muscle (HASM) cells, accounting for > 20% of all small RNA reads. Overexpression of miR-10a reduced mitogen-induced HASM proliferation by ∼50%, whereas inhibition of miR-10a increased HASM proliferation by ∼40%. Microarray profiling of HASM cells expressing miR-10a mimics identified 52 significantly down-regulated genes as potential targets of miR-10a, including the catalytic subunit α of PI3K (PIK3CA), the central component of the PI3K pathway. MiR-10a directly suppresses PIK3CA expression by targeting the 3'-untranslated region (3'-UTR) of the gene. Inhibition of PIK3CA by miR-10a reduced V-akt murine thymoma viral oncogene homolog 1 (AKT) phosphorylation and blunted the expression of cyclins and cyclin-dependent kinases that are required for HASM proliferation. Together, our study identifies a novel microRNA-mediated regulatory mechanism for PI3K signaling and ASM proliferation and further suggests miR-10a as a potential therapeutic target for lung diseases whose etiology resides in abnormal ASM proliferation.
Collapse
Affiliation(s)
- Ruoxi Hu
- 1Program in Molecular and Integrative Physiological Sciences, Harvard School of Public Health, 665 Huntington Ave., Room I-305, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Gu L, Xu T, Huang W, Xie M, Sun S, Hou S. Identification and profiling of microRNAs in the embryonic breast muscle of pekin duck. PLoS One 2014; 9:e86150. [PMID: 24465928 PMCID: PMC3900480 DOI: 10.1371/journal.pone.0086150] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 12/05/2013] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs) regulate gene expression by fully or partially binding to complementary sequences and play important roles in skeletal muscle development. However, the roles of miRNAs in embryonic breast muscle of duck are unclear. In this study, we analyzed the miRNAs profiling in embryonic breast muscle of Pekin duck at E13 (the 13(th) day of hatching), E19, and E27 by high-throughput sequencing. A total of 382 miRNAs including 359 preciously identified miRNAs 23 novel miRNA candidates were obtained. The nucleotide bias analysis of identified miRNAs showed that the miRNAs in Pekin duck was high conserved. The expression of identified miRNAs were significantly different between E13 and E19 as well as between E27 and E19. Fifteen identified miRNAs validated using stem-loop qRT-PCR can be divided into three groups: those with peak expression at E19, those with minimal expression at E19, and those with continuous increase from E11 to E27. Considering that E19 is the fastest growth stage of embryonic Pekin duck breast muscle, these three groups of miRNAs might be the potential promoters, the potential inhibitors, and the potential sustainer for breast muscle growth. Among the 23 novel miRNAs, novel-miRNA-8 and novel-miRNA-14 had maximal expression at some stages. The stem-loop qRT-PCR analysis of the two novel miRNAs and their two targets (MAP2K1 and PPARα) showed that the expression of novel-mir-8 and PPARα reached the lowest points at E19, while that of novel-mir-14 and MAP2K1 peaked at E19, suggesting novel-miRNA-8 and novel-miRNA-14 may be a potential inhibitor and a potential promoter for embryonic breast muscle development of duck. In summary, these results not only provided an overall insight into the miRNAs landscape in embryonic breast muscle of duck, but also a basis for the further investigation of the miRNAs roles in duck skeletal muscle development.
Collapse
Affiliation(s)
- Lihong Gu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P.R. China
- Institute of Animal Science (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, P.R. China
| | - Tieshan Xu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P.R. China
- Institute of Animal Science (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, P.R. China
| | - Wei Huang
- Institute of Animal Science (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, P.R. China
| | - Ming Xie
- Institute of Animal Science (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, P.R. China
| | - Shiduo Sun
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, P.R. China
| | - Shuisheng Hou
- Institute of Animal Science (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, P.R. China
| |
Collapse
|
63
|
Abstract
MicroRNA MicroRNA s (miRNAs) are small noncoding RNAs acting as endogenous regulators of gene expression. Their discovery is one of the major recent breakthroughs in molecular biology. miRNAs establish a multiplicity of relationships with target mRNAs and exert pleiotropic biological effects in many cell physiological pathways during development and adult life. The dynamic nature of gene expression regulation by Retinoic Acid Retinoic acid (RA) is consistent with an extensive functional interplay with miRNA activities. In fact, RA regulates the expression of many different miRNAs, thus suggesting a relevant function of miRNAs in RA-controlled gene expression programmes. miRNAs have been extensively studied as targets and mediators of the biological activity of RA during embryonic development as well as in normal and neoplastic cells. However, relatively few studies have experimentally explored the direct contribution of miRNA function to the RA signalling pathway. Here, we provide an overview of the mechanistic aspects that allow miRNA biogenesis, functional activation and regulation, focusing on recent evidence that highlights a functional interplay between miRNAs and RA-regulated molecular networks. We report examples of tissue-specific roles of miRNAs modulated by RA in stem cell pluripotency maintenance and regeneration, embryonic development, hematopoietic and neural differentiation, and other biological model systems, underlining their role in disease pathogenesis. We also address novel areas of research linking the RA signalling pathway to the nuclear activity of miRNAs.
Collapse
MESH Headings
- Cell Differentiation
- Embryo, Mammalian
- Embryonic Development
- Gene Expression Regulation
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Pluripotent Stem Cells/cytology
- Pluripotent Stem Cells/metabolism
- Protein Binding
- Protein Multimerization
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoid X Receptors/genetics
- Retinoid X Receptors/metabolism
- Signal Transduction
- Tretinoin/metabolism
Collapse
Affiliation(s)
- Clara Nervi
- Department of Medical-Surgical Sciences and Biotechnologies, University "La Sapienza", Rome, Italy,
| | | |
Collapse
|
64
|
Choi E, Choi E, Hwang KC. MicroRNAs as novel regulators of stem cell fate. World J Stem Cells 2013; 5:172-187. [PMID: 24179605 PMCID: PMC3812521 DOI: 10.4252/wjsc.v5.i4.172] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/13/2013] [Accepted: 08/17/2013] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence in stem cell biology has shown that microRNAs (miRNAs) play a crucial role in cell fate specification, including stem cell self-renewal, lineage-specific differentiation, and somatic cell reprogramming. These functions are tightly regulated by specific gene expression patterns that involve miRNAs and transcription factors. To maintain stem cell pluripotency, specific miRNAs suppress transcription factors that promote differentiation, whereas to initiate differentiation, lineage-specific miRNAs are upregulated via the inhibition of transcription factors that promote self-renewal. Small molecules can be used in a similar manner as natural miRNAs, and a number of natural and synthetic small molecules have been isolated and developed to regulate stem cell fate. Using miRNAs as novel regulators of stem cell fate will provide insight into stem cell biology and aid in understanding the molecular mechanisms and crosstalk between miRNAs and stem cells. Ultimately, advances in the regulation of stem cell fate will contribute to the development of effective medical therapies for tissue repair and regeneration. This review summarizes the current insights into stem cell fate determination by miRNAs with a focus on stem cell self-renewal, differentiation, and reprogramming. Small molecules that control stem cell fate are also highlighted.
