1
|
Niu Y, Guo D, Wei Y, Li J, Bai Y, Liu Z, Jia X, Chen Z, Li L, Shi B, Zhang X, Zhao Z, Hu J, Wang J, Liu X, Li S. Comparative Transcriptome Analysis of mRNA and miRNA during the Development of Longissimus Dorsi Muscle of Gannan Yak and Tianzhu White Yak. Animals (Basel) 2024; 14:2278. [PMID: 39123804 PMCID: PMC11311108 DOI: 10.3390/ani14152278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
The Gannan yak, a superior livestock breed found on the Tibetan Plateau, exhibits significantly enhanced body size, weight, and growth performance in comparison to the Tianzhu white yak. MiRNAs play a pivotal role in regulating muscle growth by negatively modulating target genes. In this study, we found the average diameter, area, and length of myofibers in Gannan yaks were significantly higher than those of Tianzhu white yaks. Further, we focused on analyzing the longissimus dorsi muscle from both Gannan yaks and Tianzhu white yaks through transcriptome sequencing to identify differentially expressed (DE)miRNAs that influence skeletal muscle development. A total of 254 DE miRNAs were identified, of which 126 miRNAs were up-regulated and 128 miRNAs were down-regulated. GO and KEGG enrichment analysis showed that the target genes of these DE miRNAs were significantly enriched in signaling pathways associated with muscle growth and development. By constructing a DE miRNA- DE mRNA interaction network, we screened 18 key miRNAs, and notably, four of the candidates (novel-m0143-3p, novel-m0024-3p, novel-m0128-5p, and novel-m0026-3p) targeted six genes associated with muscle growth and development (DDIT4, ADAMTS1, CRY2, AKIRIN2, SIX1, and FOXO1). These findings may provide theoretical references for further studies on the role of miRNAs in muscle growth and development in Gannan yaks.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Zhidong Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.N.); (D.G.); (Y.W.); (J.L.); (Y.B.); (Z.L.); (X.J.); (Z.C.); (L.L.); (B.S.); (X.Z.); (J.H.); (J.W.); (X.L.); (S.L.)
| | | | | | | | | |
Collapse
|
2
|
O'Bryan SM, Lavin KM, Graham ZA, Drummer DJ, Tuggle SC, Van Keuren-Jensen K, Reiman R, Alsop E, Kadakia MP, Craig MP, Zhang J, Bamman MM. Muscle-derived microRNAs correlated with thigh lean mass gains during progressive resistance training in older adults. J Appl Physiol (1985) 2024; 137:262-273. [PMID: 38932684 PMCID: PMC11424181 DOI: 10.1152/japplphysiol.00680.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Resistance training (RT) remains the most effective treatment for age-related declines in muscle mass. However, many older adults experience attenuated muscle hypertrophy in response to RT when compared with younger adults. This may be attributed to underlying molecular processes that are dysregulated by aging and exacerbated by improperly prescribed RT weekly volume, intensity, and/or frequency doses. MicroRNAs (miRNAs) are key epigenetic regulators that impact signaling pathways and protein expression within cells, are dynamic and responsive to exercise stimuli, and are often dysregulated in diseases. In this study, we used untargeted miRNA-seq to examine miRNA in skeletal muscle and serum-derived exosomes of older adults (n = 18, 11 M/7 F, 66 ± 1 yr) who underwent three times per wk RT for 30 wk [e.g., high intensity three times/wk (HHH, n = 9) or alternating high-low-high (HLH) intensity (n = 9)], after a standardized 4-wk washin. Within each tissue, miRNAs were clustered into modules based on pairwise correlation using weighted gene correlation network analysis (WGCNA). Modules were tested for association with the magnitude of RT-induced thigh lean mass (TLM) change [as measured by dual-energy X-ray absorptiometry (DXA)]. Although no modules were unique to training dose, we identified miRNA modules in skeletal muscle associated with TLM gains irrespective of exercise dose. Using miRNA-target interactions, we analyzed key miRNAs in significant modules for their potential regulatory involvement in biological pathways. Findings point toward potential miRNAs that may be informative biomarkers and could also be evaluated as potential therapeutic targets as an adjuvant to RT to maximize skeletal muscle mass accrual in older adults.NEW & NOTEWORTHY In this work, we identified a set of microRNAs correlated with thigh lean mass gains in a group of older adults. To our knowledge, this is the first time these microRNAs have been identified as novel predictive biomarkers correlating with lean mass gains in aging adults. As biomarkers, these may help interventionalists identify older individuals that are positively responding to an exercise intervention.
Collapse
Affiliation(s)
- Samia M O'Bryan
- UAB Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kaleen M Lavin
- Healthspan, Resilience, and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, Florida, United States
| | - Zachary A Graham
- Healthspan, Resilience, and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, Florida, United States
| | - Devin J Drummer
- UAB Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - S Craig Tuggle
- Healthspan, Resilience, and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, Florida, United States
| | | | - Rebecca Reiman
- Translational Genomics Research Institute, Phoenix, Arizona, United States
| | - Eric Alsop
- Translational Genomics Research Institute, Phoenix, Arizona, United States
| | - Madhavi P Kadakia
- Boonshoft School of Medicine, Wright State University, Dayton, Ohio, United States
| | - Michael P Craig
- Boonshoft School of Medicine, Wright State University, Dayton, Ohio, United States
| | - Jin Zhang
- Boonshoft School of Medicine, Wright State University, Dayton, Ohio, United States
| | - Marcas M Bamman
- UAB Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, United States
- Healthspan, Resilience, and Performance Research, Florida Institute for Human and Machine Cognition, Pensacola, Florida, United States
| |
Collapse
|
3
|
Artigas-Arias M, Curi R, Marzuca-Nassr GN. Myogenic microRNAs as Therapeutic Targets for Skeletal Muscle Mass Wasting in Breast Cancer Models. Int J Mol Sci 2024; 25:6714. [PMID: 38928418 PMCID: PMC11204047 DOI: 10.3390/ijms25126714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer is the type of cancer with the highest prevalence in women worldwide. Skeletal muscle atrophy is an important prognostic factor in women diagnosed with breast cancer. This atrophy stems from disrupted skeletal muscle homeostasis, triggered by diminished anabolic signalling and heightened inflammatory conditions, culminating in an upregulation of skeletal muscle proteolysis gene expression. The importance of delving into research on modulators of skeletal muscle atrophy, such as microRNAs (miRNAs), which play a crucial role in regulating cellular signalling pathways involved in skeletal muscle protein synthesis and degradation, has been recognised. This holds true for conditions of homeostasis as well as pathologies like cancer. However, the determination of specific miRNAs that modulate skeletal muscle atrophy in breast cancer conditions has not yet been explored. In this narrative review, we aim to identify miRNAs that could directly or indirectly influence skeletal muscle atrophy in breast cancer models to gain an updated perspective on potential therapeutic targets that could be modulated through resistance exercise training, aiming to mitigate the loss of skeletal muscle mass in breast cancer patients.
Collapse
Affiliation(s)
- Macarena Artigas-Arias
- Programa de Doctorado en Ciencias Mención Biología Celular y Molecular Aplicada, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo 01506-000, Brazil;
| | - Gabriel Nasri Marzuca-Nassr
- Departamento de Ciencias de la Rehabilitación, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| |
Collapse
|
4
|
García-Pérez I, Duran BOS, Dal-Pai-Silva M, Garcia de la serrana D. Exploring the Integrated Role of miRNAs and lncRNAs in Regulating the Transcriptional Response to Amino Acids and Insulin-like Growth Factor 1 in Gilthead Sea Bream ( Sparus aurata) Myoblasts. Int J Mol Sci 2024; 25:3894. [PMID: 38612703 PMCID: PMC11011856 DOI: 10.3390/ijms25073894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
In this study, gilthead sea bream (Sparus aurata) fast muscle myoblasts were stimulated with two pro-growth treatments, amino acids (AA) and insulin-like growth factor 1 (Igf-1), to analyze the transcriptional response of mRNAs, microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) and to explore their possible regulatory network using bioinformatic approaches. AA had a higher impact on transcription (1795 mRNAs changed) compared to Igf-1 (385 mRNAs changed). Both treatments stimulated the transcription of mRNAs related to muscle differentiation (GO:0042692) and sarcomere (GO:0030017), while AA strongly stimulated DNA replication and cell division (GO:0007049). Both pro-growth treatments altered the transcription of over 100 miRNAs, including muscle-specific miRNAs (myomiRs), such as miR-133a/b, miR-206, miR-499, miR-1, and miR-27a. Among 111 detected lncRNAs (>1 FPKM), only 30 were significantly changed by AA and 11 by Igf-1. Eight lncRNAs exhibited strong negative correlations with several mRNAs, suggesting a possible regulation, while 30 lncRNAs showed strong correlations and interactions with several miRNAs, suggesting a role as sponges. This work is the first step in the identification of the ncRNAs network controlling muscle development and growth in gilthead sea bream, pointing out potential regulatory mechanisms in response to pro-growth signals.
