101
|
Zilahi E, Adamecz Z, Bodoki L, Griger Z, Póliska S, Nagy-Vincze M, Dankó K. Dysregulated expression profile of myomiRs in the skeletal muscle of patients with polymyositis. EJIFCC 2019; 30:237-245. [PMID: 31372109 PMCID: PMC6599196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
MicroRNA (miRNA) research has intensively developed over the past decade. Characterization of dysregulated miRNA expression profiles could give a better understanding of the development of pathological conditions and clinical disorders, such as autoimmune diseases with polygenic etiology, including idiopathic inflammatory myopathies (IIMs). IIMs are a group of rare autoimmune disorders characterized by skeletal weakness and inflammation. Polymyositis (PM) is one of the conditions of autoimmune myopathies with proximal skeletal muscle weakness. A novel group of miRNAs, known as myomiRs are described as striated muscle-specific or muscle-enriched miRNAs. They are involved in myoblast proliferation/differentiation as well as muscle regeneration. To determine the role of myomiRs in the development and progression of PM, we performed an initial skeletal muscle miRNA profiling using microarray technique at diagnosis. The aim of the study was to examine myomiRs expression profile in patients with PM in order to remark the association between the dysregulated myomiRs' expression and the development of the disease. As a results of microarray investigation, most of the myomiRs showed altered expression patterns in the muscle samples of PM patients compared to controls. These results suggest that myomiRs, especially miR-1, miR-133a, miR-208b, miR-486, and miR-499 function in a network, and are associated with the development of PM.
Collapse
Affiliation(s)
- Erika Zilahi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Hungary
| | - Zsuzsanna Adamecz
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Hungary
| | - Levente Bodoki
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Hungary
| | - Zoltán Griger
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatic Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Hungary
| | - Melinda Nagy-Vincze
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary
| | - Katalin Dankó
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Hungary
| |
Collapse
|
102
|
Lim TB, Aliwarga E, Luu TDA, Li YP, Ng SL, Annadoray L, Sian S, Ackers-Johnson MA, Foo RSY. Targeting the highly abundant circular RNA circSlc8a1 in cardiomyocytes attenuates pressure overload induced hypertrophy. Cardiovasc Res 2019; 115:1998-2007. [DOI: 10.1093/cvr/cvz130] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/02/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022] Open
Abstract
AbstractAimsWe and others have previously described the expression landscape of circular RNA (circRNA) in mouse and human hearts. However, the functional relevance of many of these abundantly expressed cardiomyocyte circRNA remains to be fully explored. Among the most abundant circRNA, one stems from the sodium-calcium exchanger gene, Slc8a1, exon 2 locus. Because of its very high abundance in cardiomyocytes we investigated the possible role of circSlc8a1 in the heart.Methods and resultsWe performed a miRNA screen using an array of 752 miRNAs with RNA recovered from a pull-down of endogenous cardiomyocyte circSlc8a1. MicroRNA-133a (miR-133a), with a prior well-recognized role in cardiac hypertrophy, was highly enriched in the fraction of circSlc8a1 pull-down (adjusted P-value < 0.001). We, therefore, followed-up validation of the functional interaction between circSlc8a1 and miR-133 using luciferase assays and reciprocal pull-down assays. In vivo, AAV9-mediated RNAi knockdown of circSlc8a1 attenuates cardiac hypertrophy from pressure-overload, whereas forced cardiomyocyte specific overexpression of circSlc8a1 resulted in heart failure. Molecular analyses showed targets of miR-133a including serum response factor (Srf), connective tissue growth factor (Ctgf), adrenoceptor beta 1 (Adrb1), and adenylate cyclase 6 (Adcy6) to be regulated by circSlc8a1-directed intervention of knockdown and overexpression.ConclusionIn summary, circSlc8a1 can function as an endogenous sponge for miR-133a in cardiomyocytes. We propose that circSlc8a1 may serve as a novel therapeutic target for cardiac hypertrophy.
Collapse
Affiliation(s)
- Tingsen Benson Lim
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
- Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore, Singapore
| | - Edita Aliwarga
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
- Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore, Singapore
| | - Tuan Danh Anh Luu
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
| | - Yiqing Peter Li
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
| | - Shi Ling Ng
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
- Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore, Singapore
| | - Lavenniah Annadoray
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
- Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore, Singapore
| | - Stephanie Sian
- Cancer Science Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
| | - Matthew Andrew Ackers-Johnson
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
| | - Roger Sik-Yin Foo
- Cardiovascular Research Institute, National University Health Systems, MD6 Centre for Translational Medicine, 14 Medical Drive, Singapore, Singapore
- Genome Institute of Singapore, Genome, 60 Biopolis Street, Singapore, Singapore
| |
Collapse
|
103
|
miR-206 inhibits cell proliferation, invasion, and migration by down-regulating PTP1B in hepatocellular carcinoma. Biosci Rep 2019; 39:BSR20181823. [PMID: 31048362 PMCID: PMC6522750 DOI: 10.1042/bsr20181823] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/11/2019] [Accepted: 04/17/2019] [Indexed: 01/06/2023] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) has been reported as an oncogene in hepatocellular carcinoma (HCC). However, how PTP1B is regulated in HCC remains unclear. MicroRNAs (miRNAs) are a class of small non-coding RNAs involved many biological processes including tumorigenesis. In this study, we investigated whether miRNA participated in the regulation of PTP1B in HCC. We found that miR-206, which was down-regulated during tumorigenesis, inhibited HCC cell proliferation and invasion. Overexpression of miR-206 inhibited proliferation, invasion, and migration of HCC cell lines HepG2 and Huh7. Mechanistically, we demonstrated that miR-206 directly targeted PTP1B by binding to the 3′-UTR of PTP1B mRNA as demonstrated by the luciferase reporter assay. Overexpression miR-206 inhibited PTP1B expression while miR-206 inhibition enhanced PTP1B expression in HepG2 and Huh7 cells. Functionally, the regulatory effect on cell proliferation/migration/invasion of miR-206 was reversed by PTP1B overexpression. Furthermore, tumor inoculation nude mice model was used to explore the function of miR-206 in vivo. Our results showed that overexpression of miR-206 drastically inhibited tumor development. In summary, our data suggest that miR-206 inhibits HCC development by targeting PTP1B.
Collapse
|
104
|
Wang Y, Luo X, Liu Y, Han G, Sun D. Long noncoding RNA RMRP promotes proliferation and invasion via targeting miR‐1‐3p in non–small‐cell lung cancer. J Cell Biochem 2019; 120:15170-15181. [PMID: 31050363 DOI: 10.1002/jcb.28779] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/30/2018] [Accepted: 01/09/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Yi Wang
- Department of Clinical Laboratory The Third Affiliated Hospital of Jinzhou Medical University Jinzhou Liaoning P.R. China
| | - Xigang Luo
- Department of Clinical Laboratory The Third Affiliated Hospital of Jinzhou Medical University Jinzhou Liaoning P.R. China
| | - Yang Liu
- Department of Clinical Laboratory The Third Affiliated Hospital of Jinzhou Medical University Jinzhou Liaoning P.R. China
| | - Guanying Han
- Department of Medical The First Affiliated Hospital of Jinzhou Medical University Jinzhou Liaoning P.R. China
| | - Dapeng Sun
- Department of Medical The First Affiliated Hospital of Jinzhou Medical University Jinzhou Liaoning P.R. China
| |
Collapse
|
105
|
Zhang Z, Chen Y, Li B, Yao Y, Jiang A, Wei W, Liu H, Wu W. Identification of a novel miR-206-Notch3 pathway regulating mouse myoblasts proliferation. Gene 2019; 695:57-64. [DOI: 10.1016/j.gene.2019.01.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/05/2019] [Accepted: 01/22/2019] [Indexed: 10/27/2022]
|
106
|
Genome-Wide Investigation and Functional Analysis of Sus scrofa RNA Editing Sites across Eleven Tissues. Genes (Basel) 2019; 10:genes10050327. [PMID: 31052161 PMCID: PMC6562383 DOI: 10.3390/genes10050327] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/08/2019] [Accepted: 04/17/2019] [Indexed: 01/24/2023] Open
Abstract
Recently, the prevalence and importance of RNA editing have been illuminated in mammals. However, studies on RNA editing of pigs, a widely used biomedical model animal, are rare. Here we collected RNA sequencing data across 11 tissues and identified more than 490,000 RNA editing sites. We annotated their biological features, detected flank sequence characteristics of A-to-I editing sites and the impact of A-to-I editing on miRNA-mRNA interactions, and identified RNA editing quantitative trait loci (edQTL). Sus scrofa RNA editing sites showed high enrichment in repetitive regions with a median editing level as 15.38%. Expectedly, 96.3% of the editing sites located in non-coding regions including intron, 3' UTRs, intergenic, and gene proximal regions. There were 2233 editing sites located in the coding regions and 980 of them caused missense mutation. Our results indicated that to an A-to-I editing site, the adjacent four nucleotides, two before it and two after it, have a high impact on the editing occurrences. A commonly observed editing motif is CCAGG. We found that 4552 A-to-I RNA editing sites could disturb the original binding efficiencies of miRNAs and 4176 A-to-I RNA editing sites created new potential miRNA target sites. In addition, we performed edQTL analysis and found that 1134 edQTLs that significantly affected the editing levels of 137 RNA editing sites. Finally, we constructed PRESDB, the first pig RNA editing sites database. The site provides necessary functions associated with Sus scrofa RNA editing study.
Collapse
|
107
|
Sannicandro AJ, Soriano-Arroquia A, Goljanek-Whysall K. Micro(RNA)-managing muscle wasting. J Appl Physiol (1985) 2019; 127:619-632. [PMID: 30991011 DOI: 10.1152/japplphysiol.00961.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Progressive skeletal muscle wasting is a natural consequence of aging and is common in chronic and acute diseases. Loss of skeletal muscle mass and function (strength) often leads to frailty, decreased independence, and increased risk of hospitalization. Despite progress made in our understanding of the mechanisms underlying muscle wasting, there is still no treatment available, with exercise training and dietary supplementation improving, but not restoring, muscle mass and/or function. There has been slow progress in developing novel therapies for muscle wasting, either during aging or disease, partially due to the complex nature of processes underlying muscle loss. The mechanisms of muscle wasting are multifactorial, with a combination of factors underlying age- and disease-related functional muscle decline. These factors include well-characterized changes in muscle such as changes in protein turnover and more recently described mechanisms such as autophagy or satellite cell senescence. Advances in transcriptomics and other high-throughput approaches have highlighted significant deregulation of skeletal muscle gene and protein levels during aging and disease. These changes are regulated at different levels, including posttranscriptional gene expression regulation by microRNAs. microRNAs, potent regulators of gene expression, modulate many processes in muscle, and microRNA-based interventions have been recently suggested as a promising new therapeutic strategy against alterations in muscle homeostasis. Here, we review recent developments in understanding the aging-associated mechanisms of muscle wasting and explore potential microRNA-based therapeutic avenues.