Collapse
|
65
|
Kang T, Lu W, Xu W, Anderson L, Bacanamwo M, Thompson W, Chen YE, Liu D. MicroRNA-27 (miR-27) targets prohibitin and impairs adipocyte differentiation and mitochondrial function in human adipose-derived stem cells. J Biol Chem 2013; 288:34394-402. [PMID: 24133204 DOI: 10.1074/jbc.m113.514372] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Prohibitin (PHB) has been reported to play a crucial role in adipocyte differentiation and mitochondrial function. However, the regulative mechanism of PHB during adipogenesis remains unclear. In this study, we determined that the levels of both microRNA (miR)-27a and miR-27b were down-regulated following adipogenic induction of human adipose-derived stem cells, whereas the mRNA level of PHB was up-regulated. Overexpression of miR-27a or miR-27b inhibited PHB expression and adipocyte differentiation. Using PHB 3'-UTR luciferase reporter assay, we observed that miR-27a and miR-27b directly targeted PHB in human adipose-derived stem cells. A compensation of PHB partially restored the adipogenesis inhibited by miR-27. Moreover, we demonstrated the novel finding that ectopic expression of miR-27a or miR-27b impaired mitochondrial biogenesis, structure integrity, and complex I activity accompanied by excessive reactive oxygen species production. Our data suggest that miR-27 is an anti-adipogenic microRNA partly by targeting PHB and impairing mitochondrial function. Pharmacological modulation of miR-27 function may provide a new therapeutic strategy for the treatment of obesity.
Collapse
Affiliation(s)
- Ting Kang
- From the Cardiovascular Research Institute
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Non-coding RNAs: the "dark matter" of cardiovascular pathophysiology. Int J Mol Sci 2013; 14:19987-20018. [PMID: 24113581 PMCID: PMC3821599 DOI: 10.3390/ijms141019987] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 12/17/2022] Open
Abstract
Large-scale analyses of mammalian transcriptomes have identified a significant number of different RNA molecules that are not translated into protein. In fact, the use of new sequencing technologies has identified that most of the genome is transcribed, producing a heterogeneous population of RNAs which do not encode for proteins (ncRNAs). Emerging data suggest that these transcripts influence the development of cardiovascular disease. The best characterized non-coding RNA family is represented by short highly conserved RNA molecules, termed microRNAs (miRNAs), which mediate a process of mRNA silencing through transcript degradation or translational repression. These microRNAs (miRNAs) are expressed in cardiovascular tissues and play key roles in many cardiovascular pathologies, such as coronary artery disease (CAD) and heart failure (HF). Potential links between other ncRNAs, like long non-coding RNA, and cardiovascular disease are intriguing but the functions of these transcripts are largely unknown. Thus, the functional characterization of ncRNAs is essential to improve the overall understanding of cellular processes involved in cardiovascular diseases in order to define new therapeutic strategies. This review outlines the current knowledge of the different ncRNA classes and summarizes their role in cardiovascular development and disease.
Collapse
|
67
|
Benoit C, Ould-Hamouda H, Crepin D, Gertler A, Amar L, Taouis M. Early leptin blockade predisposes fat-fed rats to overweight and modifies hypothalamic microRNAs. J Endocrinol 2013; 218:35-47. [PMID: 23576026 DOI: 10.1530/joe-12-0561] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Perinatal leptin impairment has long-term consequences on energy homeostasis leading to body weight gain. The underlying mechanisms are still not clearly established. We aimed to analyze the long-term effects of early leptin blockade. In this study, newborn rats received daily injection of a pegylated rat leptin antagonist (pRLA) or saline from day 2 (d2) to d13 and then body weight gain, insulin/leptin sensitivity, and expression profile of microRNAs (miRNAs) at the hypothalamic level were determined at d28, d90, or d153 (following 1 month of high-fat diet (HFD) challenge). We show that pRLA treatment predisposes rats to overweight and promotes leptin/insulin resistance in both hypothalamus and liver at adulthood. pRLA treatment also modifies the hypothalamic miRNA expression profile at d28 leading to the upregulation of 34 miRNAs and the downregulation of four miRNAs. For quantitative RT-PCR confirmation, we show the upregulation of rno-miR-10a at d28 and rno-miR-200a, rno-miR-409-5p, and rno-miR-125a-3p following HFD challenge. Finally, pRLA treatment modifies the expression of genes involved in energy homeostasis control such as UCPs and AdipoRs. In pRLA rat muscle, Ucp2/3 and Adipor1/r2 are upregulated at d90. In liver, pRLA treatment upregulates Adipor1/r2 following HFD challenge. These genes are known to be involved in insulin resistance and type 2 diabetes. In conclusion, we demonstrate that the impairment of leptin action in early life promotes insulin/leptin resistance and modifies the hypothalamic miRNA expression pattern in adulthood, and finally, this study highlights the potential link between hypothalamic miRNA expression pattern and insulin/leptin responsiveness.
Collapse
Affiliation(s)
- Charlotte Benoit
- Neuroendocrinologie Moléculaire de la Prise Alimentaire, University of Paris-Sud, UMR 8195, Orsay F-91405, France
| | | | | | | | | | | |
Collapse
|
68
|
Abstract
The rising epidemic of T2DM (Type 2 diabetes mellitus) worldwide is of significant concern. The inherently silent nature of the disease in its early stages precludes early detection; hence cardiovascular disease is often established by the time diabetes is diagnosed. This increased cardiovascular risk leads to significant morbidity and mortality in these individuals. Progressive development of complications as a result of previous exposure to metabolic disturbances appears to leave a long-lasting impression on cells of the vasculature that is not easily reversed and is termed 'metabolic memory'. SMCs (smooth muscle cells) of blood vessel walls, through their inherent ability to switch between a contractile quiescent phenotype and an active secretory state, maintain vascular homoeostasis in health and development. This plasticity also confers SMCs with the essential capacity to adapt and remodel in pathological states. Emerging clinical and experimental studies propose that SMCs in diabetes may be functionally impaired and thus contribute to the increased incidence of macrovascular complications. Although this idea has general support, the underlying molecular mechanisms are currently unknown and hence are the subject of intense research. The aim of the present review is to explore and evaluate the current literature relating to the problem of vascular disease in T2DM and to discuss the critical role of SMCs in vascular remodelling. Possibilities for therapeutic strategies specifically at the level of T2DM SMCs, including recent novel advances in the areas of microRNAs and epigenetics, will be evaluated. Since restoring glucose control in diabetic patients has limited effect in ameliorating their cardiovascular risk, discovering alternative strategies that restrict or reverse disease progression is vital. Current research in this area will be discussed.