Collapse
Affiliation(s)
- Isabel García-Pérez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain;
| | - Bruno Oliveira Silva Duran
- Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil;
| | - Maeli Dal-Pai-Silva
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, Brazil;
| | - Daniel Garcia de la serrana
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona (UB), 08028 Barcelona, Spain;
| |
Collapse
|
5
|
Yang Y, GuangXuan H, GenMeng W, MengHuan L, Bo C, XueJie Y. Idiopathic inflammatory myopathy and non-coding RNA. Front Immunol 2023; 14:1227945. [PMID: 37744337 PMCID: PMC10512060 DOI: 10.3389/fimmu.2023.1227945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/11/2023] [Indexed: 09/26/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are common autoimmune diseases that affect skeletal muscle quality and function. The lack of an early diagnosis and treatment can lead to irreversible muscle damage. Non-coding RNAs (ncRNAs) play an important role in inflammatory transfer, muscle regeneration, differentiation, and regulation of specific antibody levels and pain in IIMs. ncRNAs can be detected in blood and hair; therefore, ncRNAs detection has great potential for diagnosing, preventing, and treating IIMs in conjunction with other methods. However, the specific roles and mechanisms underlying the regulation of IIMs and their subtypes remain unclear. Here, we review the mechanisms by which micro RNAs and long non-coding RNA-messenger RNA networks regulate IIMs to provide a basis for ncRNAs use as diagnostic tools and therapeutic targets for IIMs.
Collapse
Affiliation(s)
- Yang Yang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Hu GuangXuan
- School of Physical Education, Liaoning Normal University, Dalian, Liaoning, China
| | - Wan GenMeng
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Li MengHuan
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Chang Bo
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Yi XueJie
- Social Science Research Center, Shenyang Sport University, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Fostiak K, Bichowska M, Trybulski R, Trabka B, Krzysztofik M, Rolnick N, Filip-Stachnik A, Wilk M. Acute Effects of Ischemic Intra-Conditioning on 30 m Sprint Performance. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12633. [PMID: 36231933 PMCID: PMC9566271 DOI: 10.3390/ijerph191912633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
The present study aimed to evaluate the effects of ischemic intra-conditioning applied during rest intervals on 30 m sprint performance. Thirty-four trained male (n = 12) and female (n = 22) track and field and rugby athletes volunteered to participate in the study (age = 19.6 ± 4 years; training experience = 5.3 ± 1.9 years). In a randomized and counterbalanced order, participants performed six sets of 30 m sprints under three different testing conditions: without ischemic intra-conditioning, and with ischemic intra-conditioning at 60% or 80% arterial occlusion pressure applied bilaterally before the first trial of the sprint and during the rest periods between all sprint trials. During experimental sessions, subjects perform 6 × 30 m sprints with a 7 min rest interval between attempts. The cuffs were applied following a 1 min rest period and lasted for 5 min before being released at the 6th minute to allow for reperfusion (1 min + 5 min ischemic intra-conditioning + 1 min reperfusion). The two-way repeated measures ANOVA did not show statistically significant condition × set interaction for time of the sprint (p = 0.06; η2 = 0.05). There was also no main effect of ischemic intra-conditioning for any condition (p = 0.190; η2 = 0.05). This study indicates that ischemic intra-conditioning did not enhance the performance of 30 m sprints performed by athletes. However, ischemic intra-conditioning did not decrease performance either.
Collapse
Affiliation(s)
- Krzysztof Fostiak
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland
| | - Marta Bichowska
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland
| | - Robert Trybulski
- Provita Zory Medical Center, 44-240 Zory, Poland
- Department of Medical Sciences, The Wojciech Korfanty School of Economics, 40-065 Katowice, Poland
| | - Bartosz Trabka
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland
| | - Michal Krzysztofik
- Faculty of Physical Education and Sport, Charles University, 500 05 Prague, Czech Republic
| | - Nicholas Rolnick
- The Human Performance Mechanic, CUNY Lehman College, Bronx, New York, NY 10468, USA
| | - Aleksandra Filip-Stachnik
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education in Katowice, 40-065 Katowice, Poland
| | - Michal Wilk
- Institute of Sport Sciences, Jerzy Kukuczka Academy of Physical Education in Katowice, 40-065 Katowice, Poland
| |
Collapse
|
7
|
Ramadan F, Saab R, Hussein N, Clézardin P, Cohen PA, Ghayad SE. Non-coding RNA in rhabdomyosarcoma progression and metastasis. Front Oncol 2022; 12:971174. [PMID: 36033507 PMCID: PMC9403786 DOI: 10.3389/fonc.2022.971174] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/25/2022] [Indexed: 12/12/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a soft tissue sarcoma of skeletal muscle differentiation, with a predominant occurrence in children and adolescents. One of the major challenges facing treatment success is the presence of metastatic disease at the time of diagnosis, commonly associated with the more aggressive fusion-positive subtype. Non-coding RNA (ncRNA) can regulate gene transcription and translation, and their dysregulation has been associated with cancer development and progression. MicroRNA (miRNA) are short non-coding nucleic acid sequences involved in the regulation of gene expression that act by targeting messenger RNA (mRNA), and their aberrant expression has been associated with both RMS initiation and progression. Other ncRNA including long non-coding RNA (lncRNA), circular RNA (circRNA) and ribosomal RNA (rRNA) have also been associated with RMS revealing important mechanistic roles in RMS biology, but these studies are still limited and require further investigation. In this review, we discuss the established roles of ncRNA in RMS differentiation, growth and progression, highlighting their potential use in RMS prognosis, as therapeutic agents or as targets of treatment.
Collapse
Affiliation(s)
- Farah Ramadan
- Department of Biology, Faculty of Science II, Lebanese University, Beirut, Lebanon
- Université Claude Bernard Lyon 1, Lyon, France
- INSERM, Unit 1033, LYOS, Lyon, France
- Department of Chemistry and Biochemistry, Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science I, Lebanese University, Hadat, Lebanon
| | - Raya Saab
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nader Hussein
- Department of Chemistry and Biochemistry, Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science I, Lebanese University, Hadat, Lebanon
| | - Philippe Clézardin
- Université Claude Bernard Lyon 1, Lyon, France
- INSERM, Unit 1033, LYOS, Lyon, France
| | - Pascale A. Cohen
- Université Claude Bernard Lyon 1, Lyon, France
- INSERM, Unit 1033, LYOS, Lyon, France
| | - Sandra E. Ghayad
- Department of Biology, Faculty of Science II, Lebanese University, Beirut, Lebanon
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, Marseille, France
| |
Collapse
|
8
|
Ischemia during rest intervals between sets prevents decreases in fatigue during the explosive squat exercise: a randomized, crossover study. Sci Rep 2022; 12:5922. [PMID: 35396528 PMCID: PMC8993849 DOI: 10.1038/s41598-022-10022-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/29/2022] [Indexed: 01/14/2023] Open
Abstract
The study aimed to evaluate the impact of ischemia, used only before particular sets of a lower limb resistance exercise on power output. Ten healthy resistance-trained males (age = 26 ± 6 years; body mass = 90 ± 9 kg; training experience = 9 ± 7 years) performed two experimental sessions (with ischemia; control without ischemia) following a randomized crossover design. During the ischemic condition, the cuffs were inflated to 60% of arterial occlusion pressure. The cuffs were applied before each set for 4.5 min and released 30 s before the start of the set as the reperfusion (4.5 min ischemia + 0.5 min reperfusion). In the control condition, ischemia was not applied. During the experimental sessions, the subjects performed the Keiser machine squat exercise protocol which consisted of 5 sets of two repetitions, at a load of 60% of one-repetition maximum (1RM), with 5 min rest intervals between sets. The repetitions were performed with maximal velocity. The two-way repeated-measures ANOVA showed a statistically significant interaction effect for power output (p < 0.01; η2 = 0.26). There was also a statistically significant main effect of condition for power output (p = 0.02; η2 = 0.40). The post hoc analysis for interaction did not show significant differences between conditions in particular sets. The post hoc analysis for the main effect of the condition revealed that power output was significantly lower in the control group compared to the group where ischemic was used (p = 0.02). The t-test comparisons for particular sets showed a significant lower power output in set 3 (p = 0.03); set 4 (p < 0.01) and set 5 (p < 0.01) for the control condition when compared to the ischemic condition. The results indicate that ischemia applied before each set and released 30 s prior to the start of the squat exercise did not increase power output performance. However, we observed a significantly lower decline in power for the ischemic condition (4.5 min ischemia + 0.5 min reperfusion) in sets 3–5 compared to the control condition. Thus repeated ischemia with reperfusion used between sets can be an effective form of performance enhancement by preventing or at least diminishing fatigue during resistance exercise.