Collapse
Affiliation(s)
- Anthony J Sannicandro
- Department of Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - Ana Soriano-Arroquia
- Institute of Ageing and Chronic Disease, University of Liverpool, United Kingdom
| | - Katarzyna Goljanek-Whysall
- Department of Physiology, School of Medicine, National University of Ireland, Galway, Ireland.,Institute of Ageing and Chronic Disease, University of Liverpool, United Kingdom
| |
Collapse
|
108
|
Cui S, Li L, Mubarokah SN, Meech R. Wnt/β‐catenin signaling induces the myomiRs miR‐133b and miR‐206 to suppress Pax7 and induce the myogenic differentiation program. J Cell Biochem 2019; 120:12740-12751. [DOI: 10.1002/jcb.28542] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Shuang Cui
- Department of Clinical Pharmacology, College of Medicine and Public Health Flinders University Bedford Park South Australia Australia
- Department of Physiology Shandong University School of Medicine Jinan Shandong China
| | - Liang Li
- Department of Biochemistry Flinders University, College of Medicine and Public Health Bedford Park South Australia Australia
- Department of Biochemistry University of Adelaide Adelaide South Australia Australia
| | - Siti Nurul Mubarokah
- Department of Clinical Pharmacology, College of Medicine and Public Health Flinders University Bedford Park South Australia Australia
| | - Robyn Meech
- Department of Clinical Pharmacology, College of Medicine and Public Health Flinders University Bedford Park South Australia Australia
| |
Collapse
|
109
|
Vechetti IJ, Wen Y, Chaillou T, Murach KA, Alimov AP, Figueiredo VC, Dal-Pai-Silva M, McCarthy JJ. Life-long reduction in myomiR expression does not adversely affect skeletal muscle morphology. Sci Rep 2019; 9:5483. [PMID: 30940834 PMCID: PMC6445125 DOI: 10.1038/s41598-019-41476-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 03/11/2019] [Indexed: 12/20/2022] Open
Abstract
We generated an inducible, skeletal muscle-specific Dicer knockout mouse to deplete microRNAs in adult skeletal muscle. Following tamoxifen treatment, Dicer mRNA expression was significantly decreased by 87%. Wild-type (WT) and Dicer knockout (KO) mice were subjected to either synergist ablation or hind limb suspension for two weeks. There was no difference in muscle weight with hypertrophy or atrophy between WT and KO groups; however, even with the significant loss of Dicer expression, myomiR (miR-1, -133a and -206) expression was only reduced by 38% on average. We next aged WT and KO mice for ~22 months following Dicer inactivation to determine if myomiR expression would be further reduced over a prolonged timeframe and assess the effects of myomiR depletion on skeletal muscle phenotype. Skeletal muscle Dicer mRNA expression remained significantly decreased by 80% in old KO mice and sequencing of cloned Dicer mRNA revealed the complete absence of the floxed exons in KO skeletal muscle. Despite a further reduction of myomiR expression to ~50% of WT, no change was observed in muscle morphology between WT and KO groups. These results indicate the life-long reduction in myomiR levels did not adversely affect skeletal muscle phenotype and suggest the possibility that microRNA expression is uniquely regulated in skeletal muscle.
Collapse
Affiliation(s)
- Ivan J Vechetti
- Department of Physiology, College of Medicine, University of Kentucky, Kentucky, USA
- Center for Muscle Biology University of Kentucky, Lexington, Kentucky, USA
- Department of Morphology, São Paulo State University, Institute of Biosciences, Botucatu, Brazil
| | - Yuan Wen
- Department of Physiology, College of Medicine, University of Kentucky, Kentucky, USA
- Center for Muscle Biology University of Kentucky, Lexington, Kentucky, USA
| | - Thomas Chaillou
- Örebro University, School of Health Sciences, Örebro, Sweden
| | - Kevin A Murach
- Department of Rehabilitation Sciences, College of Health Sciences, Kentucky, USA
- Center for Muscle Biology University of Kentucky, Lexington, Kentucky, USA
| | - Alexander P Alimov
- Department of Physiology, College of Medicine, University of Kentucky, Kentucky, USA
- Center for Muscle Biology University of Kentucky, Lexington, Kentucky, USA
| | - Vandre C Figueiredo
- Department of Physiology, College of Medicine, University of Kentucky, Kentucky, USA
- Department of Rehabilitation Sciences, College of Health Sciences, Kentucky, USA
- Center for Muscle Biology University of Kentucky, Lexington, Kentucky, USA
| | - Maeli Dal-Pai-Silva
- Department of Morphology, São Paulo State University, Institute of Biosciences, Botucatu, Brazil
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Kentucky, USA.
- Center for Muscle Biology University of Kentucky, Lexington, Kentucky, USA.
| |
Collapse
|
110
|
Wang Y, Tai Q, Zhang J, Kang J, Gao F, Zhong F, Cai L, Fang F, Gao Y. MiRNA-206 inhibits hepatocellular carcinoma cell proliferation and migration but promotes apoptosis by modulating cMET expression. Acta Biochim Biophys Sin (Shanghai) 2019; 51:243-253. [PMID: 30805592 DOI: 10.1093/abbs/gmy119] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/22/2018] [Indexed: 12/25/2022] Open
Abstract
A close relationship between cancer progression and microRNAs (miRNAs) regulation has been demonstrated. Abnormal microRNA-206 (miR-206) expression has been shown to be related to the development of malignancies. However, the role of miR-206 in hepatocellular carcinoma (HCC) remains unclear. Here, we evaluated the function of miR-206 in HCC. Results showed that miR-206 expression was decreased in 27 human HCC tissues compared with that of adjacent normal tissues. Conversely, cMET was up-regulated in human HCC cancer tissues, and cMET levels were shown to be negatively correlated with miR-206 expression. Abnormally increased miR-206 expression in three HCC cell lines (SMMC-7721, HepG2, and Huh7) attenuated cell viability, migration, and invasion. Increased apoptosis was also observed in these miR-206 expressing cells. Furthermore, we identified that miR-206 targets the 3'-UTR of the cMET gene for silencing, and restoration of cMET expression reversed the inhibitory effect of miR-206 on HCC. Tumor cells expressing miR-206 also showed delayed growth in the in vivo experiments compared with the controls. Altogether, our findings provide new insights into the molecular mechanisms of HCC oncogenesis.
Collapse
Affiliation(s)
- Yuanxi Wang
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Qinwen Tai
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jinhui Zhang
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Junsheng Kang
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Feng Gao
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Feng Zhong
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Liquan Cai
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Fa Fang
- Department of General Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
111
|
Shrestha S, Phay M, Kim HH, Pouladvand P, Lee SJ, Yoo S. Differential regulation of brain-derived neurotrophic factor (BDNF) expression in sensory neuron axons by miRNA-206. FEBS Open Bio 2019; 9:374-383. [PMID: 30761261 PMCID: PMC6356166 DOI: 10.1002/2211-5463.12581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 01/27/2023] Open
Abstract
Distinct subcellular localization and subsequent translational control of 3′ UTR variants of mRNA encoding brain‐derived neurotrophic factor (BDNF) are critical for the development and plasticity of neurons. Although the processes that lead to preferential localization of BDNF have been well studied, it is still not clear how neurons ensure differential BDNF production in a spatial‐specific manner. Here, we identified that microRNA (miRNA)‐206 has the potential to specifically regulate BDNF with a long 3′ UTR without affecting its short 3′ UTR counterpart. Overexpression of miRNA‐206 in sensory neurons resulted in a 30% and 45% reduction of BDNF protein expression in the cell bodies and axons, respectively. The work described in the present study indicates that miRNAs can differentially and specifically regulate the expression of transcript variants with different localization patterns.
Collapse
Affiliation(s)
- Shiva Shrestha
- Nemours Biomedical Research Alfred I. duPont Hospital for Children Wilmington DE USA.,Department of Biological Sciences University of Delaware Newark DE USA.,Gene Therapy Program Perelman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Monichan Phay
- Nemours Biomedical Research Alfred I. duPont Hospital for Children Wilmington DE USA.,Department of Biological Sciences University of Delaware Newark DE USA
| | - Hak Hee Kim
- Nemours Biomedical Research Alfred I. duPont Hospital for Children Wilmington DE USA
| | - Pedram Pouladvand
- Department of Biological Sciences University of Delaware Newark DE USA.,Department of Biology Pennsylvania State University-Brandywine Media PA USA
| | - Seung Joon Lee
- Department of Biological Sciences University of South Carolina Columbia SC USA
| | - Soonmoon Yoo
- Nemours Biomedical Research Alfred I. duPont Hospital for Children Wilmington DE USA.,Department of Biological Sciences University of Delaware Newark DE USA
| |
Collapse
|
112
|
Pegoraro V, Merico A, Angelini C. MyomiRNAs Dysregulation in ALS Rehabilitation. Brain Sci 2019; 9:8. [PMID: 30634563 PMCID: PMC6356197 DOI: 10.3390/brainsci9010008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/21/2018] [Accepted: 01/05/2019] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare, progressive, neurodegenerative disorder caused by degeneration of upper and lower motor neurons. The disease process leads, because of lower motor neuron involvement, to progressive muscle atrophy, weakness, and fasciculations and for the upper motor neuron involvement leads to spasticity. Muscle atrophy in ALS is caused by a neural dysregulation in the molecular network controlling fast and slow muscle fibers. Denervation and reinnervation processes in skeletal muscle occur in the course of ALS and are modulated by rehabilitation. MicroRNAs (miRNAs) are small, non-coding RNAs that are involved in different biological functions under various pathophysiological conditions. MiRNAs can be secreted by various cell types and they are markedly stable in body fluids. MiR-1, miR-133 a miR-133b, and miR-206 are called "myomiRs" and are considered markers of myogenesis during muscle regeneration and contribute to neuromuscular junction stabilization or sprouting. We observed a positive effect of a standard aerobic exercise rehabilitative protocol conducted for six weeks in 18 ALS patients during hospitalization in our center. This is a preliminary study, in which we correlated clinical scales with molecular data on myomiRs. After six weeks of moderate aerobic exercise, we found lower levels in serum of myomiRNAs. Our data suggest that circulating miRNAs changed during skeletal muscle recovery in response to physical rehabilitation in ALS. However, no firm conclusions can be made on the ALS-specific effect of exercise on miRNA levels.
Collapse
Affiliation(s)
- Valentina Pegoraro
- Fondazione Ospedale San Camillo IRCCS, via Alberoni 70, 30126 Venezia, Italy.
| | - Antonio Merico
- Fondazione Ospedale San Camillo IRCCS, via Alberoni 70, 30126 Venezia, Italy.
| | - Corrado Angelini
- Fondazione Ospedale San Camillo IRCCS, via Alberoni 70, 30126 Venezia, Italy.
| |
Collapse
|
113
|
Tarnowski M, Tkacz M, Kopytko P, Bujak J, Piotrowska K, Pawlik A. Trichostatin A Inhibits Rhabdomyosarcoma Proliferation and Induces Differentiation through MyomiR Reactivation. Folia Biol (Praha) 2019; 65:43-52. [PMID: 31171081 DOI: 10.14712/fb2019065010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Rhabdomyosarcoma (RMS) is a malignant tumour of soft tissues, occurring mainly in children and young adults. RMS cells derive from muscle cells, which due to mutations and epigenetic modifications have lost their ability to differentiate. Epigenetic modifications regulate expression of genes responsible for cell proliferation, maturation, differentiation and apoptosis. HDAC inhibitors suppress histone acetylation; therefore, they are a promising tool used in cancer therapy. Trichostatin A (TsA) is a pan-inhibitor of HDAC. In our study, we investigated the effect of TsA on RMS cell biology. Our findings strongly suggest that TsA inhibits RMS cell proliferation, induces cell apoptosis, and reactivates tumour cell differentiation. TsA up-regulates miR-27b expression, which is involved in the process of myogenesis. Moreover, TsA increases susceptibility of RMS cells to routinely used chemotherapeutics. In conclusion, TsA exhibits anti-cancer properties, triggers differentiation, and thereby can complement an existing spectrum of chemotherapeutics used in RMS therapy.