Collapse
|
69
|
Ragni E, Montemurro T, Montelatici E, Lavazza C, Viganò M, Rebulla P, Giordano R, Lazzari L. Differential microRNA signature of human mesenchymal stem cells from different sources reveals an “environmental-niche memory” for bone marrow stem cells. Exp Cell Res 2013; 319:1562-74. [DOI: 10.1016/j.yexcr.2013.04.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 03/14/2013] [Accepted: 04/01/2013] [Indexed: 01/20/2023]
|
70
|
Abstract
BACKGROUND MicroRNAs (miRNA, miR) are noncoding RNAs that regulate gene expression by hindering translation. miRNA expression profiles have been shown to differ in vivo and in vitro in many cellular processes associated with cardiovascular diseases (CVDs). The progression of CVDs has also been shown to alter the blood miRNA profile in humans. CONTENT We summarize the results of animal and cell experiments concerning the miRNA profile in the atherosclerotic process and the changes which occur in the blood miRNA profile of individuals with CVD. We also survey the relationship of these CVD-related miRNAs and their expression in the human advanced atherosclerotic plaque, thereby providing more insight into miRNA function in human atherosclerotic lesions. The miRNAs miR-126, -134, -145, -146a, -198, -210, -340*, and -92a were found to be expressed differently in the blood of individuals affected and unaffected by CVD. These differences paralleled those seen in tissue comparisons of miRNA expression in advanced atherosclerotic plaques and healthy arteries. Furthermore, several miRNAs associated with atherosclerosis in in vitro studies (such as miR-10a, -126, -145, -146a/b, -185, -210, and -326) were expressed in plaques in a similar pattern as was predicted by the in vitro experiments. The clinical implications of miRNAs in atherosclerosis as biomarkers and as possible drug targets are also reviewed. SUMMARY miRNA profiles in in vitro and in vivo studies as well as in human peripheral blood are quite representative of the miRNA expression in human atherosclerotic plaques. miRNAs appear promising in terms of future clinical applications.
Collapse
Affiliation(s)
- Emma Raitoharju
- Department of Clinical Chemistry, Pirkanmaa Hospital District, Fimlab Laboratories and University of Tampere, School of Medicine, Finland
| | | | | |
Collapse
|
71
|
Neth P, Nazari-Jahantigh M, Schober A, Weber C. MicroRNAs in flow-dependent vascular remodelling. Cardiovasc Res 2013; 99:294-303. [DOI: 10.1093/cvr/cvt096] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
72
|
Chen KC, Juo SHH. MicroRNAs in atherosclerosis. Kaohsiung J Med Sci 2012; 28:631-40. [DOI: 10.1016/j.kjms.2012.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 02/20/2012] [Indexed: 02/04/2023] Open
|
73
|
Wen Z, Zheng S, Zhou C, Yuan W, Wang J, Wang T. Bone marrow mesenchymal stem cells for post-myocardial infarction cardiac repair: microRNAs as novel regulators. J Cell Mol Med 2012; 16:657-71. [PMID: 22004043 PMCID: PMC3822837 DOI: 10.1111/j.1582-4934.2011.01471.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transplantation of bone marrow-derived mesenchymal stem cells (MSCs) is safe and may improve cardiac function and structural remodelling in patients following myocardial infarction (MI). Cardiovascular cell differentiation and paracrine effects to promote endogenous cardiac regeneration, neovascularization, anti-inflammation, anti-apoptosis, anti-remodelling and cardiac contractility, may contribute to MSC-based cardiac repair following MI. However, current evidence indicates that the efficacy of MSC transplantation was unsatisfactory, due to the poor viability and massive death of the engrafted MSCs in the infarcted myocardium. MicroRNAs are short endogenous, conserved, non-coding RNAs and important regulators involved in numerous facets of cardiac pathophysiologic processes. There is an obvious involvement of microRNAs in almost every facet of putative repair mechanisms of MSC-based therapy in MI, such as stem cell differentiation, neovascularization, apoptosis, cardiac remodelling, cardiac contractility and arrhythmias, and others. It is proposed that therapeutic modulation of individual cardiovascular microRNA of MSCs, either mimicking or antagonizing microRNA actions, will hopefully enhance MSC therapeutic efficacy. In addition, MSCs may be manipulated to enhance functional microRNA expression or to inhibit aberrant microRNA levels in a paracrine manner. We hypothesize that microRNAs may be used as novel regulators in MSC-based therapy in MI and MSC transplantation by microRNA regulation may represent promising therapeutic strategy for MI patients in the future.
Collapse
Affiliation(s)
- Zhuzhi Wen
- The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
74
|
Dip N, Reis ST, Timoszczuk LS, Viana NI, Piantino CB, Morais DR, Moura CM, Abe DK, Silva IA, Srougi M, Dall'Oglio MF, Leite KRM. Stage, grade and behavior of bladder urothelial carcinoma defined by the microRNA expression profile. J Urol 2012; 188:1951-6. [PMID: 22999546 DOI: 10.1016/j.juro.2012.07.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Indexed: 01/04/2023]
Abstract
PURPOSE We identified miRNA expression profiles in urothelial carcinoma that are associated with grade, stage, and recurrence-free and disease specific survival. MATERIALS AND METHODS The expression of 14 miRNAs was evaluated by quantitative reverse transcriptase-polymerase chain reaction in surgical specimens from 30 patients with low grade, noninvasive (pTa) and 30 with high grade, invasive (pT2-3) urothelial carcinoma. Controls were normal bladder tissue from 5 patients who underwent surgical treatment for benign prostatic hyperplasia. Endogenous controls were RNU-43 and RNU-48. miRNA profiles were compared and Kaplan-Meier curves were constructed to analyze disease-free and disease specific survival. RESULTS miR-100 was under expressed in 100% of low grade pTa specimens (p <0.001) and miR-10a was over expressed in 73.3% (p <0.001). miR-21 and miR-205 were over expressed in high grade pT2-3 disease (p = 0.02 and <0.001, respectively). The other miRNAs were present at levels similar to those of normal bladder tissue or under expressed in each tumor group. miR-21 over expression (greater than 1.08) was related to shorter disease-free survival in patients with low grade pTa urothelial carcinoma. Higher miR-10a levels (greater than 2.30) were associated with shorter disease-free and disease specific survival in patients with high grade pT2-3 urothelial carcinoma. CONCLUSIONS Four miRNAs were differentially expressed in the 2 urothelial carcinoma groups. miR-100 and miR-10a showed under expression and over expression, respectively, in low grade pTa tumors. miR-21 and miR-205 were over expressed in pT2-3 disease. In addition, miR-10a and miR-21 over expression was associated with shorter disease-free and disease specific survival. miRNAs could be incorporated into the urothelial carcinoma molecular pathway. These miRNAs could also serve as new diagnostic or prognostic markers and new target drugs.