Collapse
|
9
|
Saiyed AN, Vasavada AR, Johar SRK. Recent trends in miRNA therapeutics and the application of plant miRNA for prevention and treatment of human diseases. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022; 8:24. [PMID: 35382490 PMCID: PMC8972743 DOI: 10.1186/s43094-022-00413-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/21/2022] [Indexed: 02/17/2023] Open
Abstract
Background Researchers now have a new avenue to investigate when it comes to miRNA-based therapeutics. miRNAs have the potential to be valuable biomarkers for disease detection. Variations in miRNA levels may be able to predict changes in normal physiological processes. At the epigenetic level, miRNA has been identified as a promising candidate for distinguishing and treating various diseases and defects. Main body In recent pharmacology, plants miRNA-based drugs have demonstrated a potential role in drug therapeutics. The purpose of this review paper is to discuss miRNA-based therapeutics, the role of miRNA in pharmacoepigenetics modulations, plant miRNA inter-kingdom regulation, and the therapeutic value and application of plant miRNA for cross-kingdom approaches. Target prediction and complementarity with host genes, as well as cross-kingdom gene interactions with plant miRNAs, are also revealed by bioinformatics research. We also show how plant miRNA can be transmitted from one species to another by crossing kingdom boundaries in this review. Despite several unidentified barriers to plant miRNA cross-transfer, plant miRNA-based gene regulation in trans-kingdom gene regulation may soon be valued as a possible approach in plant-based drug therapeutics. Conclusion This review summarised the biochemical synthesis of miRNAs, pharmacoepigenetics, drug therapeutics and miRNA transkingdom transfer.
Collapse
Affiliation(s)
- Atiyabanu N. Saiyed
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat India
- Ph.D. scholar of Manipal Academy of Higher Education, Manipal, Karnataka India
| | - Abhay R. Vasavada
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat India
| | - S. R. Kaid Johar
- Department of Zoology, BMTC, Human Genetics, USSC, Gujarat University, Ahmedabad, Gujarat India
| |
Collapse
|
10
|
Yedigaryan L, Sampaolesi M. Therapeutic Implications of miRNAs for Muscle-Wasting Conditions. Cells 2021; 10:cells10113035. [PMID: 34831256 PMCID: PMC8616481 DOI: 10.3390/cells10113035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules that are mainly involved in translational repression by binding to specific messenger RNAs. Recently, miRNAs have emerged as biomarkers, relevant for a multitude of pathophysiological conditions, and cells can selectively sort miRNAs into extracellular vesicles for paracrine and endocrine effects. In the overall context of muscle-wasting conditions, a multitude of miRNAs has been implied as being responsible for the typical dysregulation of anabolic and catabolic pathways. In general, chronic muscle disorders are associated with the main characteristic of a substantial loss in muscle mass. Muscular dystrophies (MDs) are a group of genetic diseases that cause muscle weakness and degeneration. Typically, MDs are caused by mutations in those genes responsible for upholding the integrity of muscle structure and function. Recently, the dysregulation of miRNA levels in such pathological conditions has been reported. This revelation is imperative for both MDs and other muscle-wasting conditions, such as sarcopenia and cancer cachexia. The expression levels of miRNAs have immense potential for use as potential diagnostic, prognostic and therapeutic biomarkers. Understanding the role of miRNAs in muscle-wasting conditions may lead to the development of novel strategies for the improvement of patient management.
Collapse
Affiliation(s)
- Laura Yedigaryan
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
- Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
11
|
Wang Y, Zhen D, Fu D, Fu Y, Zhang X, Gong G, Wei C. 1, 8-cineole attenuates cardiac hypertrophy in heart failure by inhibiting the miR-206-3p/SERP1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153672. [PMID: 34385094 DOI: 10.1016/j.phymed.2021.153672] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/06/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND 1,8-Cineole (1,8-CIN) is a monoterpene found in diverse dietary and medicinal herbs that has been reported to be effective against cardiovascular diseases. PURPOSE The present research was designed to elucidate the treatment effects and the underlying mechanism of 1,8-CIN on heart failure (HF). METHOD An in vitro cardiac hypertrophy model and an in vivo heart failure (HF) model induced by isoprenaline (ISO) were established and treated with or without 1,8-CIN. In vitro miR-206-3p mimic or inhibitors were created. MiR-206-3p, SERP1 and related mRNAs or proteins were detected using qPCR or western blotting. Cell viability was tested by MTT assay, and apoptosis was measured using TUNEL assay, AO/EB assay and flow cytometry. Actin was stained with FITC-phalloidin. MiR-206-3p and related mRNAs or proteins in cardiac muscle tissues were measured using qPCR or western blotting, HE staining, Masson staining. RESULTS ISO subcutaneous injection increased cardiac hypertrophy, cytoplasmic vacuole formation, myofiber loss and fibrosis and decreased cardiomyocyte viability. 1,8-CIN treatment improved cardiomyocyte viability and reduced cardiac hypertrophy, cytoplasmic vacuole formation, myofibre loss and fibrosis. We found that 1,8-CIN attenuated apoptosis. We observed that expression of miR-206-3p was dramatically increased in ISO-exposed cardiomyocytes or ISO-treated rat hearts. MiR-206-3p was identified to target the 3'UTR of SERP1, resulting in the accumulation of un- or misfolded proteins, leading to endoplasmic reticulum (ER) stress. CONCLUSION These results suggest that 1,8-CIN reduces the apoptosis induced by ER stress through inhibiting miR-206-3p, which inhibits the expression of SERP1.
Collapse
Affiliation(s)
- Yu Wang
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Dong Zhen
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Danni Fu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Yao Fu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Xuan Zhang
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Guohua Gong
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China; Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, PR China.
| | - Chengxi Wei
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China.
| |
Collapse
|
12
|
Histone deacetylase HDAC4 participates in the pathological process of myocardial ischemia-reperfusion injury via MEKK1/JNK pathway by binding to miR-206. Cell Death Discov 2021; 7:240. [PMID: 34526481 PMCID: PMC8443671 DOI: 10.1038/s41420-021-00601-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/29/2021] [Accepted: 07/26/2021] [Indexed: 12/29/2022] Open
Abstract
Histone deacetylases (HDACs) and microRNAs (miRs) have been reported to exert pivotal roles on the pathogenesis of myocardial ischemia-reperfusion injury (MIRI). Therefore, the present study was performed to define the underlying role of HDAC4 and miR-206 in the pathological process of MIRI. An IRI rat model was established. The interaction between HDAC4 and the promoter region of miR-206 was determined using ChIP, and that between miR-206 and mitogen-activated protein kinase kinase kinase 1 (MEKK1) was determined using dual luciferase reporter gene assay. After the loss- or gain-of-function assay in cardiomyocytes, western blot analysis, RT-qPCR, TUNEL, and ELISA assay were performed to define the roles of HDAC4, miR-206, and MEKK1. Up-regulation of HDAC4 and down-regulation of miR-206 occurred in rat myocardial tissues and cardiomyocytes in MIRI. HDAC4 down-regulation or miR-206 up-regulation contributed to reduced cell apoptosis and the levels of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and malondialdehyde (MDA), while elevating the superoxide dismutase (SOD) and glutathione (GSH) contents. Meanwhile, HDAC4 silencing promoted the expression of miR-206, which targeted and negatively regulated MEKK1. Then inhibition of JNK phosphorylation reduced the cardiomyocyte apoptosis to alleviate MIRI. Coherently, HDAC4 silencing could up-regulate the expression of miR-206 to reduce cardiomyocyte apoptosis and inhibit oxidative stress, and exerting a protective effect on MIRI via the MEKK1/JNK pathway.