Collapse
Affiliation(s)
- M Tarnowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - M Tkacz
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - P Kopytko
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - J Bujak
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - K Piotrowska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - A Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
114
|
Kemp PR, Griffiths M, Polkey MI. Muscle wasting in the presence of disease, why is it so variable? Biol Rev Camb Philos Soc 2018; 94:1038-1055. [PMID: 30588725 DOI: 10.1111/brv.12489] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 12/16/2022]
Abstract
Skeletal muscle wasting is a common clinical feature of many chronic diseases and also occurs in response to single acute events. The accompanying loss of strength can lead to significant disability, increased care needs and have profound negative effects on quality of life. As muscle is the most abundant source of amino acids in the body, it appears to function as a buffer for fuel and substrates that can be used to repair damage elsewhere and to feed the immune system. In essence, the fundamentals of muscle wasting are simple: less muscle is made than is broken down. However, although well-described mechanisms modulate muscle protein turnover, significant individual differences in the amount of muscle lost in the presence of a given severity of disease complicate the understanding of underlying mechanisms and suggest that individuals have different sensitivities to signals for muscle loss. Furthermore, the rate at which muscle protein is turned over under normal conditions means that clinically significant muscle loss can occur with changes in the rate of protein synthesis and/or breakdown that are too small to be measurable. Consequently, the changes in expression of factors regulating muscle turnover required to cause a decline in muscle mass are small and, except in cases of rapid wasting, there is no consistent pattern of change in the expression of factors that regulate muscle mass. MicroRNAs are fine tuners of cell phenotype and are therefore ideally suited to cause the subtle changes in proteome required to tilt the balance between synthesis and degradation in a way that causes clinically significant wasting. Herein we present a model in which muscle loss as a consequence of disease in non-muscle tissue is modulated by a set of microRNAs, the muscle expression of which is associated with severity of disease in the non-muscle tissue. These microRNAs alter fundamental biological processes including the synthesis of ribosomes and mitochondria leading to reduced protein synthesis and increased protein breakdown, thereby freeing amino acids from the muscle. We argue that the variability in muscle loss observed in the human population arises from at least two sources. The first is from pre-existing or disease-induced variation in the expression of microRNAs controlling the sensitivity of muscle to the atrophic signal and the second is from the expression of microRNAs from imprinted loci (i.e. only expressed from the maternally or paternally inherited allele) and may control the rate of myonuclear recruitment. In the absence of disease, these factors do not correlate with muscle mass, since there is no challenge to the established balance. However, in the presence of such a challenge, these microRNAs determine the rate of decline for a given disease severity. Together these mechanisms provide novel insight into the loss of muscle mass and its variation in the human population. The involvement of imprinted loci also suggests that genes that regulate early development also contribute to the ability of individuals to resist muscle loss in response to disease.
Collapse
Affiliation(s)
- Paul R Kemp
- National Heart & Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, U.K
| | - Mark Griffiths
- National Heart & Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, U.K
| | - Michael I Polkey
- National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, Sydney Street, London SW3 6NP, U.K
| |
Collapse
|
115
|
Lock MC, Tellam RL, Botting KJ, Wang KCW, Selvanayagam JB, Brooks DA, Seed M, Morrison JL. The role of miRNA regulation in fetal cardiomyocytes, cardiac maturation and the risk of heart disease in adults. J Physiol 2018; 596:5625-5640. [PMID: 29785790 PMCID: PMC6265572 DOI: 10.1113/jp276072] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction is a primary contributor towards the global burden of cardiovascular disease. Rather than repairing the existing damage of myocardial infarction, current treatments only address the symptoms of the disease and reducing the risk of a secondary infarction. Cardiac regenerative capacity is dependent on cardiomyocyte proliferation, which concludes soon after birth in humans and precocial species such as sheep. Human fetal cardiac tissue has some ability to repair following tissue damage, whereas a fully matured human heart has minimal capacity for cellular regeneration. This is in contrast to neonatal mice and adult zebrafish hearts, which retain the ability to undergo cardiomyocyte proliferation and can regenerate cardiac tissue after birth. In mice and zebrafish models, microRNAs (miRNAs) have been implicated in the regulation of genes involved in cardiac cell cycle progression and regeneration. However, the significance of miRNA regulation in cardiomyocyte proliferation for humans and other large mammals, where the timing of heart development in relation to birth is similar, remains unclear. miRNAs may be valuable targets for therapies that promote cardiac repair after injury. Therefore, elucidating the role of specific miRNAs in large animals, where heart development closely resembles that of humans, remains vitally important for identifying therapeutic targets that may be translated into clinical practice focused on tissue repair.
Collapse
Affiliation(s)
- Mitchell C. Lock
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Ross L. Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Kimberley J. Botting
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Kimberley C. W. Wang
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
- School of Human SciencesUniversity of Western AustraliaCrawleyWA 6009Australia
| | - Joseph B. Selvanayagam
- Cardiac Imaging Research Group, Department of Heart HealthSouth Australian Health & Medical Research Institute, and Flinders UniversityGPO Box 2100AdelaideSA 5001Australia
| | - Doug A. Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Mike Seed
- Hospital for Sick Children, Division of Cardiology555 University AvenueTorontoON M5G 1X8Canada
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| |
Collapse
|
116
|
Wu N, Gu T, Lu L, Cao Z, Song Q, Wang Z, Zhang Y, Chang G, Xu Q, Chen G. Roles of miRNA‐1 and miRNA‐133 in the proliferation and differentiation of myoblasts in duck skeletal muscle. J Cell Physiol 2018; 234:3490-3499. [DOI: 10.1002/jcp.26857] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/23/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Ningzhao Wu
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Tiantian Gu
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Lu Lu
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Zhengfeng Cao
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Qianqian Song
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Zhixiu Wang
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Yang Zhang
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Guobin Chang
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Qi Xu
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| | - Guohong Chen
- Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province Yangzhou University Yangzhou China
| |
Collapse
|
117
|
Sessa F, Salerno M, Di Mizio G, Bertozzi G, Messina G, Tomaiuolo B, Pisanelli D, Maglietta F, Ricci P, Pomara C. Anabolic Androgenic Steroids: Searching New Molecular Biomarkers. Front Pharmacol 2018; 9:1321. [PMID: 30524281 PMCID: PMC6256094 DOI: 10.3389/fphar.2018.01321] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Even if anabolic androgenic steroid (AAS) abuse is clearly associated with a wide spectrum of collateral effects, adolescents and athletes frequently use a large group of synthetic derivatives of testosterone, both for aesthetic uses and for improving performance. Over the last few years, the development of MicroRNA (miRNA) technologies has become an essential part of research projects and their role as potential molecular biomarkers is being investigated by the scientific community. The circulating miRNAs detection as a diagnostic or prognostic tool for the diagnosis and treatment of several diseases is very useful, because with a minimal quantity of sample (peripheral blood), miRNAs are very sensitive. Even more, miRNAs remain stable both at room temperature and during freeze-thaw cycles. These characteristics highlight the important role of miRNAs in the near future as new tools for anti-doping. The article provides a systematic review and meta-analysis on the role of miRNAs as new potential molecular biomarkers of AAS use/abuse. Particularly, this paper analyzed the “miRNA signature” use as biomarkers for health disorders, focusing on the organ damages which are related to ASS use/abuse. Moreover, this review aims to provide a future prospect for less invasive or non-invasive procedures for the detection of circulating miRNA biomarkers as doping assumption signaling.
Collapse
Affiliation(s)
- Francesco Sessa
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Monica Salerno
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giulio Di Mizio
- Department of Legal, Historical, Economic and Social Sciences, University of Catanzaro, Catanzaro, Italy
| | - Giuseppe Bertozzi
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Benedetta Tomaiuolo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Daniela Pisanelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Francesca Maglietta
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Pietrantonio Ricci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.,Medical and Surgical Sciences, University of Catanzaro, Catanzaro, Italy
| | - Cristoforo Pomara
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| |
Collapse
|
118
|
Mikovic J, Sadler K, Butchart L, Voisin S, Gerlinger-Romero F, Della Gatta P, Grounds MD, Lamon S. MicroRNA and Long Non-coding RNA Regulation in Skeletal Muscle From Growth to Old Age Shows Striking Dysregulation of the Callipyge Locus. Front Genet 2018; 9:548. [PMID: 30505320 PMCID: PMC6250799 DOI: 10.3389/fgene.2018.00548] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) undergo high levels of regulation in skeletal muscle development and control skeletal muscle mass, function and metabolism over the lifespan. More recently, the role of long non-coding RNAs (lncRNAs) in skeletal muscle regulation has started to emerge. Following up on our recent study describing the expression pattern and putative roles of 768 miRNAs in the quadriceps muscle of mice at early life stages, we used a high-throughput miRNA qPCR-based array to assess the expression of the same miRNAs in 28-month old male mouse quadriceps muscle. In addition, we report the expression patterns of lncRNAs playing a putative role in muscle development and adaptation from growth to old age. Twelve miRNAs were significantly downregulated in 28-month old muscle when compared with 12-week old muscle. Ten of them clustered at the Dlk1-Dio3 locus, known as ‘Callipyge,’ which is associated with muscle development and hypertrophy. This collective downregulation was paralleled by decreases in the expression levels of the maternally expressed imprinted LncRNA coding genes Meg3 and Rian stemming from the same chromosomal region. In contrast, the paternally expressed imprinted Dlk1-Dio3 locus members Rtl1, Dio3, and Dlk1 and the muscle related lncRNAs lncMyoD1, Neat_v1, Neat_v2, and Malat1 underwent significant changes during growth, but their expression levels were not altered past the age of 12 weeks, suggesting roles limited to hyperplasia and early hypertrophy. In conclusion, collective muscle miRNA expression gradually decreases over the lifespan and a cluster of miRNAs and maternally expressed lncRNAs stemming from the Callipyge locus is significantly dysregulated in aging muscle. The Dlk1-Dio3 locus therefore represents a potential new mechanism for age-related muscle decline.
Collapse
Affiliation(s)
- Jasmine Mikovic
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin University, Geelong, VIC, Australia
| | - Kate Sadler
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin University, Geelong, VIC, Australia
| | - Lauren Butchart
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Sarah Voisin
- Institute of Health and Sport, Victoria University, Footscray, VIC, Australia
| | - Frederico Gerlinger-Romero
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin University, Geelong, VIC, Australia
| | - Paul Della Gatta
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin University, Geelong, VIC, Australia
| | - Miranda D Grounds
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Séverine Lamon
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
119
|
The Role of MicroRNAs in Patients with Amyotrophic Lateral Sclerosis. J Mol Neurosci 2018; 66:617-628. [PMID: 30415446 DOI: 10.1007/s12031-018-1204-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a serious neurodegenerative disease that affects motor neurons and leads to death within 2 to 3 years after the first symptoms manifest. MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression in fundamental cellular processes and, post-transcriptionally, the translation levels of target mRNA transcripts. We searched PubMed for studies that examined miRNAs in ALS patients and attempted to group the results in order to find the strongest miRNA candidate for servings as an ALS biomarker. The studies on humans so far have been diverse, yielding considerably heterogeneous results, as they were performed on a wide variety of tissues and subjects. Among the miRNAs that were found consistently deregulated are miR-206, miR-133, miR-149, and miR-338-3p. Additively, the deregulation of some specific miRNAs seems to compose a miRNA expression profile that is specific for ALS. More research is required in order for the scientific community to reach a consensus.