Collapse
Affiliation(s)
- Nelson Dip
- Laboratory of Medical Investigation, Urology Department, University of São Paulo Medical School, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Wang M, Moisá S, Khan MJ, Wang J, Bu D, Loor JJ. MicroRNA expression patterns in the bovine mammary gland are affected by stage of lactation. J Dairy Sci 2012; 95:6529-35. [PMID: 22959945 DOI: 10.3168/jds.2012-5748] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 07/26/2012] [Indexed: 01/08/2023]
Abstract
The objective of this work was to determine the expression pattern of microRNA (miR) associated with cellular proliferation, lipid metabolism, and innate immunity in dairy cow mammary gland tissue at different stages of lactation. The expression of miR-10a, miR-15b, miR-16, miR-21, miR-31, miR-33b, miR-145, miR-146b, miR-155, miR-181a, miR-205, miR-221, and miR-223 was studied by real-time reverse-transcription PCR in tissue (n=7/stage) harvested via repeated biopsies during the dry period (-30 d prepartum), the fresh period (7 d postpartum), and early lactation (30 d postpartum). Except for miR-31, all miR studied increased in expression between the dry and fresh periods. Among those upregulated, the expression of miR-221 increased further at early lactation, suggesting a role in the control of endothelial cell proliferation or angiogenesis, whereas the expression of miR-223 decreased at early lactation but to a level that was greater than in the dry period, suggesting it could play a role in the mammary response to pathogens soon after parturition. The expression of miR-31, a hormonally regulated miR that inhibits cyclin gene expression, was greater at early lactation compared with the dry period. From a metabolic standpoint, the consistent upregulation of miR-33b during early lactation compared with the dry period suggests that this miR may exert some control over lipogenesis in mammary tissue. Overall, results indicate that expression of miR associated with transcriptional regulation of genes across diverse biological functions is altered by stage of lactation. The specific roles of these miR during lactation will require further research.
Collapse
Affiliation(s)
- M Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | | | | | | | | | | |
Collapse
|
76
|
Joshi SR, McLendon JM, Comer BS, Gerthoffer WT. MicroRNAs-control of essential genes: Implications for pulmonary vascular disease. Pulm Circ 2012; 1:357-64. [PMID: 22140625 PMCID: PMC3224427 DOI: 10.4103/2045-8932.87301] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
During normal lung development and in lung diseases structural cells in the lungs adapt to permit changes in lung function. Fibroblasts, myofibroblasts, smooth muscle, epithelial cells, and various progenitor cells can all undergo phenotypic modulation. In the pulmonary vasculature occlusive vascular lesions that occur in severe pulmonary arterial hypertension are multifocal, polyclonal lesions containing cells presumed to have undergone phenotypic transition resulting in altered proliferation, cell lifespan or contractility. Dynamic changes in gene expression and protein composition that underlie processes responsible for such cellular plasticity are not fully defined. Advances in molecular biology have shown that multiple classes of ribonucleic acid (RNA) collaborate to establish the set of proteins expressed in a cell. Both coding Messenger Ribonucleic acid (mRNA) and small noncoding RNAs (miRNA) act via multiple parallel signaling pathways to regulate transcription, mRNA processing, mRNA stability, translation and possibly protein lifespan. Rapid progress has been made in describing dynamic features of miRNA expression and miRNA function in some vascular tissues. However posttranscriptional gene silencing by microRNA-mediated mRNA degradation and translational blockade is not as well defined in the pulmonary vasculature. Recent progress in defining miRNAs that modulate vascular cell phenotypes is reviewed to illustrate both functional and therapeutic significance of small noncoding RNAs in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Sachindra R Joshi
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | | | | | | |
Collapse
|
77
|
Abstract
PURPOSE OF REVIEW In this review, we summarize the recent advances regarding microRNA (miRNA) functions in the regulation of vascular smooth muscle cell (VSMC) differentiation and phenotypic modulation. RECENT FINDINGS Multiple miRNAs are found to be responsible for VSMC differentiation and proliferation under physiological or pathological condition. A single miRNA downregulates multiple targets, whereas a single gene is regulated by multiple miRNAs to modulate a specific aspect of VSMC phenotype. SUMMARY The phenotype of VSMCs is dynamically regulated in response to environmental stimuli. Deregulation of phenotype switching is associated with vascular diseases. Several miRNAs have been found to be highly expressed in the vasculature, to modulate VSMC phenotype, and to be dysregulated in vascular diseases. By regulating mRNA and/or protein levels posttranscriptionally, miRNAs provide a delicate regulation in the complex molecular networks that regulate the vascular system. Understanding the functions of miRNAs in the regulation of VSMC differentiation and phenotype switching provides new insights into the mechanisms of vascular development, function, and dysfunction.
Collapse
|
78
|
Heinrich EM, Dimmeler S. MicroRNAs and stem cells: control of pluripotency, reprogramming, and lineage commitment. Circ Res 2012; 110:1014-22. [PMID: 22461365 DOI: 10.1161/circresaha.111.243394] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Stem cells hold great promise for regenerative medicine and the treatment of cardiovascular diseases. The mechanisms regulating self-renewal, pluripotency, and differentiation are not fully understood. MicroRNAs (miRs) are small noncoding RNAs controlling gene expression, either by inducing mRNA degradation or by blocking mRNA translation. The expression of miRs was shown to regulate various aspects of stem cell functions, including the maintenance and induction of pluripotency for reprogramming. In addition, some miRs control cell fate decisions. This review summarizes the role of miRs in reprogramming and embryonic stem cell self-renewal, and specifically addresses the regulation of cardiovascular cell fate decisions by miRs.
Collapse
Affiliation(s)
- Eva-Marie Heinrich
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, University of Frankfurt, Frankfurt, Germany
| | | |
Collapse
|
79
|
Descamps B, Emanueli C. Vascular differentiation from embryonic stem cells: Novel technologies and therapeutic promises. Vascul Pharmacol 2012; 56:267-79. [DOI: 10.1016/j.vph.2012.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 12/04/2011] [Indexed: 01/25/2023]
|
80
|
Hu J, Xie C, Ma H, Yang B, Ma PX, Chen YE. Construction of vascular tissues with macro-porous nano-fibrous scaffolds and smooth muscle cells enriched from differentiated embryonic stem cells. PLoS One 2012; 7:e35580. [PMID: 22545119 PMCID: PMC3335865 DOI: 10.1371/journal.pone.0035580] [Citation(s) in RCA: 16] [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: 01/13/2012] [Accepted: 03/18/2012] [Indexed: 12/31/2022] Open
Abstract
Vascular smooth muscle cells (SMCs) have been broadly used for constructing tissue-engineered blood vessels. However, the availability of mature SMCs from donors or patients is very limited. Derivation of SMCs by differentiating embryonic stem cells (ESCs) has been reported, but not widely utilized in vascular tissue engineering due to low induction efficiency and, hence, low SMC purity. To address these problems, SMCs were enriched from retinoic acid induced mouse ESCs with LacZ genetic labeling under the control of SM22α promoter as the positive sorting marker in the present study. The sorted SMCs were characterized and then cultured on three-dimensional macro-porous nano-fibrous scaffolds in vitro or implanted subcutaneously into nude mice after being seeded on the scaffolds. Our data showed that the LacZ staining, which reflected the corresponding SMC marker SM22α expression level, was efficient as a positive selection marker to dramatically enrich SMCs and eliminate other cell types. After the sorted cells were seeded into the three-dimensional nano-fibrous scaffolds, continuous retinoic acid treatment further enhanced the SMC marker gene expression level while inhibited pluripotent maker gene expression level during the in vitro culture. Meanwhile, after being implanted subcutaneously into nude mice, the implanted cells maintained the positive LacZ staining within the constructs and no teratoma formation was observed. In conclusion, our results demonstrated the potential of SMCs derived from ESCs as a promising cell source for therapeutic vascular tissue engineering and disease model applications.