Collapse
|
13
|
Torma F, Gombos Z, Fridvalszki M, Langmar G, Tarcza Z, Merkely B, Naito H, Ichinoseki-Sekine N, Takeda M, Murlasits Z, Osvath P, Radak Z. Blood flow restriction in human skeletal muscle during rest periods after high-load resistance training down-regulates miR-206 and induces Pax7. JOURNAL OF SPORT AND HEALTH SCIENCE 2021; 10:470-477. [PMID: 32813644 PMCID: PMC8343007 DOI: 10.1016/j.jshs.2019.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/24/2019] [Accepted: 06/26/2019] [Indexed: 05/25/2023]
Abstract
BACKGROUD Blood flow restriction (BFR) with low-intensity resistance training has been shown to result in hypertrophy of skeletal muscle. In this study, we tested the hypothesis that BFR during the rest periods between acute, high-intensity resistance exercise sessions (70% of 1 repetition maximum, 7 sets with 10 repetitions) enhances the effects of the resistance training. METHODS A total of 7 healthy young men performed squats, and between sets BFR was carried out on one leg while the other leg served as a control. Because BFR was applied during rest periods, even severe occlusion pressure (approximately 230 mmHg), which almost completely blocked blood flow, was well-tolerated by the participants. Five muscle-specific microRNAs were measured from the biopsy samples, which were taken 2 h after the acute training. RESULTS Doppler data showed that the pattern of blood flow recovery changed significantly between the first and last BFR. microRNA-206 levels significantly decreased in the BFR leg compared to the control. The mRNA levels of RAC-β serine/threonine-protein kinase v22, nuclear respiratory factor 1, vascular endothelial growth factor, lupus Ku autoantigen protein p70 genes (p < 0.05), and paired box 7 (p < 0.01) increased in the BFR leg. The protein levels of paired box 7, nuclear respiratory factor 1, and peroxisome proliferator-activated receptor γ coactivator 1α did not differ between the BFR leg and the control leg. CONCLUSION BFR, during the rest periods of high-load resistance training, could lead to mRNA elevation of those proteins that regulate angiogenesis, mitochondrial biogenesis, and muscle hypertrophy and repair. However, BFR also can cause DNA damage, judging from the increase in mRNA levels of lupus Ku autoantigen protein p70.
Collapse
Affiliation(s)
- Ferenc Torma
- Research Center for Molecular Exercise Science, University of Physical Education, Budapest 1123, Hungary
| | - Zoltan Gombos
- Research Center for Molecular Exercise Science, University of Physical Education, Budapest 1123, Hungary
| | - Marcell Fridvalszki
- Department of Kinesiology, University of Physical Education, Budapest 1123, Hungary
| | - Gergely Langmar
- Department of Kinesiology, University of Physical Education, Budapest 1123, Hungary
| | - Zsofia Tarcza
- Heart and Vascular Center, Semmelweis University, Budapest 1122, Hungary
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest 1122, Hungary
| | - Hisashi Naito
- Faculty of Health and Sports Science, Juntendo University, Chiba 270-1695, Japan
| | | | - Masaki Takeda
- Faculty of Health and Sports Science, Doshisha University, Kyotanabe 610-0394, Japan
| | - Zsolt Murlasits
- Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Peter Osvath
- Department of Health Sciences and Sport Medicine, University of Physical Education, Budapest 1123, Hungary
| | - Zsolt Radak
- Research Center for Molecular Exercise Science, University of Physical Education, Budapest 1123, Hungary.
| |
Collapse
|
14
|
da Paixão AO, Bolin AP, Silvestre JG, Rodrigues AC. Palmitic Acid Impairs Myogenesis and Alters Temporal Expression of miR-133a and miR-206 in C2C12 Myoblasts. Int J Mol Sci 2021; 22:ijms22052748. [PMID: 33803124 PMCID: PMC7963199 DOI: 10.3390/ijms22052748] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/26/2022] Open
Abstract
Palmitic acid (PA), a saturated fatty acid enriched in high-fat diet, has been implicated in the development of sarcopenic obesity. Herein, we chose two non-cytotoxic concentrations to better understand how excess PA could impact myotube formation or diameter without inducing cell death. Forty-eight hours of 100 µM PA induced a reduction of myotube diameter and increased the number of type I fibers, which was associated with increased miR-206 expression. Next, C2C12 myotube growth in the presence of PA was evaluated. Compared to control cells, 150 µM PA reduces myoblast proliferation and the expression of MyoD and miR-206 and miR-133a expression, leading to a reduced number and diameter of myotubes. PA (100 µM), despite not affecting proliferation, impairs myotube formation by reducing the expression of Myf5 and miR-206 and decreasing protein synthesis. Interestingly, 100 and 150 µM PA-treated myotubes had a higher number of type II fibers than control cells. In conclusion, PA affects negatively myotube diameter, fusion, and metabolism, which may be related to myomiRs. By providing new insights into the mechanisms by which PA affects negatively skeletal muscle, our data may help in the discovery of new targets to treat sarcopenic obesity.
Collapse
Affiliation(s)
- Ailma O. da Paixão
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (A.O.d.P.); (A.P.B.)
| | - Anaysa Paola Bolin
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (A.O.d.P.); (A.P.B.)
| | - João G. Silvestre
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil;
| | - Alice Cristina Rodrigues
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (A.O.d.P.); (A.P.B.)
- Correspondence: ; Tel.: +55-11-3091-7406
| |
Collapse
|
15
|
Yoshihara T, Naito H. Protective effects of acute exercise preconditioning on disuse-induced muscular atrophy in aged muscle: a narrative literature review. J Physiol Sci 2020; 70:55. [PMID: 33246401 PMCID: PMC10717045 DOI: 10.1186/s12576-020-00783-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/19/2020] [Indexed: 11/10/2022]
Abstract
Aging is associated with a progressive loss of skeletal muscle mass and strength, resulting in frailty and lower quality of life in older individuals. At present, a standard of clinical or pharmacological care to prevent the adverse effects of aging does not exist. Determining the mechanism(s) responsible for muscular atrophy in disused aged muscle is a required key step for the development of effective countermeasures. Studies suggest an age-related differential response of genes and signalings to muscle disuse in both rodents and humans, implying the possibility that effective countermeasures to prevent disuse muscle atrophy may be age-specific. Notably, exercise preconditioning can attenuate disuse-induced muscular atrophy in rodent and human skeletal muscles; however, information on age-specific mechanisms of this exercise-induced protection remains limited. This mini-review aimed to summarize the protective effects of acute exercise preconditioning on muscular atrophy in aged muscle and provide potential mechanisms for its preventive effect on skeletal muscle wasting.