Collapse
|
120
|
Li S, Chen W, Zhan A, Liang J. Identification and characterization of microRNAs involved in scale biomineralization in the naked carp Gymnocypris przewalskii. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2018; 28:196-203. [PMID: 30317123 DOI: 10.1016/j.cbd.2018.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 11/25/2022]
Abstract
The mineralized scale derived from skin plays a protective role for the fish body and also possesses important application values in the biomaterial field. However, little is known about fish scale biomineralization and related molecular regulatory mechanisms. Here, we used a comparative microRNA sequencing approach to identify and characterize differentially expressed microRNAs (DEMs) involved in scale biomineralization in the naked carp Gymnocypris przewalskii. A total of 18, 43, and 66 DEMs were obtained from skin tissues covered with initial, developing, and mature scales (IS, DS, and MS) compared with scale-uncovered skin. The target genes of these DEMs were significantly enriched in a sole biomineralization-related sphingolipid signaling pathway. Seven DEMs (dre-miR-124-3p, dre-miR-133a-2-5p, dre-miR-184, dre-miR-206-3p, novel_33, novel_56 and novel_75) were common in IS, DS, and MS. Dre-miR-124-3p, dre-miR-206-3p, and novel_33 were predicted to be able to target biomineralization-related genes. Stem-loop real-time quantitative PCR further confirmed that the common DEMs had higher expression levels in scale-covered skin tissues than that in the gill, intestine, and brain, except for dre-miR-133a-2-5p. Our results suggest that these identified microRNAs may play a role in scale biomineralization in G. przewalskii, and the obtained microRNAs are expected to be candidates in understanding the molecular mechanism of scale biomineralization in fish species.
Collapse
Affiliation(s)
- Shiguo Li
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Weiwei Chen
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Aibin Zhan
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China.
| | - Jian Liang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China.
| |
Collapse
|
121
|
Wang J, Song C, Cao X, Li H, Cai H, Ma Y, Huang Y, Lan X, Lei C, Ma Y, Bai Y, Lin F, Chen H. MiR-208b regulates cell cycle and promotes skeletal muscle cell proliferation by targeting CDKN1A. J Cell Physiol 2018; 234:3720-3729. [PMID: 30317561 DOI: 10.1002/jcp.27146] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 07/05/2018] [Indexed: 01/18/2023]
Abstract
Skeletal muscle is the most abundant tissue in the body. The development of skeletal muscle cell is complex and affected by many factors. A sea of microRNAs (miRNAs) have been identified as critical regulators of myogenesis. MiR-208b, a muscle-specific miRNA, was reported to have a connection with fiber type determination. However, whether miR-208b has effect on proliferation of muscle cell was under ascertained. In our study, cyclin-dependent kinase inhibitor 1A (CDKN1A), which participates in cell cycle regulation, was predicted and then validated as one target gene of miR-208b. We found that overexpression of miR-208b increased the expression of cyclin D1, cyclin E1, and cyclin-dependent kinase 2 at the levels of messenger RNA and protein in cattle primary myoblasts in vivo and in vitro. Flow cytometry showed that forced expression of miR-208b increased the percentage of cells at the S phase and decreased the percentage of cells at the G0/G1 phase. These results indicated that miR-208b participates in the cell cycle regulation of cattle primary myoblast cells. 5-Ethynyl-20-deoxyuridine and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays showed that overexpression of miR-208b promoted the proliferation of cattle primary myoblasts. Therefore, we conclude that miR-208b participates in the cell cycle and proliferation regulation of cattle primary skeletal muscle cell through the posttranscriptional downregulation of CDKN1A.
Collapse
Affiliation(s)
- Jian Wang
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chengchuang Song
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiukai Cao
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hui Li
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hanfang Cai
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yilei Ma
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yongzhen Huang
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xianyong Lan
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuzhao Lei
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yun Ma
- College of Life Sciences, Institute for Conservation and Utilization of Agro-Bioresources in Dabie Mountains, Xinyang Normal University, Xinyang, China
| | - Yueyu Bai
- Animal Health Supervision in Henan Province, Zhengzhou, China
| | - Fengpeng Lin
- Bureau of Animal Husbandry of Biyang County, Biyang, China
| | - Hong Chen
- Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
122
|
Kemp GJ, Birrell F, Clegg PD, Cuthbertson DJ, De Vito G, van Dieën JH, Del Din S, Eastell R, Garnero P, Goljanek–Whysall K, Hackl M, Hodgson R, Jackson MJ, Lord S, Mazzà C, McArdle A, McCloskey EV, Narici M, Peffers MJ, Schiaffino S, Mathers JC. Developing a toolkit for the assessment and monitoring of musculoskeletal ageing. Age Ageing 2018; 47:iv1-iv19. [PMID: 30203052 PMCID: PMC6127513 DOI: 10.1093/ageing/afy143] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
The complexities and heterogeneity of the ageing process have slowed the development of consensus on appropriate biomarkers of healthy ageing. The Medical Research Council–Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA) is a collaboration between researchers and clinicians at the Universities of Liverpool, Sheffield and Newcastle. One of CIMA’s objectives is to ‘Identify and share optimal techniques and approaches to monitor age-related changes in all musculoskeletal tissues, and to provide an integrated assessment of musculoskeletal function’—in other words to develop a toolkit for assessing musculoskeletal ageing. This toolkit is envisaged as an instrument that can be used to characterise and quantify musculoskeletal function during ‘normal’ ageing, lend itself to use in large-scale, internationally important cohorts, and provide a set of biomarker outcome measures for epidemiological and intervention studies designed to enhance healthy musculoskeletal ageing. Such potential biomarkers include: biochemical measurements in biofluids or tissue samples, in vivo measurements of body composition, imaging of structural and physical properties, and functional tests. This review assesses candidate biomarkers of musculoskeletal ageing under these four headings, details their biological bases, strengths and limitations, and makes practical recommendations for their use. In addition, we identify gaps in the evidence base and priorities for further research on biomarkers of musculoskeletal ageing.
Collapse
Affiliation(s)
- Graham J Kemp
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Fraser Birrell
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Peter D Clegg
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Daniel J Cuthbertson
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Giuseppe De Vito
- School of Public Health, Physiotherapy and Sports Science, Institute for Sport and Health, University College Dublin, Belfield, Dublin, Ireland
| | - Jaap H van Dieën
- Department of Human Movement Sciences, VU University Amsterdam, Amsterdam Movement Sciences, Van der Boechorststraat 9, Amsterdam, The Netherlands
| | - Silvia Del Din
- Clinical Ageing Research Unit, Institute of Neuroscience/Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Richard Eastell
- Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Patrick Garnero
- Division of Bone Diseases, Geneva University Hospital and Faculty of Medicine, 1205 Geneva, Switzerland
| | - Katarzyna Goljanek–Whysall
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | | | - Richard Hodgson
- Centre for Imaging Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, UK
| | - Malcolm J Jackson
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Sue Lord
- Clinical Ageing Research Unit, Institute of Neuroscience/Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Claudia Mazzà
- Department of Mechanical Engineering & INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Anne McArdle
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Eugene V McCloskey
- Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Marco Narici
- MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, Derby Royal Hospital, Uttoxeter Road, Derby, UK
| | - Mandy J Peffers
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease (IACD), University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| | - Stefano Schiaffino
- Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, Padova, Italy
| | - John C Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine and Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
- The MRC-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA)
| |
Collapse
|
123
|
Li Y, Liang Y, Zhu Y, Zhang Y, Bei Y. Noncoding RNAs in Cardiac Hypertrophy. J Cardiovasc Transl Res 2018; 11:439-449. [PMID: 30171598 DOI: 10.1007/s12265-018-9797-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/19/2018] [Indexed: 01/07/2023]
Abstract
Cardiac hypertrophy is classified as pathological and physiological hypertrophy. Pathological hypertrophy typically precedes the onset of heart failure, one of the largest contributors to disease burden and deaths worldwide. In contrast, physiological hypertrophy is an adaptive response and protects against adverse cardiac remodeling. Noncoding RNAs (ncRNAs) have drawn significant attention over the last couple of decades, and their dysregulation is increasingly being linked to cardiac hypertrophy and cardiovascular diseases. In this review, we will summarize the profiling, function, and molecular mechanism of microRNAs, long noncoding RNAs, and circular RNAs in pathological cardiac hypertrophy. Additionally, we also review microRNAs responsible for physiological hypertrophy. With better understanding of ncRNAs in cardiac hypertrophy, manipulation of the important ncRNAs will offer exciting avenues for the prevention and therapy of heart failure.
Collapse
Affiliation(s)
- Yongqin Li
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Yajun Liang
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Yujiao Zhu
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Yuhui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Bei Li Tu Road, Beijing, 100037, China.
| | - Yihua Bei
- Cardiac Regeneration and Ageing Lab, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.
| |
Collapse
|
124
|
Lovett JAC, Durcan PJ, Myburgh KH. Investigation of Circulating Extracellular Vesicle MicroRNA Following Two Consecutive Bouts of Muscle-Damaging Exercise. Front Physiol 2018; 9:1149. [PMID: 30177888 PMCID: PMC6109634 DOI: 10.3389/fphys.2018.01149] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022] Open
Abstract
Background: Extracellular vesicles (EVs) are nano-sized vesicles that are known to be powerful mediators of intercellular communication via their microRNA (miR) content. A paucity of information on EV-mediated communication arising from skeletal muscle (SkM) in response to exercise-induced muscle damage is present in the published literature. Lack of such information inhibits our understanding of muscle injury and repair processes. Aims: To assess circulating EV levels and selected miR content within them, in response to two consecutive bouts of muscle-damaging exercise. Methods: Serum creatine kinase activity (CK) and EVs were analyzed from the blood of 9 healthy, untrained males at baseline, and at 2 and 24 h post-exercise. The exercise regimen consisted of a combination of plyometric jumping and downhill running. Perceived muscle pain (PMP) was assessed on a scale from 1 to 10. Plasma EVs were isolated using size exclusion columns and visualized with transmission electron microscopy (TEM). EV size and number were quantified using nanoparticle tracking analysis (NTA). miR expression was quantified using qPCR, with normalization to an exogenous control (cel-miR-39). Results: PMP and CK were significantly elevated post-exercise compared to baseline levels, providing indirect evidence for muscle damage. EV visualization using TEM revealed an abundant and heterogeneously sized pool of intact particles within the exosome size range (30-150 nm). No significant change in mean EV size or number was seen over time. The SkM-specific miR-206 in EVs was found to be variable among participants and no significant change occurred in SkM-important miRs; 1, 133a, 133b, 486, and 499a. However, EV miR-31 decreased from baseline to 24 h post-exercise (p = 0.027). Conclusion: Mild to moderate exercise-induced muscle damage altered the miR-31 profile of circulating EVs within the first 24 h post-exercise, but not that of myomiRs in EVs. These data demonstrate that EVs carry selectively packaged cargo which can be affected by exercise. Future research into the total miR content of EVs in response to exercise-induced muscle damage may reveal other miRs responsive to this relatively mild perturbation. More time points post-muscle-damaging exercise would provide a better understanding of the temporal EV myomiR response.