Collapse
Affiliation(s)
- Jiang Hu
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Changqing Xie
- Department of Internal Medicine, Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Haiyun Ma
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bo Yang
- Department of Cardiac Surgery, Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Peter X. Ma
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (PXM); (YEC)
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (PXM); (YEC)
| |
Collapse
|
81
|
Hunkapiller J, Shen Y, Diaz A, Cagney G, McCleary D, Ramalho-Santos M, Krogan N, Ren B, Song JS, Reiter JF. Polycomb-like 3 promotes polycomb repressive complex 2 binding to CpG islands and embryonic stem cell self-renewal. PLoS Genet 2012; 8:e1002576. [PMID: 22438827 PMCID: PMC3305387 DOI: 10.1371/journal.pgen.1002576] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 01/18/2012] [Indexed: 12/25/2022] Open
Abstract
Polycomb repressive complex 2 (PRC2) trimethylates lysine 27 of histone H3 (H3K27me3) to regulate gene expression during diverse biological transitions in development, embryonic stem cell (ESC) differentiation, and cancer. Here, we show that Polycomb-like 3 (Pcl3) is a component of PRC2 that promotes ESC self-renewal. Using mass spectrometry, we identified Pcl3 as a Suz12 binding partner and confirmed Pcl3 interactions with core PRC2 components by co-immunoprecipitation. Knockdown of Pcl3 in ESCs increases spontaneous differentiation, yet does not affect early differentiation decisions as assessed in teratomas and embryoid bodies, indicating that Pcl3 has a specific role in regulating ESC self-renewal. Consistent with Pcl3 promoting PRC2 function, decreasing Pcl3 levels reduces H3K27me3 levels while overexpressing Pcl3 increases H3K27me3 levels. Furthermore, chromatin immunoprecipitation and sequencing (ChIP-seq) reveal that Pcl3 co-localizes with PRC2 core component, Suz12, and depletion of Pcl3 decreases Suz12 binding at over 60% of PRC2 targets. Mutation of conserved residues within the Pcl3 Tudor domain, a domain implicated in recognizing methylated histones, compromises H3K27me3 formation, suggesting that the Tudor domain of Pcl3 is essential for function. We also show that Pcl3 and its paralog, Pcl2, exist in different PRC2 complexes but bind many of the same PRC2 targets, particularly CpG islands regulated by Pcl3. Thus, Pcl3 is a component of PRC2 critical for ESC self-renewal, histone methylation, and recruitment of PRC2 to a subset of its genomic sites.
Collapse
Affiliation(s)
- Julie Hunkapiller
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Yin Shen
- Ludwig Institute for Cancer Research, School of Medicine, University of California San Diego, San Diego, California, United States of America
| | - Aaron Diaz
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Gerard Cagney
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - David McCleary
- Ludwig Institute for Cancer Research, School of Medicine, University of California San Diego, San Diego, California, United States of America
| | - Miguel Ramalho-Santos
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Nevan Krogan
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, United States of America
| | - Bing Ren
- Ludwig Institute for Cancer Research, School of Medicine, University of California San Diego, San Diego, California, United States of America
| | - Jun S. Song
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Department of Biostatistics and Epidemiology, Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (JSS); (JFR)
| | - Jeremy F. Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (JSS); (JFR)
| |
Collapse
|
82
|
Xie C, Guo Y, Zhu T, Zhang J, Ma PX, Chen YE. Yap1 protein regulates vascular smooth muscle cell phenotypic switch by interaction with myocardin. J Biol Chem 2012; 287:14598-605. [PMID: 22411986 DOI: 10.1074/jbc.m111.329268] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The Hippo-Yap (Yes-associated protein) signaling pathway has emerged as one of the critical pathways regulating cell proliferation, differentiation, and apoptosis in response to environmental and developmental cues. However, Yap1 roles in vascular smooth muscle cell (VSMC) biology have not been investigated. VSMCs undergo phenotypic switch, a process characterized by decreased gene expression of VSMC contractile markers and increased proliferation, migration, and matrix synthesis. The goals of the present studies were to investigate the relationship between Yap1 and VSMC phenotypic switch and to determine the molecular mechanisms by which Yap1 affects this essential process in VSMC biology. Results demonstrated that the expression of Yap1 was rapidly up-regulated by stimulation with PDGF-BB (a known inducer of phenotypic switch in VSMCs) and in the injured vessel wall. Knockdown of Yap1 impaired VSMC proliferation in vitro and enhanced the expression of VSMC contractile genes as well by increasing serum response factor binding to CArG-containing regions of VSMC-specific contractile genes within intact chromatin. Conversely, the interaction between serum response factor and its co-activator myocardin was reduced by overexpression of Yap1 in a dose-dependent manner. Taken together, these results indicate that down-regulation of Yap1 promotes VSMC contractile phenotype by both up-regulating myocardin expression and promoting the association of the serum response factor-myocardin complex with VSMC contractile gene promoters and suggest that the Yap1 signaling pathway is a central regulator of phenotypic switch of VSMCs.
Collapse
Affiliation(s)
- Changqing Xie
- Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | |
Collapse
|
83
|
Chen ML, Liang LS, Wang XK. miR-200c inhibits invasion and migration in human colon cancer cells SW480/620 by targeting ZEB1. Clin Exp Metastasis 2012; 29:457-69. [PMID: 22407310 DOI: 10.1007/s10585-012-9463-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 02/26/2012] [Indexed: 12/19/2022]
Abstract
MicroRNAs are a class of ≈22-nt noncoding single-strand RNAs regulating gene expression postscriptionally. Metastasis caused poor prognosis in colorectal cancer patients and half of the patients developed metastatic lesions when admission. Here we investigated the possible roles of microRNAs in regulating metastasis in the paired colon cancer cells SW480 and SW620. Among those dysregulated microRNAs, miR-200c was speculated to inhibit metastasis by targeting ZEB1. Overexpression of miR-200c was concurrent with downregulation of ZEB1 mRNA and protein. Functional assays demonstrated that modulation of miR-200c with mimics or inhibitors changed potential of metastasis in SW480/620 cancer cells in vitro. Taken together, our study demonstrated that miR-200c inhibits metastatic ability by targeting ZEB1 in colon cancer cells SW480/620 and suggested that modulation of miR-200c could serve as therapeutic tool for inhibiting metastasis in colorectal cancer.