Collapse
Affiliation(s)
- Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| |
Collapse
|
16
|
Marceca GP, Nigita G, Calore F, Croce CM. MicroRNAs in Skeletal Muscle and Hints on Their Potential Role in Muscle Wasting During Cancer Cachexia. Front Oncol 2020; 10:607196. [PMID: 33330108 PMCID: PMC7732629 DOI: 10.3389/fonc.2020.607196] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Cancer-associated cachexia is a heterogeneous, multifactorial syndrome characterized by systemic inflammation, unintentional weight loss, and profound alteration in body composition. The main feature of cancer cachexia is represented by the loss of skeletal muscle tissue, which may or may not be accompanied by significant adipose tissue wasting. Such phenotypic alteration occurs as the result of concomitant increased myofibril breakdown and reduced muscle protein synthesis, actively contributing to fatigue, worsening of quality of life, and refractoriness to chemotherapy. According to the classical view, this condition is primarily triggered by interactions between specific tumor-induced pro-inflammatory cytokines and their cognate receptors expressed on the myocyte membrane. This causes a shift in gene expression of muscle cells, eventually leading to a pronounced catabolic condition and cell death. More recent studies, however, have shown the involvement of regulatory non-coding RNAs in the outbreak of cancer cachexia. In particular, the role exerted by microRNAs is being widely addressed, and several mechanistic studies are in progress. In this review, we discuss the most recent findings concerning the role of microRNAs in triggering or exacerbating muscle wasting in cancer cachexia, while mentioning about possible roles played by long non-coding RNAs and ADAR-mediated miRNA modifications.
Collapse
Affiliation(s)
- Gioacchino P Marceca
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giovanni Nigita
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Federica Calore
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
17
|
Chen R, Lei S, Jiang T, She Y, Shi H. Regulation of Skeletal Muscle Atrophy in Cachexia by MicroRNAs and Long Non-coding RNAs. Front Cell Dev Biol 2020; 8:577010. [PMID: 33043011 PMCID: PMC7523183 DOI: 10.3389/fcell.2020.577010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle atrophy is a common complication of cachexia, characterized by progressive bodyweight loss and decreased muscle strength, and it significantly increases the risks of morbidity and mortality in the population with atrophy. Numerous complications associated with decreased muscle function can activate catabolism, reduce anabolism, and impair muscle regeneration, leading to muscle wasting. microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), types of non-coding RNAs, are important for regulation of skeletal muscle development. Few studies have specifically identified the roles of miRNAs and lncRNAs in cellular or animal models of muscular atrophy during cachexia, and the pathogenesis of skeletal muscle wasting in cachexia is not entirely understood. To develop potential approaches to improve skeletal muscle mass, strength, and function, a more comprehensive understanding of the known key pathophysiological processes leading to muscular atrophy is needed. In this review, we summarize the known miRNAs, lncRNAs, and corresponding signaling pathways involved in regulating skeletal muscle atrophy in cachexia and other diseases. A comprehensive understanding of the functions and mechanisms of miRNAs and lncRNAs during skeletal muscle wasting in cachexia and other diseases will, therefore, promote therapeutic treatments for muscle atrophy.
Collapse
Affiliation(s)
- Rui Chen
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Si Lei
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ting Jiang
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanling She
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Huacai Shi
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
18
|
Chen Z, Zhang Z, Guo L, Wei X, Zhang Y, Wang X, Wei L. The role of histone deacetylase 4 during chondrocyte hypertrophy and endochondral bone development. Bone Joint Res 2020; 9:82-89. [PMID: 32435460 PMCID: PMC7229302 DOI: 10.1302/2046-3758.92.bjr-2019-0172.r1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chondrocyte hypertrophy represents a crucial turning point during endochondral bone development. This process is tightly regulated by various factors, constituting a regulatory network that maintains normal bone development. Histone deacetylase 4 (HDAC4) is the most well-characterized member of the HDAC class IIa family and participates in different signalling networks during development in various tissues by promoting chromatin condensation and transcriptional repression. Studies have reported that HDAC4-null mice display premature ossification of developing bones due to ectopic and early-onset chondrocyte hypertrophy. Overexpression of HDAC4 in proliferating chondrocytes inhibits hypertrophy and ossification of developing bones, which suggests that HDAC4, as a negative regulator, is involved in the network regulating chondrocyte hypertrophy. Overall, HDAC4 plays a key role during bone development and disease. Thus, understanding the role of HDAC4 during chondrocyte hypertrophy and endochondral bone formation and its features regarding the structure, function, and regulation of this process will not only provide new insight into the mechanisms by which HDAC4 is involved in chondrocyte hypertrophy and endochondral bone development, but will also create a platform for developing a therapeutic strategy for related diseases. Cite this article:Bone Joint Res. 2020;9(2):82–89.
Collapse
Affiliation(s)
- Zhi Chen
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhiwei Zhang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Li Guo
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaochun Wei
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yang Zhang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaojian Wang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Wei
- Department of Orthopedics, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
19
|
Li Y, Liu J. MicroRNA-206 predicts raised fetal growth retardation risk through the interaction with vascular endothelial growth factor in pregnancies. Medicine (Baltimore) 2020; 99:e18897. [PMID: 32049790 PMCID: PMC7035023 DOI: 10.1097/md.0000000000018897] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
This study aimed to investigate the correlation of microRNA (miR)-206, vascular endothelial growth factor (VEGF) and miR-206/VEGF axis at different gestational ages with fetal growth retardation (FGR) risk in pregnancies.Eight hundred twenty pregnancies were consecutively recruited and their plasma samples were collected at early pregnancy (gestational age ≤ 13 weeks), middle pregnancy (gestational age: 14-27 weeks) and late pregnancy (gestational age ≥ 28 weeks), respectively. miR-206 expression and VEGF level in plasma were detected by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay respectively. FGR was diagnosed based on the actual birth weight of fetus.miR-206 expression was negatively correlated with VEGF expression at early pregnancy, middle pregnancy and late pregnancy. Besides, miR-206 expression and miR-206/VEGF axis were elevated, but VEGF expression was decreased along with the increased gestational age. There were 74 FGR pregnancies and 746 non-FGR pregnancies. And both miR-206 expression and miR-206/VEGF axis were increased, but VEGF expression was reduced in FGR group compared to non-FGR group at early pregnancy, middle pregnancy and late pregnancy. Additionally, miR-206, VEGF and miR-206/VEGF axis at middle pregnancy and late pregnancy all showed good predictive values for FGR risk, and these indexes at late pregnancy exhibited the numerically highest predictive value for FGR risk. Furthermore, compared to miR-206 or VEGF alone, miR-206/VEGF axis presented with numerically higher predictive value for FGR risk.miR-206 predicts raised FGR risk through the interaction with VEGF in pregnancies, and it may serve as a novel biomarker for FGR prevention.
Collapse
Affiliation(s)
| | - Jiaqiang Liu
- Department of Hematology, People's Hospital of Rizhao, Shandong, China
| |
Collapse
|
20
|
Ooi JYY, Bernardo BC. Translational Potential of Non-coding RNAs for Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:343-354. [PMID: 32285423 DOI: 10.1007/978-981-15-1671-9_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jenny Y Y Ooi
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
21
|
In silico Design of Novel Histone Deacetylase 4 Inhibitors: Design Guidelines for Improved Binding Affinity. Int J Mol Sci 2019; 21:ijms21010219. [PMID: 31905609 PMCID: PMC6981887 DOI: 10.3390/ijms21010219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/19/2019] [Accepted: 12/25/2019] [Indexed: 02/02/2023] Open
Abstract
Histone deacetylases (HDAC) are being targeted for a number of diseases such as cancer, inflammatory disease, and neurological disorders. Within this family of 18 isozymes, HDAC4 is a prime target for glioma, one of the most aggressive brain tumors reported. Thus, the development of HDAC4 inhibitors could present a novel therapeutic route for glioma. In this work, molecular docking studies on cyclopropane hydroxamic acid derivatives identified five novel molecular interactions to the HDAC4 receptor that could be harnessed to enhance inhibitor binding. Thus, design guidelines for the optimization of potent HDAC4 inhibitors were developed which can be utilized to further the development of HDAC4 inhibitors. Using the developed guidelines, eleven novel cyclopropane hydroxamic acid derivatives were designed that outcompeted all original cyclopropane hydroxamic acids HDAC4 inhibitors studied in silico. The results of this work will be an asset to paving the way for further design and optimization of novel potent HDAC4 inhibitors for gliomas.