Collapse
Affiliation(s)
- Jason A C Lovett
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Peter J Durcan
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Kathryn H Myburgh
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
125
|
Ren D, Zheng H, Fei S, Zhao JL. MALAT1 induces osteosarcoma progression by targeting miR-206/CDK9 axis. J Cell Physiol 2018; 234:950-957. [PMID: 30076726 DOI: 10.1002/jcp.26923] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 06/13/2018] [Indexed: 12/29/2022]
Abstract
Long noncoding RNAs (LncRNAs) have been reported to participate in cancer development, including osteosarcoma. Here, in our study, we observed that lncRNA metastasis-associated lung adenocarcinoma transcription 1 (MALAT1) was remarkably overexpressed in osteosarcoma. However, the role it plays in osteosarcoma proliferation mediated by miR-206/cyclin-dependent kinase 9 (CDK9) axis remains uninvestigated. It was found that miR-206 was decreased and CDK9 was elevated in human osteosarcoma cells including MG63, Saos-2, U2OS, and KHOS compared with human osteoblast cell line hFOB 1.19. In addition, it was exhibited that knockdown of MALAT1 was able to inhibit osteosarcoma cell proliferation, which suggested that MALAT1 played an oncogenic role in osteosarcoma development. Bioinformatics analysis indicated that MALAT1 can function as a competing endogenous RNA by sponging miR-206. Because miR-206 has been identified as a significant tumor suppressive gene in multiple cancers, we validated that mimics of miR-206 can restrain osteosarcoma progression. Furthermore, dual-luciferase reporter assay, RNA binding protein immunoprecipitation, and RNA pull-down assay demonstrated the correlation between miR-206 and MALAT1. Besides these, CDK9 was predicted as a downstream gene of miR-206, and we observed that MALAT1 can regulate osteosarcoma progress by modulating CDK9 expression via sponging miR-206. In conclusion, our study implied that MALAT1/miR-206/CDK9 axis can provide novel insights into the biological mechanism of osteosarcoma progression.
Collapse
Affiliation(s)
- Dong Ren
- Department of Hand Surgery, Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hao Zheng
- Department of Radiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sang Fei
- Department of Orthopedics, Lian Shui People's Hospital, Lianshui, Jiangsu, China
| | - Jia-Li Zhao
- Department of Orthopaedics, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| |
Collapse
|
126
|
McClure MJ, Cohen DJ, Ramey AN, Bivens CB, Mallu S, Isaacs JE, Imming E, Huang YC, Sunwoo M, Schwartz Z, Boyan BD. Decellularized Muscle Supports New Muscle Fibers and Improves Function Following Volumetric Injury. Tissue Eng Part A 2018; 24:1228-1241. [DOI: 10.1089/ten.tea.2017.0386] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Michael J. McClure
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - David J. Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Allison N. Ramey
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Caroline B. Bivens
- Department of School of Art, Virginia Commonwealth University, Richmond, Virginia
| | - Satya Mallu
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Jonathan E. Isaacs
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Emily Imming
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - Yen-Chen Huang
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - MoonHae Sunwoo
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
127
|
Liu B, Cong W, Liu C, Tang Y, Zhou N, Li N, Zhang Y, Jin Y, Xiao J. miR-27b-3p Was Involved in Retinoic Acid-induced Abnormal Early Myogenic Differentiation of C2C12 Cells via Targeting CaMKIIδ. J HARD TISSUE BIOL 2018. [DOI: 10.2485/jhtb.27.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Bo Liu
- Institute of Genome Engineered Animal Models for Human Diseases, Dalian Medical University
| | - Wei Cong
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Chao Liu
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Yi Tang
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Nan Zhou
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Nan Li
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Ying Zhang
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Yaru Jin
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| | - Jing Xiao
- Department of Oral Pathology, College of Stomatology, Dalian Medical University
| |
Collapse
|
128
|
Assis AF, Li J, Donate PB, Dernowsek JA, Manley NR, Passos GA. Predicted miRNA-mRNA-mediated posttranscriptional control associated with differences in cervical and thoracic thymus function. Mol Immunol 2018; 99:39-52. [DOI: 10.1016/j.molimm.2018.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 03/09/2018] [Accepted: 04/05/2018] [Indexed: 12/12/2022]
|
129
|
Mattiotti A, Prakash S, Barnett P, van den Hoff MJB. Follistatin-like 1 in development and human diseases. Cell Mol Life Sci 2018; 75:2339-2354. [PMID: 29594389 PMCID: PMC5986856 DOI: 10.1007/s00018-018-2805-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/27/2018] [Accepted: 03/22/2018] [Indexed: 12/19/2022]
Abstract
Follistatin-like 1 (FSTL1) is a secreted glycoprotein displaying expression changes during development and disease, among which cardiovascular disease, cancer, and arthritis. The cardioprotective role of FSTL1 has been intensively studied over the last years, though its mechanism of action remains elusive. FSTL1 is involved in multiple signaling pathways and biological processes, including vascularization and regulation of the immune response, a feature that complicates its study. Binding to the DIP2A, TLR4 and BMP receptors have been shown, but other molecular partners probably exist. During cancer progression and rheumatoid arthritis, controversial data have been reported with respect to the proliferative, apoptotic, migratory, and inflammatory effects of FSTL1. This controversy might reside in the extensive post-transcriptional regulation of FSTL1. The FSTL1 primary transcript also encodes for a microRNA (miR-198) in primates and multiple microRNA-binding sites are present in the 3'UTR. The switch between expression of the FSTL1 protein and miR-198 is an important regulator of tumour metastasis and wound healing. The glycosylation state of FSTL1 is a determinant of biological activity, in cardiomyocytes the glycosylated form promoting proliferation and the non-glycosylated working anti-apoptotic. Moreover, the glycosylation state shows differences between species and tissues which might underlie the differences observed in in vitro studies. Finally, regulation at the level of protein secretion has been described.
Collapse
Affiliation(s)
- Andrea Mattiotti
- Department of Medical Biology, Academic Medical Center, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - Stuti Prakash
- Department of Medical Biology, Academic Medical Center, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - Phil Barnett
- Department of Medical Biology, Academic Medical Center, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - Maurice J B van den Hoff
- Department of Medical Biology, Academic Medical Center, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
130
|
mir-127-3p inhibits the proliferation of myocytes by targeting KMT5a. Biochem Biophys Res Commun 2018; 503:970-976. [PMID: 29932923 DOI: 10.1016/j.bbrc.2018.06.104] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 06/19/2018] [Indexed: 01/06/2023]
Abstract
MicroRNAs are a class of highly conserved ∼20 nucleotides non-coding RNAs that post-transcriptionally regulate gene expression. Many miRNAs were studied in the development of skeletal muscle, such as miR-1, miR-206, and miR-133. In our previous study, miR-127-3p was found highly expressed in porcine fetal skeletal muscle, whereas the detailed functions of miR-127-3p in muscle development is still unclear. In this study, we detected that miR-127-3p also highly expressed in skeletal muscle, cardiac muscle of adult mice and proliferative C2C12 cell lines. Overexpression of miR-127-3p almost has no effects on differentiation of C2C12 cell lines. However, miR-127-3p significantly inhibited the cell proliferation of C2C12 cells. Moreover, we identified KMT5a as a target gene that was down-regulated in both mRNA and protein level when miR-127-3p mimics were introduced. Furthermore, KMT5a overexpression in miR-127-3p treated cells rescued the influence of miR-127-3p on C2C12 proliferation. In brief, our data reveals that miR-127-3p regulates the proliferation of myocytes through KMT5a.
Collapse
|
131
|
Watt K, Newsted D, Voorand E, Gooding RJ, Majewski A, Truesdell P, Yao B, Tuschl T, Renwick N, Craig AW. MicroRNA-206 suppresses TGF-β signalling to limit tumor growth and metastasis in lung adenocarcinoma. Cell Signal 2018; 50:25-36. [PMID: 29935234 DOI: 10.1016/j.cellsig.2018.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/18/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022]
Abstract
MicroRNA-206 (miR-206) has demonstrated tumor suppressive effects in a variety of cancers. Numerous studies have identified aberrantly expressed targets of miR-206 that contribute to tumor progression and metastasis, however, the broader gene-networks and pathways regulated by miR-206 remain poorly defined. Here, we have ectopically expressed miR-206 in lung adenocarcinoma cell lines and tumors to identify differentially expressed genes, and study the effects on tumor growth and metastasis. In H1299 tumor xenograft assays, stable expression of miR-206 suppressed both tumor growth and metastasis in mice. Profiling of xenograft tumors using small RNA sequencing and a targeted panel of tumor progression and metastasis-related genes revealed a network of genes involved in TGF-β signalling that were regulated by miR-206. Among these were the TGFB1 ligand, as well as direct transcriptional targets of Smad3. Other differentially expressed genes included components of the extracellular matrix involved in TGF-β activation and signalling, including Thrombospondin-1, which is responsible for the activation of latent TGF-β in the stroma. In cultured lung adenocarcinoma cells treated with recombinant TGF-β, ectopic expression of miR-206 impaired canonical signalling, and expression of TGF-β target genes linked to epithelial-mesenchymal transition. This was due at least in part to the suppression of Smad3 protein levels in lung adenocarcinoma cells with ectopic miR-206 expression. Together, these findings indicate that miR-206 can suppress tumor progression and metastasis by limiting autocrine production of TGF-β, and highlight the potential utility of TGF-β inhibitors for the treatment of lung adenocarcinomas.
Collapse
Affiliation(s)
- Kathleen Watt
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Daniel Newsted
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Elena Voorand
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Robert J Gooding
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Physics, Queen's University, Kingston, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Adrianna Majewski
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Peter Truesdell
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Binchen Yao
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Thomas Tuschl
- HHMI Laboratory of RNA Molecular Biology, The Rockefeller University, New York, USA
| | - Neil Renwick
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada; HHMI Laboratory of RNA Molecular Biology, The Rockefeller University, New York, USA
| | - Andrew W Craig
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada.
| |
Collapse
|
132
|
Vergara HM, Ramirez J, Rosing T, Nave C, Blandino R, Saw D, Saraf P, Piexoto G, Coombes C, Adams M, Domingo CR. miR-206 is required for changes in cell adhesion that drive muscle cell morphogenesis in Xenopus laevis. Dev Biol 2018; 438:94-110. [PMID: 29596841 DOI: 10.1016/j.ydbio.2018.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/14/2018] [Accepted: 03/22/2018] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are highly conserved small non-coding RNA molecules that post-transcriptionally regulate gene expression in multicellular organisms. Within the set of muscle-specific miRNAs, miR-206 expression is largely restricted to skeletal muscle and is found exclusively within the bony fish lineage. Although many studies have implicated miR-206 in muscle maintenance and disease, its role in skeletal muscle development remains largely unknown. Here, we examine the role of miR-206 during Xenopus laevis somitogenesis. In Xenopus laevis, miR-206 expression coincides with the onset of somitogenesis. We show that both knockdown and over-expression of miR-206 result in abnormal somite formation affecting muscle cell rotation, attachment, and elongation. In particular, our data suggests that miR-206 regulates changes in cell adhesion that affect the ability of newly formed somites to adhere to the notochord as well as to the intersomitic boundaries. Additionally, we show that β-dystroglycan and F-actin expression levels are significantly reduced, suggesting that knockdown of miR-206 levels affects cellular mechanics necessary for cell shape changes and attachments that are required for proper muscle formation.