Collapse
Affiliation(s)
- Mian Ling Chen
- Department of General Surgery, Affiliated First People's Municipal Hospital of Guang Zhou, Guang Zhou Medical College, No. 602, Ren Min Bei Road, Guang Zhou 510180, Guang Dong, People's Republic of China.
| | | | | |
Collapse
|
84
|
Zhang L, Zhou Y, Zhu J, Xu Q. An updated view on stem cell differentiation into smooth muscle cells. Vascul Pharmacol 2012; 56:280-7. [PMID: 22421140 DOI: 10.1016/j.vph.2012.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 02/17/2012] [Accepted: 02/28/2012] [Indexed: 12/16/2022]
Abstract
Stem cells possess the ability of self-renewal and give rise to specific cell types. The differentiation of stem cells involves environmental factors, transduction of extra and intra-cellular signals, regulation of gene expression by transcriptional factors, microRNAs and chromosome structural modifiers. Vascular SMCs play a profound role in blood vessel physiology and participate in a number of cardiovascular diseases such as atherosclerosis, hypertension and restenosis. In addition, SMCs could be a crucial cell component for vascular tissue engineering. In this review, we aim to update the recent progress on the mechanisms of SMC differentiation from stem cells, which involve reactive oxygen species, epigenetic modifiers, transcription factors and microRNAs coordinately regulated during stem cell differentiation. We will also discuss the potential application of stem cell therapy for patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou 310003, PR China
| | | | | | | |
Collapse
|
85
|
Spin JM, Maegdefessel L, Tsao PS. Vascular smooth muscle cell phenotypic plasticity: focus on chromatin remodelling. Cardiovasc Res 2012; 95:147-55. [PMID: 22362814 DOI: 10.1093/cvr/cvs098] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Differentiated vascular smooth muscle cells (SMCs) retain the capacity to modify their phenotype in response to inflammation or injury. This phenotypic switching is a crucial component of vascular disease, and is partly dependent on epigenetic regulation. An appreciation has been building in the literature for the essential role chromatin remodelling plays both in SMC lineage determination and in influencing changes in SMC behaviour and state. This process includes numerous chromatin regulatory elements and pathways such as histone acetyltransferases, deacetylases, and methyltransferases and other factors that act at SMC-specific marker sites to silence or permit access to the cellular transcriptional machinery and on other key regulatory elements such as myocardin and Kruppel-like factor 4 (KLF4). Various stimuli known to alter the SMC phenotype, such as transforming growth factor beta (TGF-β), platelet-derived growth factor (PDGF), oxidized phospholipids, and retinoic acid, appear to act in part through effects upon SMC chromatin structure. In recent years, specific covalent histone modifications that appear to establish SMC determinacy have been identified, while others alter the differentiation state. In this article, we review the mechanisms of chromatin remodelling as it applies to the SMC phenotype.
Collapse
Affiliation(s)
- Joshua M Spin
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA 94305, USA
| | | | | |
Collapse
|
86
|
Joshi SR, Comer BS, McLendon JM, Gerthoffer WT. MicroRNA Regulation of Smooth Muscle Phenotype. MOLECULAR AND CELLULAR PHARMACOLOGY 2012; 4:1-16. [PMID: 25309675 PMCID: PMC4190587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Advances in studies of microRNA (miRNA) expression and function in smooth muscles illustrate important effects of small noncoding RNAs on cell proliferation, hypertrophy and differentiation. An emerging theme in miRNA research in a variety of cell types including smooth muscles is that miRNAs regulate protein expression networks to fine tune phenotype. Some widely expressed miRNAs have been described in smooth muscles that regulate important processes in many cell types, such as miR-21 control of proliferation and cell survival. Other miRNAs that are prominent regulators of smooth muscle-restricted gene expression also have targets that control pluripotent cell differentiation. The miR-143~145 cluster which targets myocardin and Kruppel-like factor 4 (KLF4) is arguably the best-described miRNA family in smooth muscles with profound effects on gene expression networks that promote serum response factor (SRF)-dependent contractile and cytoskeletal protein expression and the mature contractile phenotype. Kruppel-family members KLF4 and KLF5 have multiple effects on cell differentiation and are targets for multiple miRNAs in smooth muscles (miR-145, miR-146a, miR-25). The feedback and feedforward loops being defined appear to contribute significantly to vascular and airway remodeling in cardiovascular and respiratory diseases. RNA interference approaches applied to animal models of vascular and respiratory diseases prove that miRNAs and RNA-induced silencing are valid targets for novel anti-remodeling therapies that alter pathological smooth muscle hyperplasia and hypertrophy.
Collapse
Affiliation(s)
- Sachindra R Joshi
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama
| | - Brian S Comer
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama
| | - Jared M McLendon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama
| | - William T Gerthoffer
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama
| |
Collapse
|
87
|
Mansfield JH, McGlinn E. Evolution, Expression, and Developmental Function of Hox-Embedded miRNAs. Curr Top Dev Biol 2012; 99:31-57. [DOI: 10.1016/b978-0-12-387038-4.00002-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
88
|
Rebustini IT, Hayashi T, Reynolds AD, Dillard ML, Carpenter EM, Hoffman MP. miR-200c regulates FGFR-dependent epithelial proliferation via Vldlr during submandibular gland branching morphogenesis. Development 2011; 139:191-202. [PMID: 22115756 DOI: 10.1242/dev.070151] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of epithelial proliferation during organ morphogenesis is crucial for normal development, as dysregulation is associated with tumor formation. Non-coding microRNAs (miRNAs), such as miR-200c, are post-transcriptional regulators of genes involved in cancer. However, the role of miR-200c during normal development is unknown. We screened miRNAs expressed in the mouse developing submandibular gland (SMG) and found that miR-200c accumulates in the epithelial end buds. Using both loss- and gain-of-function, we demonstrated that miR-200c reduces epithelial proliferation during SMG morphogenesis. To identify the mechanism, we predicted miR-200c target genes and confirmed their expression during SMG development. We discovered that miR-200c targets the very low density lipoprotein receptor (Vldlr) and its ligand reelin, which unexpectedly regulate FGFR-dependent epithelial proliferation. Thus, we demonstrate that miR-200c influences FGFR-mediated epithelial proliferation during branching morphogenesis via a Vldlr-dependent mechanism. miR-200c and Vldlr may be novel targets for controlling epithelial morphogenesis during glandular repair or regeneration.
Collapse
Affiliation(s)
- Ivan T Rebustini
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
89
|
Ishida M, El-Mounayri O, Kattman S, Zandstra P, Sakamoto H, Ogawa M, Keller G, Husain M. Regulated expression and role of c-Myb in the cardiovascular-directed differentiation of mouse embryonic stem cells. Circ Res 2011; 110:253-64. [PMID: 22116818 DOI: 10.1161/circresaha.111.259499] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
RATIONALE c-myb null (knockout) embryonic stem cells (ESC) can differentiate into cardiomyocytes but not contractile smooth muscle cells (SMC) in embryoid bodies (EB). OBJECTIVE To define the role of c-Myb in SMC differentiation from ESC. METHODS AND RESULTS In wild-type (WT) EB, high c-Myb levels on days 0-2 of differentiation undergo ubiquitin-mediated proteosomal degradation on days 2.5-3, resurging on days 4-6, without changing c-myb mRNA levels. Activin-A and bone morphogenetic protein 4-induced cardiovascular progenitors were isolated by FACS for expression of vascular endothelial growth factor receptor (VEGFR)2 and platelet-derived growth factor receptor (PDGFR)α. By day 3.75, hematopoesis-capable VEGFR2+ cells were fewer, whereas cardiomyocyte-directed VEGFR2+/PDGFRα+ cells did not differ in abundance in knockout versus WT EB. Importantly, highest and lowest levels of c-Myb were observed in VEGFR2+ and VEGFR2+/PDGFRα+ cells, respectively. Proteosome inhibitor MG132 and lentiviruses enabling inducible expression or knockdown of c-myb were used to regulate c-Myb in WT and knockout EB. These experiments showed that c-Myb promotes expression of VEGFR2 over PDGFRα, with chromatin immunopreciptation and promoter-reporter assays defining specific c-Myb-responsive binding sites in the VEGFR2 promoter. Next, FACS-sorted VEGFR2+ cells expressed highest and lowest levels of SMC- and fibroblast-specific markers, respectively, at days 7-14 after retinoic acid (RA) as compared with VEGFR2+/PDGFRα+ cells. By contrast, VEGFR2+/PDGFRα+ cells cultured without RA beat spontaneously, like cardiomyocytes between days 7 and 14, and expressed cardiac troponin. Notably, RA was required to more fully differentiate SMC from VEGFR2+ cells and completely blocked differentiation of cardiomyocytes from VEGFR2+/PDGFRα+ cells. CONCLUSIONS c-Myb is tightly regulated by proteosomal degradation during cardiovascular-directed differentiation of ESC, expanding early-stage VEGFR2+ progenitors capable of RA-responsive SMC formation.