Collapse
|
22
|
Bao T, Han H, Li B, Zhao Y, Bou G, Zhang X, Du M, Zhao R, Mongke T, Laxima, Ding W, Jia Z, Dugarjaviin M, Bai D. The distinct transcriptomes of fast-twitch and slow-twitch muscles in Mongolian horses. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2019; 33:100649. [PMID: 31869634 DOI: 10.1016/j.cbd.2019.100649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 01/19/2023]
Abstract
Skeletal muscle is the largest organ system in the mammalian body and plays a key role in locomotion of horses. Fast and slow muscle fibers have different abilities and functions to adapt to exercises. To investigate the RNA and miRNA expression profiles in the muscles with different muscle fiber compositions on Mongolian horses. We examined the muscle fiber type population and produced deep RNA sequencing for different parts of skeletal muscles. And chose two of them with the highest difference in fast and slow muscle fiber population (splenius and gluteus medius) for comparing the gene expression profile of slow and fast muscle fiber types. We identified a total of 275 differentially expressed genes (DEGs), and 11 differentially expressed miRNAs (DEmiRs). In addition, target gene prediction and alternative splicing analysis were also performed. Significant correlations were found between the differentially expressed gene, miRNAs, and alternative splicing events. The result indicated that differentially expressed muscle-specific genes and target genes of miRNAs might co-regulating the performance of slow and fast muscle fiber types in Mongolian horses.
Collapse
Affiliation(s)
- Tugeqin Bao
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Haige Han
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Bei Li
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yiping Zhao
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Gerelchimeg Bou
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xinzhuang Zhang
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Ming Du
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Ruoyang Zhao
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Togtokh Mongke
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Laxima
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wenqi Ding
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Zijie Jia
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Manglai Dugarjaviin
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Dongyi Bai
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China.
| |
Collapse
|
23
|
Fan RF, Cao CY, Chen MH, Shi QX, Xu SW. Gga-let-7f-3p promotes apoptosis in selenium deficiency-induced skeletal muscle by targeting selenoprotein K. Metallomics 2019; 10:941-952. [PMID: 29905752 DOI: 10.1039/c8mt00083b] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Selenoprotein K (SELENOK) is primarily observed in the endoplasmic reticulum, and serves to maintain the normal physiological functions of skeletal muscle. Skeletal muscle development and regeneration are associated with significant changes in the expression of specific microRNAs (miRNAs). Downregulated SELENOK expression is observed in chicken muscles deficient of Se. However, the mechanisms of miRNA regulation of SELENOK expression remain elusive. Here, deep sequencing was used to detect the miRNA profiles of muscle in Se deficient (-Se group) and normal (C group) chickens. A dual-luciferase reporter assay was adopted to verify the relationship between SELENOK and gga-let-7f-3p. In addition, gga-let-7f-3p was either overexpressed or knocked-down in chicken myoblasts. Furthermore, the cells were treated with N-acetyl-l-cysteine (NAC) or hydrogen peroxide (H2O2) in order to probe the factors involved in oxidative stress, endoplasmic reticulum stress (ERS) and apoptosis, respectively. Relative to the C group, there were 132 differentially expressed miRNAs (including 57 upregulated and 75 downregulated) in the muscles of the -Se group. The dual-luciferase reporter assay showed that SELENOK was a primary target of gga-let-7f-3p. It was also observed that the overexpression or knock-down of gga-let-7f-3p significantly influenced the SELENOK expression. Moreover, NAC blocked mimics of ga-let-7f-3p, thus inducing oxidative stress, ERS and apoptosis. Simultaneously, gga-let-7f-3p inhibitors blocked the stimulant effects caused by H2O2 in chicken myoblasts. Furthermore, Se deficiency downregulated the SELENOK protein expression and induced oxidative stress, ERS and apoptosis in chicken muscles. In conclusion, the gga-let-7f-3p-SELENOK pathway played a pivotal role in Se deficiency mediated muscle injuries through the induction of oxidative stress and ERS, ultimately promoting apoptosis.
Collapse
Affiliation(s)
- Rui-Feng Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | | | | | | | | |
Collapse
|
24
|
Bittel DC, Jaiswal JK. Contribution of Extracellular Vesicles in Rebuilding Injured Muscles. Front Physiol 2019; 10:828. [PMID: 31379590 PMCID: PMC6658195 DOI: 10.3389/fphys.2019.00828] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/13/2019] [Indexed: 12/22/2022] Open
Abstract
Skeletal myofibers are injured due to mechanical stresses experienced during physical activity, or due to myofiber fragility caused by genetic diseases. The injured myofiber needs to be repaired or regenerated to restore the loss in muscle tissue function. Myofiber repair and regeneration requires coordinated action of various intercellular signaling factors-including proteins, inflammatory cytokines, miRNAs, and membrane lipids. It is increasingly being recognized release and transmission of these signaling factors involves extracellular vesicle (EV) released by myofibers and other cells in the injured muscle. Intercellular signaling by these EVs alters the phenotype of their target cells either by directly delivering the functional proteins and lipids or by modifying longer-term gene expression. These changes in the target cells activate downstream pathways involved in tissue homeostasis and repair. The EVs are heterogeneous with regards to their size, composition, cargo, location, as well as time-course of genesis and release. These differences impact on the subsequent repair and regeneration of injured skeletal muscles. This review focuses on how intracellular vesicle production, cargo packaging, and secretion by injured muscle, modulates specific reparative, and regenerative processes. Insights into the formation of these vesicles and their signaling properties offer new understandings of the orchestrated response necessary for optimal muscle repair and regeneration.
Collapse
Affiliation(s)
- Daniel C Bittel
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC, United States
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC, United States.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
25
|
Li T, Yu SS, Zhou CY, Wang K, Wan YC. MicroRNA-206 inhibition and activation of the AMPK/Nampt signalling pathway enhance sevoflurane post-conditioning-induced amelioration of myocardial ischaemia/reperfusion injury. J Drug Target 2019; 28:80-91. [PMID: 31092059 DOI: 10.1080/1061186x.2019.1616744] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Tao Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Shan-Shan Yu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Chang-Yu Zhou
- Department of Gastroenterology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Ke Wang
- Department of Gynaecology and Obstetrics, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Ying-Chun Wan
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
26
|
Rusanova I, Fernández-Martínez J, Fernández-Ortiz M, Aranda-Martínez P, Escames G, García-García FJ, Mañas L, Acuña-Castroviejo D. Involvement of plasma miRNAs, muscle miRNAs and mitochondrial miRNAs in the pathophysiology of frailty. Exp Gerontol 2019; 124:110637. [PMID: 31199979 DOI: 10.1016/j.exger.2019.110637] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023]
Abstract
Frailty is a geriatric syndrome that leads not only to the loss of physical functions, but also to a generalized decline of the organism and a high risk of disability and dependency. Frailty's detection and management represent important goals for current gerontology. The advance in its rapid diagnosis could play a relevant role in taking measures to reduce the negative consequences it exerts on the body and to take preventive measures. microRNAs are the one of multiple epigenetic biomarkers that reflect functional changes in aged subject. In this review we analyze microRNAs as molecules involved in the control of the pathways leading to the development of frailty. miRNAs can be present in different body fluids, including plasma/serum and saliva, can be associated with organelles like the mitochondria, and can be expressed in tissues. Based on the multifactorial physiopathology of frailty, we analyzed here the microRNAs linked to "inflammaging" (inflamma-miRs), to musculoskeletal health (myomiRs), and microRNAs that can directly or indirectly affect the mitochondria (mitomiRs). Subsequently, we analyze those microRNAs that can be modified by physical exercise. In this review we will analyze the latest experimental studies carried out in animals, cell cultures, and human samples, with the aim to identify gaps in the research and in order to try to dazzle the information about the pathways regulated by each miRNA. Multiple studies revised here suggest that several miRs can be considered as possible markers of frailty, including miR-1, miR-21, miR-34a, miR-146a, miR-185, and miR-206, miR-223, among others. Normalization of miRNAs data and standardization of the protocols used for their measurement to avoid confounding variables influencing the results, are important to use miRNAs as disease biomarkers.