Collapse
Affiliation(s)
- Hernando Martínez Vergara
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Julio Ramirez
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Trista Rosing
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Ceazar Nave
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Rebecca Blandino
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Daniel Saw
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Parag Saraf
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Gabriel Piexoto
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Coohleen Coombes
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Melissa Adams
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA
| | - Carmen R Domingo
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA, 94132, USA.
| |
Collapse
|
133
|
Umeh-Garcia M, Sweeney C. Cancer prevention through miRNAs: miR-206 prevents the initiation and progression of hepatocellular carcinoma by attenuating c-MET signaling and cell-cycle progression via cyclin D1 and CDK6. ACTA ACUST UNITED AC 2018; 2. [PMID: 31930188 DOI: 10.21037/ncri.2018.06.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Maxine Umeh-Garcia
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| | - Colleen Sweeney
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
134
|
Di Pietro L, Lattanzi W, Bernardini C. Skeletal Muscle MicroRNAs as Key Players in the Pathogenesis of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2018; 19:ijms19051534. [PMID: 29786645 PMCID: PMC5983603 DOI: 10.3390/ijms19051534] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder, for which, to date, no effective treatment to ameliorate the clinical manifestations is available. The long-standing view of ALS as affecting only motor neurons has been challenged by the finding that the skeletal muscle plays an active role in the disease pathogenesis and can be a valuable target for therapeutic strategies. In recent years, non-coding RNAs, including microRNAs, have emerged as important molecules that play key roles in several cellular mechanisms involved in the pathogenic mechanisms underlying various human conditions. In this review, we summarize how the expression of some microRNAs is dysregulated in the skeletal muscle of ALS mouse models and patients. Shedding light on the mechanisms underlying microRNAs dysregulation in the skeletal muscle could clarify some of the processes involved in the pathogenesis of ALS and especially identify new promising therapeutic targets in patients.
Collapse
Affiliation(s)
- Lorena Di Pietro
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Wanda Lattanzi
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Camilla Bernardini
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
135
|
miR-133b, a particular member of myomiRs, coming into playing its unique pathological role in human cancer. Oncotarget 2018; 8:50193-50208. [PMID: 28422730 PMCID: PMC5564843 DOI: 10.18632/oncotarget.16745] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs, a family of single-stranded and non-coding RNAs, play a crucial role in regulating gene expression at posttranscriptional level, by which it can mediate various types of physiological and pathological process in normal developmental progress and human disease, including cancer. The microRNA-133b originally defined as canonical muscle-specific microRNAs considering their function to the development and health of mammalian skeletal and cardiac muscles, but new findings coming from our group and others revealed that miR-133b have frequently abnormal expression in various kinds of human cancer and its complex complicated regulatory networks affects the tumorigenicity and development of malignant tumors. Very few existing reviews on miR-133b, until now, are principally about its role in homologous cluster (miR-1, −133 and -206s), however, most of constantly emerging new researches now are focused mainly on one of them, so In this article, to highlight the unique pathological role of miR-133b playing in tumor, we conduct a review to summarize the current understanding about one of the muscle-specific microRNAs, namely miR-133b, acting in human cancer. The review focused on the following four aspects: the overview of miR-133b, the target genes of miR-133b involved in human cancer, the expression of miR-133b and regulatory mechanisms leading to abnormal expression of miR-133b.
Collapse
|
136
|
Chen S, Fan X, Gu H, Zhang L, Zhao W. Competing endogenous RNA regulatory network in papillary thyroid carcinoma. Mol Med Rep 2018; 18:695-704. [PMID: 29767230 PMCID: PMC6059698 DOI: 10.3892/mmr.2018.9009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 11/15/2017] [Indexed: 11/16/2022] Open
Abstract
The present study aimed to screen all types of RNAs involved in the development of papillary thyroid carcinoma (PTC). RNA-sequencing data of PTC and normal samples were used for screening differentially expressed (DE) microRNAs (DE-miRNAs), long non-coding RNAs (DE-lncRNAs) and genes (DEGs). Subsequently, lncRNA-miRNA, miRNA-gene (that is, miRNA-mRNA) and gene-gene interaction pairs were extracted and used to construct regulatory networks. Feature genes in the miRNA-mRNA network were identified by topological analysis and recursive feature elimination analysis. A support vector machine (SVM) classifier was built using 15 feature genes, and its classification effect was validated using two microarray data sets that were downloaded from the Gene Expression Omnibus (GEO) database. In addition, Gene Ontology function and Kyoto Encyclopedia Genes and Genomes pathway enrichment analyses were conducted for genes identified in the ceRNA network. A total of 506 samples, including 447 tumor samples and 59 normal samples, were obtained from The Cancer Genome Atlas (TCGA); 16 DE-lncRNAs, 917 DEGs and 30 DE-miRNAs were screened. The miRNA-mRNA regulatory network comprised 353 nodes and 577 interactions. From these data, 15 feature genes with high predictive precision (>95%) were extracted from the network and were used to form an SVM classifier with an accuracy of 96.05% (486/506) for PTC samples downloaded from TCGA, and accuracies of 96.81 and 98.46% for GEO downloaded data sets. The ceRNA regulatory network comprised 596 lines (or interactions) and 365 nodes. Genes in the ceRNA network were significantly enriched in ‘neuron development’, ‘differentiation’, ‘neuroactive ligand-receptor interaction’, ‘metabolism of xenobiotics by cytochrome P450’, ‘drug metabolism’ and ‘cytokine-cytokine receptor interaction’ pathways. Hox transcript antisense RNA, miRNA-206 and kallikrein-related peptidase 10 were nodes in the ceRNA regulatory network of the selected feature gene, and they may serve import roles in the development of PTC.
Collapse
Affiliation(s)
- Shouhua Chen
- Department of Breast and Thyroid Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Xiaobin Fan
- Department of Operation Room, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - He Gu
- Department of Breast and Thyroid Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Lili Zhang
- Department of Breast and Thyroid Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Wenhua Zhao
- Department of Oncology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
137
|
Wang Y, Xu H, Si L, Li Q, Zhu X, Yu T, Gang X. MiR-206 inhibits proliferation and migration of prostate cancer cells by targeting CXCL11. Prostate 2018. [PMID: 29542173 DOI: 10.1002/pros.23468] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Our study was aimed at detecting the expression levels of miR-206 in prostate cancer (PCa) tissues and PCa cell lines, and exploring the potential functions of miR-206 by targeting chemokine ligand 11 (CXCL11). METHODS RT-qPCR was applied to detect the expressions of miR-206 and CXCL11 in PCa tissues and in PCa cell lines. Expression of the CXCL11 protein was detected using Western blot. After manipulating the expression of miR-206 and CXCL11 in PC-3 and DU-145 cells, the changes of cell proliferation and cell cycle were observed through cell counting kit-8 (CCK-8) and flow cytometry. Wound healing and transwell assay were conducted for cell migration and invasion examination in vitro. The luciferase reporter assay was applied to validate the association between miR-206 and CXCL11. RESULTS MiR-206 was significantly under-expressed in PCa tissues and in PCa cell lines. Up-regulation of miR-206 could inhibit proliferation, migration, invasion and induced G1/G0 arrest of PCa cells, and vice versa. MiR-206 bound to the 3'-UTR of CXCL11 and significantly repressed the luciferase activity. Overexpression of miR-206 decreased the expression level of CXCL11 significantly. CXCL11 mRNA and protein levels were significantly decreased in PCa cells. Downregulation of CXCL11 presented tumor-suppressing effects on PCa cells as miR-206 mimics did. And co-transfection miR-206 attenuated the tumor-promoting effects induced by CXCL11 overexpression. CONCLUSION Our current finding demonstrated that miR-206 negatively regulated PCa cell proliferation and migration, and arrested cell cycle by targeting CXCL11 as a tumor suppressor in prostate cancer.
Collapse
Affiliation(s)
- Yao Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Haitao Xu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lihui Si
- Department of Gynaecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Qiuju Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xiujie Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Tong Yu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
138
|
Jiao D, Chen J, Li Y, Tang X, Wang J, Xu W, Song J, Li Y, Tao H, Chen Q. miR-1-3p and miR-206 sensitizes HGF-induced gefitinib-resistant human lung cancer cells through inhibition of c-Met signalling and EMT. J Cell Mol Med 2018; 22:3526-3536. [PMID: 29664235 PMCID: PMC6010770 DOI: 10.1111/jcmm.13629] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 03/08/2018] [Indexed: 12/20/2022] Open
Abstract
Hepatocyte growth factor (HGF) overexpression is an important mechanism in acquired epidermal growth factor receptor (EGFR) kinase inhibitor gefitinib resistance in lung cancers with EGFR activating mutations. MiR-1-3p and miR-206 act as suppressors in lung cancer proliferation and metastasis. However, whether miR-1-3p and miR-206 can overcome HGF-induced gefitinib resistance in EGFR mutant lung cancer is not clear. In this study, we showed that miR-1-3p and miR-206 restored the sensitivities of lung cancer cells PC-9 and HCC-827 to gefitinib in present of HGF. For the mechanisms, we demonstrated that both miR-1-3p and miR-206 directly target HGF receptor c-Met in lung cancer. Knockdown of c-Met mimicked the effects of miR-1-3p and miR-206 transfections Meanwhile, c-Met overexpression attenuated the effects of miR-1-3p and miR-206 in HGF-induced gefitinib resistance of lung cancers. Furthermore, we showed that miR-1-3p and miR-206 inhibited c-Met downstream Akt and Erk pathway and blocked HGF-induced epithelial-mesenchymal transition (EMT). Finally, we demonstrated that miR-1-3p and miR-206 can increase gefitinib sensitivity in xenograft mouse models in vivo. Our study for the first time indicated the new function of miR-1-3p and miR-206 in overcoming HGF-induced gefitinib resistance in EGFR mutant lung cancer cell.