Collapse
Affiliation(s)
- Masayoshi Ishida
- Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON, Canada, M5G 1L7
| | | | | | | | | | | | | | | |
Collapse
|
90
|
McDonald RA, Hata A, MacLean MR, Morrell NW, Baker AH. MicroRNA and vascular remodelling in acute vascular injury and pulmonary vascular remodelling. Cardiovasc Res 2011; 93:594-604. [PMID: 22065733 DOI: 10.1093/cvr/cvr299] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vascular remodelling is an integral pathological process central to a number of cardiovascular diseases. The complex interplay between distinct cell populations in the vessel wall following vascular injury leads to inflammation, cellular dysfunction, pro-growth signals in the smooth muscle cell (SMC) compartment, and the acquisition of a synthetic phenotype. Although the signals for vascular remodelling are diverse in different pathological contexts, SMC proliferation and migration are consistently observed. It is therefore critical to elucidate key mechanisms central to these processes. MicroRNAs (miRNAs) are small non-coding sequences of RNA that have the capacity to regulate many genes, pathways, and complex biological networks within cells, acting either alone or in concert with one another. In diseases such as cancer and cardiac disease, the role of miRNA in disease pathogenesis has been documented in detail. In contrast, despite a great deal of interest in miRNA, relatively few studies have directly assessed the role of miRNA in vascular remodelling. The potential for modulation of miRNA to achieve therapeutic benefits in this setting is attractive. Here, we focus on the role of miRNA in vascular inflammation and remodelling associated with acute vascular injury (vein graft disease, angioplasty restenosis, and in-stent restenosis) as well as in vascular remodelling associated with the development of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Robert A McDonald
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | | | | | | | | |
Collapse
|
91
|
Manavalan TT, Teng Y, Appana SN, Datta S, Kalbfleisch TS, Li Y, Klinge CM. Differential expression of microRNA expression in tamoxifen-sensitive MCF-7 versus tamoxifen-resistant LY2 human breast cancer cells. Cancer Lett 2011; 313:26-43. [PMID: 21955614 DOI: 10.1016/j.canlet.2011.08.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 08/16/2011] [Accepted: 08/17/2011] [Indexed: 12/17/2022]
Abstract
Microarrays identified miRNAs differentially expressed and 4-hydroxytamoxifen (4-OHT) regulated in MCF-7 endocrine-sensitive versus resistant LY2 human breast cancer cells. 97 miRNAs were differentially expressed in MCF-7 versus LY2 cells. Opposite expression of miRs-10a, 21, 22, 29a, 93, 125b, 181, 200a, 200b, 200c, 205, and 222 was confirmed. Bioinformatic analyses to impute the biological significance of these miRNAs identified 36 predicted gene targets from those regulated by 4-OHT in MCF-7 cells. Agreement in the direction of anticipated regulation was detected for 12 putative targets. These miRNAs with opposite expression between the two cell lines may be involved in endocrine resistance.
Collapse
Affiliation(s)
- Tissa T Manavalan
- Department of Biochemistry and Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | | | | | | | | | | | | |
Collapse
|
92
|
Abstract
The miR-10 microRNA precursor family encodes a group of short non-coding RNAs involved in gene regulation. The miR-10 family is highly conserved and has sparked the interest of many research groups because of the genomic localization in the vicinity of, coexpression with and regulation of the Hox gene developmental regulators. Here, we review the current knowledge of the evolution, physiological function and involvement in cancer of this family of microRNAs.
Collapse
Affiliation(s)
- Disa Tehler
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
93
|
Xie C, Ritchie RP, Huang H, Zhang J, Chen YE. Smooth muscle cell differentiation in vitro: models and underlying molecular mechanisms. Arterioscler Thromb Vasc Biol 2011; 31:1485-94. [PMID: 21677291 DOI: 10.1161/atvbaha.110.221101] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Development of in vitro models by which to study smooth muscle cell (SMC) differentiation has been hindered by some peculiarities intrinsic to these cells, namely their different embryological origins and their ability to undergo phenotypic modulation in cell culture. Although many in vitro models are available for studying SMC differentiation, careful consideration should be taken so that the model chosen fits the questions being posed. In this review, we summarize several well-established in vitro models available to study SMC differentiation from stem cells and outline novel mechanisms recently identified as underlying SMC differentiation programs.
Collapse
Affiliation(s)
- Changqing Xie
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | | | | | | | | |
Collapse
|
94
|
MicroRNAs regulating cell pluripotency and vascular differentiation. Vascul Pharmacol 2011; 55:69-78. [PMID: 21854874 DOI: 10.1016/j.vph.2011.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 07/26/2011] [Accepted: 08/02/2011] [Indexed: 12/12/2022]
Abstract
Human embryonic stem cells (hESC) offer broad potential for regenerative medicine owing to their capacity for self renewal, exponential scale up and differentiation into any cell type in the adult body. hESC have been proposed as a potentially unlimited source for the generation of transplantable, healthy, functional vascular cells for repair of ischemic tissues. To optimally harness this potential necessitates precise control over biological processes that govern maintenance, pluripotency and cell differentiation including signalling cascades, gene expression profiles and epigenetic modification. Such control may be elicited by microRNAs, which are powerful negative regulators of gene expression. Here, we review the role for miRNAs in both the maintenance of pluripotency and differentiation of cells to a cardiovascular lineage including endothelial cells, vascular smooth muscle cells and cardiomyocytes and put this into context for regenerative medicine in the cardiovascular system.