Collapse
Affiliation(s)
- Iryna Rusanova
- Departamento de Fisiología, Facultad de Medicina, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain; CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain.
| | - José Fernández-Martínez
- Departamento de Fisiología, Facultad de Medicina, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Marisol Fernández-Ortiz
- Departamento de Fisiología, Facultad de Medicina, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Paula Aranda-Martínez
- Departamento de Fisiología, Facultad de Medicina, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Germaine Escames
- Departamento de Fisiología, Facultad de Medicina, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain; CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain
| | - Francisco J García-García
- CIBERfes, División de Medicina Geriátrica, Hospital Virgen del Valle, Complejo Hospitalario de Toledo, Toledo, Spain
| | - Leocadio Mañas
- CIBERfes, Servicio de Geriatría, Hospital Universitario de Getafe, Madrid, Spain
| | - Darío Acuña-Castroviejo
- Departamento de Fisiología, Facultad de Medicina, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain; CIBERfes, Ibs.Granada, and UGC de Laboratorios Clínicos, Complejo Hospitalario de Granada, Granada, Spain.
| |
Collapse
|
27
|
Yoshihara T, Tsuzuki T, Chang SW, Kakigi R, Sugiura T, Naito H. Exercise preconditioning attenuates hind limb unloading-induced gastrocnemius muscle atrophy possibly via the HDAC4/Gadd45 axis in old rats. Exp Gerontol 2019; 122:34-41. [PMID: 31009659 DOI: 10.1016/j.exger.2019.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/01/2019] [Accepted: 04/18/2019] [Indexed: 11/25/2022]
Abstract
The mechanisms involved in unloading-induced skeletal muscle loss may be age-specific, and the evidence for exercise preconditioning-induced protection against disuse muscle atrophy in aged rats is limited. Therefore, in this study, we investigated age-related differences in the activation of the HDAC4/Gadd45α pathway following hindlimb unloading (HU). We also assessed the protective effect of preconditioning exercise on this pathway in young and old rat gastrocnemius muscle. Three-month-old (young, n = 18) and 24-month-old (old, n = 18) male Wistar rats were assigned to the following groups: control group (n = 6), seven days of HU group (n = 6), and a bout of exercise preconditioning prior to HU (Ex+HU) group (n = 6). Rats of both ages in the Ex + HU group ran continuously on a motor-driven treadmill (0° slope, 20 m/min, 15 min) prior to HU. The gastrocnemius muscles were removed after 7 days of HU and analyzed for protein content and mRNA expression. Gastrocnemius muscle weight was significantly higher in the Ex+HU group than in the HU group of old rats, but not in young rats. Levels of HDAC4 protein and mRNA were significantly increased in the old HU group. However, the increase was significantly suppressed in the old Ex+HU group. Moreover, the protective effect of exercise preconditioning had a positive effect on Gadd45α mRNA and protein levels only in the old Ex+HU group. No exercise preconditioning-related protection was observed in the young rats. Our data indicated that a single bout of preconditioning exercise prior to HU may exert a protective effect in disuse muscle atrophy in old rats and that these effects may be partially mediated by the HDAC4/Gadd45α axis.
Collapse
Affiliation(s)
- Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan.
| | - Takamasa Tsuzuki
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan.
| | - Shuo-Wen Chang
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan.
| | - Ryo Kakigi
- Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Takao Sugiura
- Faculty of Education, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8513, Japan.
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba 270-1695, Japan.
| |
Collapse
|
28
|
Mukhopadhyay P, Smolenkova I, Warner D, Pisano MM, Greene RM. Spatio-Temporal Expression and Functional Analysis of miR-206 in Developing Orofacial Tissue. Microrna 2019; 8:43-60. [PMID: 30068287 DOI: 10.2174/2211536607666180801094528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/28/2018] [Accepted: 07/27/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND Development of the mammalian palate is dependent on precise, spatiotemporal expression of a panoply of genes. MicroRNAs (miRNAs), the largest family of noncoding RNAs, function as crucial modulators of cell and tissue differentiation, regulating expression of key downstream genes. OBSERVATIONS Our laboratory has previously identified several developmentally regulated miRNAs, including miR-206, during critical stages of palatal morphogenesis. The current study reports spatiotemporal distribution of miR-206 during development of the murine secondary palate (gestational days 12.5-14.5). RESULT AND CONCLUSION Potential cellular functions and downstream gene targets of miR-206 were investigated using functional assays and expression profiling, respectively. Functional analyses highlighted potential roles of miR-206 in governing TGFß- and Wnt signaling in mesenchymal cells of the developing secondary palate. In addition, altered expression of miR-206 within developing palatal tissue of TGFß3-/- fetuses reinforced the premise that crosstalk between this miRNA and TGFß3 is crucial for secondary palate development.
Collapse
Affiliation(s)
- Partha Mukhopadhyay
- Division of Craniofacial Development and Anomalies, Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY 40202, United States
| | - Irina Smolenkova
- Division of Craniofacial Development and Anomalies, Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY 40202, United States
| | - Dennis Warner
- Division of Craniofacial Development and Anomalies, Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY 40202, United States
| | - Michele M Pisano
- Division of Craniofacial Development and Anomalies, Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY 40202, United States
| | - Robert M Greene
- Division of Craniofacial Development and Anomalies, Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY 40202, United States
| |
Collapse
|
29
|
Wen J, He T, Qi F, Chen H. MiR-206-3p alleviates chronic constriction injury-induced neuropathic pain through targeting HDAC4. Exp Anim 2018; 68:213-220. [PMID: 30587671 PMCID: PMC6511522 DOI: 10.1538/expanim.18-0091] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
It was identified that microRNAs were involved in the regulation of chronic neuropathic pain. However, the role of miR-206-3p in neuropathic pain was still unclear. In the current study, the role of miR-206-3p, a type of mature miR-206, in neuropathic pain was investigated. The potential mechanisms were also explored. We found that the expression of miR-206-3p decreased in the dorsal root ganglion (DRG) of chronic constriction sciatic nerve injury (CCI) rats, whereas the While histone deacetylase 4 (HDAC4) level increased. Further exploration showed that administration of a miR-206-3p mimic alleviated neuropathic pain and reduced the level of HDAC4, a predicted target of miR-206-3p. Overexpression of HDAC4 attenuated the effects of miR-206-3p on neuropathic pain. Our data revealed a miR-206-3p-HDAC4 signal that played a potentially important role in CCI-induced neuropathic pain.
Collapse
Affiliation(s)
- Jing Wen
- Department of Histology and Embryology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, People's Republic of China
| | - Tao He
- Department of Histology and Embryology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, People's Republic of China.,Nanchang Joint Programme, Queen Mary University of London, London E1 4NS, UK
| | - Fangfang Qi
- Department of Histology and Embryology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, People's Republic of China.,Nanchang Joint Programme, Queen Mary University of London, London E1 4NS, UK
| | - Hongping Chen
- Department of Histology and Embryology, Medical College, Nanchang University, 461 Bayi Road, Nanchang 330006, People's Republic of China.,Jiangxi Province Key Laboratory of Tumor Pathogen's and Molecular Pathology, Nanchang 330006, People's Republic of China
| |
Collapse
|
30
|
Worton LE, Gardiner EM, Kwon RY, Downey LM, Ausk BJ, Bain SD, Gross TS. Botulinum toxin A-induced muscle paralysis stimulates Hdac4 and differential miRNA expression. PLoS One 2018; 13:e0207354. [PMID: 30427927 PMCID: PMC6235354 DOI: 10.1371/journal.pone.0207354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022] Open
Abstract
At sufficient dose, intramuscular injection of Botulinum toxin A causes muscle wasting that is physiologically consistent with surgical denervation and other types of neuromuscular dysfunction. The aim of this study was to clarify early molecular and micro-RNA alterations in skeletal muscle following Botulinum toxin A-induced muscle paralysis. Quadriceps were analyzed for changes in expression of micro- and messenger RNA and protein levels after a single injection of 0.4, 2 or 4U Botulinum toxin A (/100g body weight). After injection with 2.0U Botulinum toxin A, quadriceps exhibited significant reduction in muscle weight and increased levels of ubiquitin ligase proteins at 7, 14 and 28 days. Muscle miR-1 and miR-133a/b levels were decreased at these time points, whereas a dose-responsive increase in miR-206 expression at day 14 was observed. Expression of the miR-133a/b target genes RhoA, Tgfb1 and Ctfg, and the miR-1/206 target genes Igf-1 and Hdac4, were upregulated at 28 days after Botulinum toxin A injection. Increased levels of Hdac4 protein were observed after injection, consistent with anticipated expression changes in direct and indirect Hdac4 target genes, such as Myog. Our results suggest Botulinum toxin A-induced denervation of muscle shares molecular characteristics with surgical denervation and other types of neuromuscular dysfunction, and implicates miR-133/Tgf-β1/Ctfg and miR-1/Hdac4/Myog signaling during the resultant muscle atrophy.