Collapse
Affiliation(s)
- Demin Jiao
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Jun Chen
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Yu Li
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Xiali Tang
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Jian Wang
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Wei Xu
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Jia Song
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - You Li
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Huimin Tao
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China
| | - Qingyong Chen
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, China.,The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
139
|
Lin CY, He JY, Zeng CW, Loo MR, Chang WY, Zhang PH, Tsai HJ. microRNA-206 modulates an Rtn4a/Cxcr4a/Thbs3a axis in newly forming somites to maintain and stabilize the somite boundary formation of zebrafish embryos. Open Biol 2018; 7:rsob.170009. [PMID: 28701377 PMCID: PMC5541343 DOI: 10.1098/rsob.170009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
Although microRNA-206 (miR-206) is known to regulate proliferation and differentiation of muscle fibroblasts, the role of miR-206 in early-stage somite development is still unknown. During somitogenesis of zebrafish embryos, reticulon4a (rtn4a) is specifically repressed by miR-206. The somite boundary was defective, and actin filaments were crossing over the boundary in either miR-206-knockdown or rtn4a-overexpressed embryos. In these treated embryos, C-X-C motif chemokine receptor 4a (cxcr4a) was reduced, while thrombospondin 3a (thbs3a) was increased. The defective boundary was phenocopied in either cxcr4a-knockdown or thbs3a-overexpressed embryos. Repression of thbs3a expression by cxcr4a reduced the occurrence of the boundary defect. We demonstrated that cxcr4a is an upstream regulator of thbs3a and that defective boundary cells could not process epithelialization in the absence of intracellular accumulation of the phosphorylated focal adhesion kinase (p-FAK) in boundary cells. Therefore, in the newly forming somites, miR-206-mediated downregulation of rtn4a increases cxcr4a. This activity largely decreases thbs3a expression in the epithelial cells of the somite boundary, which causes epithelialization of boundary cells through mesenchymal-epithelial transition (MET) and eventually leads to somite boundary formation. Collectively, we suggest that miR-206 mediates a novel pathway, the Rtn4a/Cxcr4a/Thbs3a axis, that allows boundary cells to undergo MET and form somite boundaries in the newly forming somites of zebrafish embryos.
Collapse
Affiliation(s)
- Cheng-Yung Lin
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| | - Jun-Yu He
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Chih-Wei Zeng
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Moo-Rumg Loo
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Wen-Yen Chang
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan, Republic of China
| | - Po-Hsiang Zhang
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| | - Huai-Jen Tsai
- Institute of Biomedical Sciences, Mackay Medical College, No. 46, Section 3 Zhongzhen Road, Sanzhi Dist., New Taipei City 252, Taiwan, Republic of China
| |
Collapse
|
140
|
Meral I, Pala M, Akbas F, Ustunova S, Yildiz C, Demirel MH. Effects of thymoquinone on liver miRNAs and oxidative stress in Ehrlich acid mouse solid tumor model. Biotech Histochem 2018; 93:301-308. [PMID: 29611713 DOI: 10.1080/10520295.2018.1437472] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We investigated the effects of thymoquinone (TQ) on the expression of liver microRNAs (miRNAs), liver histopathology and oxidative stress in Ehrlich acid solid tumor model induced mice. We used 24 male BALB/c mice divided randomly into three groups. Control (C) group mice were injected intraperitoneally (i.p.) with 0.5 ml saline for four weeks. Tumor (T) group mice were injected i.p. with 0.5 ml saline for four weeks, then Ehrlich acid tumor cells were injected subcutaneously into the neck to induce solid tumor formation. TQ (T + Tq) group mice injected i.p. with 10 mg/kg TQ for four weeks, then Ehrlich acid tumor cells were injected subcutaneously into the neck of the mice in this group to induce solid tumor formation. At the end of the study, liver from all groups were removed for histopathological and miRNAs analysis, and oxidative stress measurement. We found that the expression of miR-206b-3p was up-regulated and the oxidative stress and necrosis increased in the liver tissue of mice with Ehrlich acid solid tumor. TQ application decreased the oxidative stress, prevented necrosis, increased regeneration and down-regulated the expression of miR-206b-3p in the liver tissue.
Collapse
Affiliation(s)
- I Meral
- a Department of Physiology, School of Medicine , Bezmialem Vakif University , Istanbul
| | - M Pala
- b Department of Physiology, School of Medicine , Biruni University , Istanbul
| | - F Akbas
- c Department of Medical Biology, School of Medicine , Bezmialem Vakif University , Istanbul
| | - S Ustunova
- a Department of Physiology, School of Medicine , Bezmialem Vakif University , Istanbul
| | - C Yildiz
- d School of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| | - M H Demirel
- d School of Medicine , Bezmialem Vakif University , Istanbul , Turkey
| |
Collapse
|
141
|
Mitchell CJ, D'Souza RF, Schierding W, Zeng N, Ramzan F, O'Sullivan JM, Poppitt SD, Cameron-Smith D. Identification of human skeletal muscle miRNA related to strength by high-throughput sequencing. Physiol Genomics 2018; 50:416-424. [PMID: 29602299 DOI: 10.1152/physiolgenomics.00112.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The loss of muscle size, strength, and quality with aging is a major determinant of morbidity and mortality in the elderly. The regulatory pathways that impact the muscle phenotype include the translational regulation maintained by microRNAs (miRNA). Yet the miRNAs that are expressed in human skeletal muscle and relationship to muscle size, strength, and quality are unknown. Using next-generation sequencing, we selected the 50 most abundantly expressed miRNAs and then analyzed them in vastus lateralis muscle, obtained by biopsy from middle-aged males ( n = 48; 50.0 ± 4.3 yr). Isokinetic strength testing and midthigh computed tomography was undertaken for muscle phenotype analysis. Muscle attenuation was measured by computerized tomography and is inversely proportional to myofiber lipid content. miR-486-5p accounted for 21% of total miR sequence reads, with miR-10b-5p, miR-133a-3p, and miR-22-3p accounting for a further 15, 12, and 10%, respectively. Isokinetic knee extension strength and muscle cross-sectional area were positively correlated with miR-100-5p, miR-99b-5p, and miR-191-5p expression. Muscle attenuation was negatively correlated to let-7f-5p, miR-30d-5p, and miR-125b-5p expression. In silico analysis implicates miRNAs related to strength and muscle size in the regulation of mammalian target of rapamycin, while miRNAs related to muscle attenuation may have potential roles regulating the transforming growth factor-β/SMAD3 pathway.
Collapse
Affiliation(s)
| | | | | | - Nina Zeng
- Liggins Institute, University of Auckland , Auckland , New Zealand
| | - Farha Ramzan
- Liggins Institute, University of Auckland , Auckland , New Zealand
| | | | - Sally D Poppitt
- School of Biological Sciences, University of Auckland , Auckland , New Zealand.,Riddet Institute , Palmerston North , New Zealand
| | - David Cameron-Smith
- Liggins Institute, University of Auckland , Auckland , New Zealand.,Food & Bio-based Products Group, AgResearch, Palmerston North , New Zealand.,Riddet Institute , Palmerston North , New Zealand
| |
Collapse
|
142
|
Butchart LC, Terrill JR, Rossetti G, White R, Filipovska A, Grounds MD. Expression patterns of regulatory RNAs, including lncRNAs and tRNAs, during postnatal growth of normal and dystrophic (mdx) mouse muscles, and their response to taurine treatment. Int J Biochem Cell Biol 2018; 99:52-63. [PMID: 29578051 DOI: 10.1016/j.biocel.2018.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 01/27/2023]
Abstract
Post-natal skeletal muscle growth in mice is very rapid and involves complex changes in many cells types over the first 6 weeks of life. The acute onset of dystropathology also occurs around 3 weeks of age in the mdx mouse model of the human disease Duchenne Muscular Dystrophy (DMD). This study investigated (i) alterations in expression patterns of regulatory non-coding RNAs (ncRNAs) in vivo, including miRNAs, lncRNAs and tRNAs, during early growth of skeletal muscles in normal control C57Bl/10Scsn (C57) compared with dystrophic mdx mice from 2 to 6 weeks of postnatal age, and revealed inherent differences in vivo for levels of 3 ncRNAs between C57 and mdx muscles before the onset of dystropathology. Since the amino acid taurine has many benefits and reduces disease severity in mdx mice, this study also (ii) determined the impact of taurine treatment on these expression patterns in mdx muscles at the onset of dystropathology (3 weeks) and after several bouts of myonecrosis and regeneration (6 weeks). Taurine treatment of mdx mice only altered ncRNA levels when administered from 18 days to 6 weeks of age, but a deficiency in tRNA levels was rectified earlier in mdx skeletal muscles treated from 14 days to 3 weeks. Myogenesis in tissue culture was also used to (iii) compare ncRNA expression patterns for both strains, and (iv) the response to taurine treatment. These analyses revealed intrinsic differences in ncRNA expression patterns during myogenesis between strains, as well as increased sensitivity of mdx ncRNA levels to taurine treatment.
Collapse
Affiliation(s)
- Lauren C Butchart
- School of Human Sciences, The University of Western Australia, Australia.
| | - Jessica R Terrill
- School of Molecular Sciences, The University of Western Australia, Australia
| | - Giulia Rossetti
- Harry Perkins Institute of Medical Research, Western Australia, Australia
| | - Robert White
- School of Human Sciences, The University of Western Australia, Australia
| | - Aleksandra Filipovska
- School of Molecular Sciences, The University of Western Australia, Australia; Harry Perkins Institute of Medical Research, Western Australia, Australia
| | - Miranda D Grounds
- School of Human Sciences, The University of Western Australia, Australia
| |
Collapse
|
143
|
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression by targeting mRNAs for degradation or translational repression. MiRNAs can be expressed tissue specifically and are altered in response to various physiological conditions. It has recently been shown that miRNAs are released into the circulation, potentially for the purpose of communicating with distant tissues. This manuscript discusses miRNA alterations in cardiac muscle and the circulation during heart failure, a prevalent and costly public health issue. A potential mechanism for how skeletal muscle maladaptations during heart failure could be mediated by myocardium-derived miRNAs released to the circulation is presented. An overview of miRNA alterations in skeletal muscle during the ubiquitous process of aging and perspectives on miRNA interactions during heart failure are also provided.
Collapse
Affiliation(s)
- Kevin A Murach
- Center for Muscle Biology, University of Kentucky, Lexington, KY, 40536, USA
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, KY, 40536, USA
| | - John J McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, KY, 40536, USA.
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
144
|
Walter E, Dellago H, Grillari J, Dimai HP, Hackl M. Cost-utility analysis of fracture risk assessment using microRNAs compared with standard tools and no monitoring in the Austrian female population. Bone 2018; 108:44-54. [PMID: 29269173 DOI: 10.1016/j.bone.2017.12.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/14/2017] [Accepted: 12/16/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Osteoporosis poses an immense burden to the society in terms of morbidity, mortality and financial cost. To reduce this burden, it is essential to accurately assess the individual patient's fracture risk and, where indicated, to initiate appropriate treatment that reduces fracture probability. Current screening and monitoring approaches include utilization of FRAX®, a web-based country-specific fracture risk assessment tool, and bone mineral density measurement by Dual Energy X-ray Absorptiometry (DXA). Recently, microRNAs have been recognized as important regulators of bone physiology and potential biomarkers for fracture risk assessment and monitoring. A fracture risk assessment tool based on microRNAs (osteomiR™ test) is currently being developed. The aim of this study was to estimate the cost-effectiveness of fracture risk screening, monitoring, and resulting treatment decisions for the Austrian female population using the osteomiR™ test compared with DXA, with FRAX®, or with no screening/monitoring. METHODS A cost-utility-model was developed to simulate long-term consequences of Austrian women from age 50 over lifetime or death with respect to osteoporosis. Markov-modelling techniques were used to calculate health state transitions of fracture incidence according to risk groups (high, intermediate, low). High-risk patients receive medical treatment. Probabilities were derived via systematic-literature-review; direct costs (2015, €) from published sources from the payer's perspective. Results evaluate the incremental cost-effectiveness ratios (ICER) for osteomiR™ against the comparators, gains or losses of fractures, life years (LYs), quality-adjusted life years (QALYs), and direct costs. QALYs, life years (LYs) and costs were discounted (3% p.a). RESULTS Fracture risk assessment and monitoring using the osteomiR™ test reduces fracture incidence compared with no monitoring, DXA alone, or FRAX® alone. In the per-patient analysis, the ICER/QALY of osteomiR™ vs. no-monitoring was 13,103 €, vs. FRAX® 37,813 €, and vs. DXA -19,605 €, indicating that costs can be saved while gaining QALYs. Considering the total cohort over lifetime, the osteomiR™ test can avoid 57,919 fractures compared with DXA, 31,285 fractures compared with FRAX® and 133,394 fractures compared with no monitoring. Sensitivity analysis confirmed the robustness of these findings. CONCLUSION Fracture risk assessment and monitoring using the osteomiR™ test dominates DXA-strategy and constitutes a cost-effective alternative to FRAX®, and no-monitoring, respectively.