Collapse
|
95
|
Long X, Miano JM. Transforming growth factor-beta1 (TGF-beta1) utilizes distinct pathways for the transcriptional activation of microRNA 143/145 in human coronary artery smooth muscle cells. J Biol Chem 2011; 286:30119-29. [PMID: 21712382 DOI: 10.1074/jbc.m111.258814] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
MicroRNA 143/145 (miR143/145) is restricted to adult smooth muscle cell (SMC) lineages and mediates, in part, the expression of several SMC contractile genes. Although the function of miR143/145 has begun to be elucidated, its transcriptional regulation in response to various signaling inputs is poorly understood. In an effort to define a miR signature for SMC differentiation, we screened human coronary artery SMCs for miRs modulated by TGF-β1, a known stimulus for SMC differentiation. Array analysis revealed a number of TGF-β1-induced miRs, including miR143/145. Validation studies showed that TGF-β1 stimulated miR143/145 expression in a dose- and time-dependent manner. We utilized several chemical inhibitors and found that SB203580, a specific inhibitor of p38MAPK, significantly decreased TGF-β1-induced miR143/145 expression. siRNA studies demonstrated that the effect of TGF-β1 on miR143/145 was dependent upon the myocardin and serum response factor transcriptional switch as well as SMAD4. TGF-β1 stimulated a 580-bp human miR143/145 enhancer, and mutagenesis studies revealed a critical role for both a known CArG box and an adjacent SMAD-binding element for full TGF-β1-dependent activation of the enhancer. Chromatin immunoprecipitation assays documented TGF-β1-mediated enrichment of SMAD3 and SMAD4 binding over the enhancer region containing the SMAD-binding element. Pre-miR145 strongly promoted SMC differentiation, whereas an anti-miR145 partially blocked TGF-β1-induced SMC differentiation. These results demonstrate a dual pathway for TGF-β1-induced transcription of miR143/145, thus revealing a novel mechanism underlying TGF-β1-induced human vascular SMC differentiation.
Collapse
Affiliation(s)
- Xiaochun Long
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA.
| | | |
Collapse
|
96
|
Ma X, Becker Buscaglia LE, Barker JR, Li Y. MicroRNAs in NF-kappaB signaling. J Mol Cell Biol 2011; 3:159-66. [PMID: 21502305 DOI: 10.1093/jmcb/mjr007] [Citation(s) in RCA: 476] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nuclear factor κB (NF-κB) is a transcriptional factor that regulates a battery of genes that are critical to innate and adaptive immunity, cell proliferation, inflammation, and tumor development. MicroRNAs (miRNAs) are short RNA molecules of 20-25 nucleotides in length that negatively regulate gene expression in animals and plants primarily by targeting 3' untranslated regions of mRNAs. In this work, we review the convergence of miRNAs and NF-κB signaling and dysregulation of miRNAs and NF-κB activation in human diseases, particularly in cancer. The function of miR-146, miR-155, miR-181b, miR-21, and miR-301a in NF-κB activation and their impact on tumorigenesis are discussed. Given that over 1000 human miRNAs have been identified, rendering miRNAs one of the most abundant classes of regulatory molecules, deciphering their biological function and pathological contribution in NF-κB dysregulation is essential to appreciate the complexity of immune systems and to develop therapeutics against cancer.
Collapse
Affiliation(s)
- Xiaodong Ma
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, 319 Abraham Flexner Way, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
97
|
Abstract
The peroxisome proliferator-activated receptors (PPARs) and the retinoid X receptors (RXRs) are ligand-activated transcription factors that coordinately regulate gene expression. This PPAR-RXR transcriptional complex plays a critical role in energy balance, including triglyceride metabolism, fatty acid handling and storage, and glucose homeostasis: processes whose dysregulation characterize obesity, diabetes, and atherosclerosis. PPARs and RXRs are also involved directly in inflammatory and vascular responses in endothelial and vascular smooth muscle cells. New insights into fundamental aspects of PPAR and RXR biology, and their actions in the vasculature, continue to appear. Although RXRs are obligate heterodimeric partners for PPAR action, the part that RXRs, and their endogenous retinoid mediators, exert in the vessel wall is less well understood. Biological insights into PPAR-RXRs may help inform interpretation of clinical trials with synthetic PPAR agonists and prospects for future PPAR therapeutics. Importantly, the extensive data establishing a key role for PPARs and RXRs in energy balance, inflammation, and vascular biology stands separately from the clinical experience with any given synthetic PPAR agonist. Both the basic science data and the clinical experience with PPAR agonists identify the need to better understand these important transcriptional regulators.
Collapse
Affiliation(s)
- Jorge Plutzky
- From Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
98
|
A systematic analysis of the skeletal muscle miRNA transcriptome of chicken varieties with divergent skeletal muscle growth identifies novel miRNAs and differentially expressed miRNAs. BMC Genomics 2011; 12:186. [PMID: 21486491 PMCID: PMC3107184 DOI: 10.1186/1471-2164-12-186] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Accepted: 04/13/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Functional studies have demonstrated that microRNAs (miRNAs or miRs) play critical roles in a wide spectrum of biological processes including development and disease pathogenesis. To investigate the functional roles that miRNAs play during chicken skeletal muscle development, the miRNA transcriptomes of skeletal muscles from broiler and layer chickens were profiled using Solexa deep sequencing. RESULTS Some miRNAs have multiple isoforms and several miRNAs* are present at higher levels than their corresponding miRNAs. Thirty three novel and 189 known chicken miRNAs were identified using computational approaches. Subsequent miRNA transcriptome comparisons and real-time PCR validation experiments revealed 17 miRNAs that were differentially expressed between broilers and layers, and a number of targets of these miRNAs have been implicated in myogenesis regulation. Using integrative miRNA target-prediction and network-analysis approaches an interaction network of differentially expressed and muscle-related miRNAs and their putative targets was constructed, and miRNAs that could contribute to the divergent muscle growth of broiler and layer chickens by targeting the ACVR2B gene were identified, which can causes dramatic increases in muscle mass. CONCLUSIONS The present study provides the first transcriptome profiling-based evaluation of miRNA function during skeletal muscle development in chicken. Systematic predictions aided the identification of potential miRNAs and their targets, which could contribute to divergent muscle growth in broiler and layer chickens. Furthermore, these predictions generated information that can be utilized in further research investigating the involvement of interaction networks, containing miRNAs and their targets, in the regulation of muscle development.
Collapse
|
99
|
Kane NM, Xiao Q, Baker AH, Luo Z, Xu Q, Emanueli C. Pluripotent stem cell differentiation into vascular cells: A novel technology with promises for vascular re(generation). Pharmacol Ther 2011; 129:29-49. [DOI: 10.1016/j.pharmthera.2010.10.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/05/2010] [Indexed: 12/15/2022]
|
100
|
Abstract
Vascular smooth muscle cells (VSMCs) exhibit extraordinary plasticity during postnatal development. Vascular injury initiates VSMC phenotypic switch from the contractile to proliferative phenotype, which plays a central role in vascular lesion formation and diverse vascular diseases. MicroRNAs (miRNAs) regulate gene expression posttranscriptionally by either degrading target mRNAs or repressing their translation. Emerging evidence has revealed miRNAs are critical regulators in VSMC differentiation from stem cells, phenotypic switch, and various vascular pathogenesis. Here, we review recent advances regarding functions of specific miRNAs in vasculature and discuss possible mechanisms by which miRNAs affect VSMC biology.
Collapse
Affiliation(s)
- Changqing Xie
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|