Collapse
Affiliation(s)
- Evelyn Walter
- Institute for Pharmaeconomic Research, Vienna, Austria
| | | | - Johannes Grillari
- University of Natural Resources and Life Sciences Vienna, Department of Biotechnology, Vienna, Austria
| | - Hans Peter Dimai
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
| | | |
Collapse
|
145
|
Muscle microRNA signatures as biomarkers of disease progression in amyotrophic lateral sclerosis. Neurobiol Dis 2018; 114:85-94. [PMID: 29486297 DOI: 10.1016/j.nbd.2018.02.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/28/2018] [Accepted: 02/21/2018] [Indexed: 02/06/2023] Open
Abstract
ALS is a fatal neurodegenerative disorder of motor neurons leading to progressive atrophy and weakness of muscles. Some of the earliest pathophysiological changes occur at the level of skeletal muscle and the neuromuscular junction. We previously identified distinct mRNA patterns, including members of the Smad and TGF-β family, that emerge in muscle tissue at the earliest (pre-clinical) stages. These patterns track disease progression in the mutant SOD1 mouse and are present in human ALS muscle. Because miRNAs play a direct regulatory role in mRNA expression, we hypothesized in this study that there would be distinct miRNA patterns in ALS muscle appearing in early stages that could track disease progression. We performed next-generation miRNA sequencing on muscle samples from G93A SOD1 mice at early (pre-clinical) and late (symptomatic) stages, and identified distinct miRNA patterns at both stages with some overlap. An Ingenuity Pathway Analysis predicted effects on a number of pathways relevant to ALS including TGF-β signaling, axon guidance signaling, and mitochondrial function. A subset of miRNAs was validated in the G93A SOD1 mouse at four stages of disease, and several appeared to track disease progression, including miR-206. We assessed these miRNAs in a large cohort of human ALS and disease control samples and found that some had similar changes but were not specific for ALS. Surprisingly, miR-206 levels did not change overall compared to normal controls, but did correlate with changes in strength of the muscle biopsied. In summary, we identified distinct miRNA patterns in ALS muscle that reflected disease stage which could potentially be used as biomarkers of disease activity.
Collapse
|
146
|
Inhibition of the JNK/MAPK signaling pathway by myogenesis-associated miRNAs is required for skeletal muscle development. Cell Death Differ 2018; 25:1581-1597. [PMID: 29449644 PMCID: PMC6143622 DOI: 10.1038/s41418-018-0063-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 12/24/2017] [Accepted: 01/04/2018] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle differentiation is controlled by multiple cell signaling pathways, however, the JNK/MAPK signaling pathway dominating this process has not been fully elucidated. Here, we report that the JNK/MAPK pathway was significantly downregulated in the late stages of myogenesis, and in contrast to P38/MAPK pathway, it negatively regulated skeletal muscle differentiation. Based on the PAR-CLIP-seq analysis, we identified six elevated miRNAs (miR-1a-3p, miR-133a-3p, miR-133b-3p, miR-206-3p, miR-128-3p, miR-351-5p), namely myogenesis-associated miRNAs (mamiRs), negatively controlled the JNK/MAPK pathway by repressing multiple factors for the phosphorylation of the JNK/MAPK pathway, including MEKK1, MEKK2, MKK7, and c-Jun but not JNK protein itself, and as a result, expression of transcriptional factor MyoD and mamiRs were further promoted. Our study revealed a novel double-negative feedback regulatory pattern of cell-specific miRNAs by targeting phosphorylation kinase signaling cascade responsible for skeletal muscle development.
Collapse
|
147
|
Szigyarto CAK, Spitali P. Biomarkers of Duchenne muscular dystrophy: current findings. Degener Neurol Neuromuscul Dis 2018; 8:1-13. [PMID: 30050384 PMCID: PMC6053903 DOI: 10.2147/dnnd.s121099] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Numerous biomarkers have been unveiled in the rapidly evolving biomarker discovery field, with an aim to improve the clinical management of disorders. In rare diseases, such as Duchenne muscular dystrophy, this endeavor has created a wealth of knowledge that, if effectively exploited, will benefit affected individuals, with respect to health care, therapy, improved quality of life and increased life expectancy. The most promising findings and molecular biomarkers are inspected in this review, with an aim to provide an overview of currently known biomarkers and the technological developments used. Biomarkers as cells, genetic variations, miRNAs, proteins, lipids and/or metabolites indicative of disease severity, progression and treatment response have the potential to improve development and approval of therapies, clinical management of DMD and patients’ life quality. We highlight the complexity of translating research results to clinical use, emphasizing the need for biomarkers, fit for purpose and describe the challenges associated with qualifying biomarkers for clinical applications.
Collapse
Affiliation(s)
- Cristina Al-Khalili Szigyarto
- Division of Proteomics, School of Biotechnology, AlbaNova University Center, KTH-Royal Institute of Technology, Stockholm, Sweden, .,Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden,
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands,
| |
Collapse
|
148
|
Moushi A, Michailidou K, Soteriou M, Cariolou M, Bashiardes E. MicroRNAs as possible biomarkers for screening of aortic aneurysms: a systematic review and validation study. Biomarkers 2018; 23:253-264. [PMID: 29297231 DOI: 10.1080/1354750x.2018.1423704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
CONTEXT There is an urgent need to identify non-invasive biomarkers for the early detection of aortic aneurysms, preceding a fatal event. The potential role for MicroRNAs (miRNAs) as diagnostic markers for aortic aneurysms was investigated through the present systematic review. OBJECTIVE To perform a comprehensive review on published studies examining the association of miRNAs with aortic aneurysms and further validate these results with plasma samples collected from thoracic aortic aneurysm (TAA) patients. METHODS The literature search was performed via numerous databases and articles were only included if they fulfilled the predefined eligibility criteria. The miRNAs reported three times or more with expression consistency were validated using plasma samples from TAA patients collected before and following surgery. RESULTS Twenty-four articles were selected from the literature search and 11 miRNAs were chosen for validation using our samples. The miRNAs which were further validated were found to follow the trend in the regulation pattern as with the majority of the published data. MiRNA hsa-miR-193a-5p was found to be significantly down-regulated in the plasma samples collected before the aneurysmal removal when compared with postsurgical serum samples. CONCLUSIONS Numerous miRNAs have been associated with aortic aneurysms, and specifically hsa-miR-193a-5p and hsa-miR-30b-5p; therefore they warrant further investigation as potential biomarkers. Registration: The protocol of the review was registered in Prospero Databases (ID: CRD42016039953).
Collapse
Affiliation(s)
- Areti Moushi
- a Cyprus School of Molecular Medicine , The Cyprus Institute of Neurology and Genetics , Nicosia , Cyprus
| | - Kyriaki Michailidou
- b Department of Electron Microscopy/Molecular Pathology , The Cyprus Institute of Neurology and Genetics , Nicosia, Cyprus
| | | | - Marios Cariolou
- a Cyprus School of Molecular Medicine , The Cyprus Institute of Neurology and Genetics , Nicosia , Cyprus.,d Department of Cardiovascular Genetics and The Laboratory of Forensic Genetics , The Cyprus Institute of Neurology and Genetics , Nicosia , Cyprus
| | - Evy Bashiardes
- a Cyprus School of Molecular Medicine , The Cyprus Institute of Neurology and Genetics , Nicosia , Cyprus.,d Department of Cardiovascular Genetics and The Laboratory of Forensic Genetics , The Cyprus Institute of Neurology and Genetics , Nicosia , Cyprus
| |
Collapse
|
149
|
Song Y, Lu S, Zhao J, Wang L. Nuclear Receptor SHP: A Critical Regulator of miRNA and lncRNA Expression and Function. NUCLEAR RECEPTOR RESEARCH 2017; 4:101312. [PMID: 30148159 PMCID: PMC6103530 DOI: 10.11131/2017/101312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Small heterodimer partner (SHP, NR0B2) is identified as a unique orphan nuclear receptor that acts as a transcriptional repressor. SHP plays a crucial role in the control of various physiological processes and in several diseases by regulating the expression of disease-specific genes. Non-coding RNAs (ncRNAs), including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), are encoded of RNAs that are transcribed but not translated into proteins, which are involved in diverse developmental and cellular processes in eukaryotic organisms. Research during the past decade has identified factors participating in the regulation of ncRNAs biogenesis and function. In this review, we summarize recent findings demonstrating a critical role of SHP as a transcriptional regulator of ncRNAs expression and function.
Collapse
Affiliation(s)
- Yongfeng Song
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, 250021, China
- Department of Physiology and Neurobiology, and the Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Shan Lu
- Genesis Biotechnology, Trenton, NJ 08619, USA
| | - Jiajun Zhao
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, 250021, China
| | - Li Wang
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, 250021, China
- Department of Physiology and Neurobiology, and the Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
150
|
Saliani N, Montazersaheb S, Montasser Kouhsari S. Micromanaging Glucose Tolerance and Diabetes. Adv Pharm Bull 2017; 7:547-556. [PMID: 29399544 PMCID: PMC5788209 DOI: 10.15171/apb.2017.066] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/05/2017] [Accepted: 12/12/2017] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous non-coding RNAs that have significant roles in biological processes such as glucose homoeostasis. MiRNAs fine-tune target genes expression via sequence-specific binding of their seed sequence to the untranslated region of mRNAs and degrade target mRNAs. MicroRNAs in islet β-cells regulate β-cell differentiation, proliferation, insulin transcription and glucose-stimulated insulin secretion. Furthermore, miRNAs play key roles in the regulation of glucose and lipid metabolisms and modify insulin sensitivity by controlling metabolic functions in main target organs of insulin such as skeletal muscle, liver and adipose tissue. Moreover, since circulating miRNAs are detectable and stable in serum, levels of certain miRNAs seem to be novel biomarkers for prediction of diabetes mellitus. In this article, due to the prominent impact of miRNAs on diabetes, we overviewed the microRNAs regulatory functions in organs related to insulin resistance and diabetes and shed light on their potential as diagnostic and therapeutic markers for diabetes.
Collapse
Affiliation(s)
- Negar Saliani
- Department of Cellular and Molecular Biology, School of Biology, College of Sciences, University of Tehran, Tehran, Iran
| | | | - Shideh Montasser Kouhsari
- Department of Cellular and Molecular Biology, School of Biology, College of Sciences, University of Tehran, Tehran, Iran
| |
Collapse
|