Collapse
Affiliation(s)
- Leah E. Worton
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA
- * E-mail:
| | - Edith M. Gardiner
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA
| | - Ronald Y. Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA
| | - Leah M. Downey
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA
| | - Brandon J. Ausk
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA
| | - Steven D. Bain
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA
| | - Ted S. Gross
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA
| |
Collapse
|
31
|
Bernardo BC, Gregorevic P, Ritchie RH, McMullen JR. Generation of MicroRNA-34 Sponges and Tough Decoys for the Heart: Developments and Challenges. Front Pharmacol 2018; 9:1090. [PMID: 30298011 PMCID: PMC6160554 DOI: 10.3389/fphar.2018.01090] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) is a debilitating and deadly chronic disease, with almost 50% of patients with HF dying within 5 years of diagnosis. With limited effective therapies to treat or cure HF, new therapies are greatly needed. microRNAs (miRNAs) are small non-coding RNA molecules that are powerful regulators of gene expression and play a key role in almost every biological process. Disruptions in miRNA gene expression has been functionally linked to numerous diseases, including cardiovascular disease. Molecular tools for manipulating miRNA activity have been developed, and there is evidence from preclinical studies demonstrating the potential of miRNAs to be therapeutic targets for cardiovascular disease. For clinical application, miRNA sponges and tough decoys have been developed for more stable suppression and targeted delivery of the miRNA of choice. The aim of this study was to generate miRNA sponges and tough decoys to target miR-34 in the mouse heart. We present data to show that using both approaches we were unable to get significant knockdown of miR-34 or regulate miR-34 target genes in the heart in vivo. We also review recent applications of this method in the heart and discuss further considerations for optimisation in construct design and testing, and the obstacles to be overcome before they enter the clinic.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Physiology, Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Diabetes, Central Clinical School, Monash University, Clayton, VIC, Australia.,Department of Medicine, Monash University, Clayton, VIC, Australia.,Department of Physiology, Monash University, Clayton, VIC, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
32
|
Chodkowska KA, Ciecierska A, Majchrzak K, Ostaszewski P, Sadkowski T. Effect of β-hydroxy-β-methylbutyrate on miRNA expression in differentiating equine satellite cells exposed to hydrogen peroxide. GENES AND NUTRITION 2018; 13:10. [PMID: 29662554 PMCID: PMC5892041 DOI: 10.1186/s12263-018-0598-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 03/14/2018] [Indexed: 12/25/2022]
Abstract
Background Skeletal muscle injury activates satellite cells to initiate processes of proliferation, differentiation, and hypertrophy in order to regenerate muscle fibers. The number of microRNAs and their target genes are engaged in satellite cell activation. β-Hydroxy-β-methylbutyrate (HMB) is known to prevent exercise-induced muscle damage. The purpose of this study was to evaluate the effect of HMB on miRNA and relevant target gene expression in differentiating equine satellite cells exposed to H2O2. We hypothesized that HMB may regulate satellite cell activity, proliferation, and differentiation, hence attenuate the pathological processes induced during an in vitro model of H2O2-related injury by changing the expression of miRNAs. Methods Equine satellite cells (ESC) were isolated from the samples of skeletal muscle collected from young horses. ESC were treated with HMB (24 h) and then exposed to H2O2 (1 h). For the microRNA and gene expression assessment microarrays, technique was used. Identified miRNAs and genes were validated using real-time qPCR. Cell viability, oxidative stress, and cell damage were measured using colorimetric method and flow cytometry. Results Analysis of miRNA and gene profile in differentiating ESC pre-incubated with HMB and then exposed to H2O2 revealed difference in the expression of 27 miRNAs and 4740 genes, of which 344 were potential target genes for identified miRNAs. Special attention was focused on differentially expressed miRNAs and their target genes involved in processes related to skeletal muscle injury. Western blot analysis showed protein protection in HMB-pre-treated group compared to control. The viability test confirmed that HMB enhanced cell survival after the hydrogen peroxide exposition. Conclusions Our results suggest that ESC pre-incubated with HMB and exposed to H2O2 could affect expression on miRNA levels responsible for skeletal muscle development, cell proliferation and differentiation, and activation of tissue repair after injury. Enrichment analyses for targeted genes revealed that a large group of genes was associated with the regulation of signaling pathways crucial for muscle tissue development, protein metabolism, muscle injury, and regeneration, as well as with oxidative stress response.
Collapse
Affiliation(s)
- Karolina A Chodkowska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Anna Ciecierska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Kinga Majchrzak
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Piotr Ostaszewski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Tomasz Sadkowski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| |
Collapse
|
33
|
Lack of Heme Oxygenase-1 Induces Inflammatory Reaction and Proliferation of Muscle Satellite Cells after Cardiotoxin-Induced Skeletal Muscle Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:491-506. [PMID: 29169990 DOI: 10.1016/j.ajpath.2017.10.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/12/2017] [Accepted: 10/23/2017] [Indexed: 12/29/2022]
Abstract
Heme oxygenase-1 (HO-1, Hmox1) regulates viability, proliferation, and differentiation of many cell types; hence, it may affect regeneration of injured skeletal muscle. Here, we injected cardiotoxin into gastrocnemius muscle of Hmox1+/+ and Hmox1-/- animals and analyzed cellular response after muscle injury, focusing on muscle satellite cells (SCs), inflammatory reaction, fibrosis, and formation of new blood vessels. HO-1 is strongly induced after muscle injury, being expressed mostly in the infiltrating leukocytes (CD45+ cells), including macrophages (F4/80+ cells). Lack of HO-1 augments skeletal muscle injury, evidenced by increased creatinine kinase and lactate dehydrogenase, as well as expression of monocyte chemoattractant protein-1, IL-6, IL-1β, and insulin-like growth factor-1. This, together with disturbed proportion of M1/M2 macrophages, accompanied by enhanced formation of arterioles, may be responsible for shift of Hmox1-/- myofiber size distribution toward larger one. Importantly, HO-1-deficient SCs are prone to activation and have higher proliferation on injury. This effect can be partially mimicked by stimulation of Hmox1+/+ SCs with monocyte chemoattractant protein-1, IL-6, IL-1β, and is associated with increased MyoD expression, suggesting that Hmox1-/- SCs are shifted toward more differentiated myogenic population. However, multiple rounds of degeneration/regeneration in conditions of HO-1 deficiency may lead to exhaustion of SC pool, and the number of SCs is decreased in old Hmox1-/- mice. In summary, HO-1 modulates muscle repair mechanisms preventing its uncontrolled acceleration.
Collapse
|
34
|
Simon L, Ford SM, Song K, Berner P, Vande Stouwe C, Nelson S, Bagby GJ, Molina PE. Decreased myoblast differentiation in chronic binge alcohol-administered simian immunodeficiency virus-infected male macaques: role of decreased miR-206. Am J Physiol Regul Integr Comp Physiol 2017; 313:R240-R250. [PMID: 28637658 PMCID: PMC5625276 DOI: 10.1152/ajpregu.00146.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/07/2017] [Accepted: 06/21/2017] [Indexed: 01/05/2023]
Abstract
Skeletal muscle stem cells play a critical role in regeneration of myofibers. We previously demonstrated that chronic binge alcohol (CBA) markedly attenuates myoblast differentiation potential and myogenic gene expression. Muscle-specific microRNAs (miRs) are implicated in regulation of myogenic genes. The aim of this study was to determine whether myoblasts isolated from asymptomatic CBA-administered simian immunodeficiency virus (SIV)-infected macaques treated with antiretroviral therapy (ART) showed similar impairments and, if so, to elucidate potential underlying mechanisms. Myoblasts were isolated from muscle at 11 mo after SIV infection from CBA/SIV macaques and from time-matched sucrose (SUC)-treated SIV-infected (SUC/SIV) animals and age-matched controls. Myoblast differentiation and myogenic gene expression were significantly decreased in myoblasts from SUC/SIV and CBA/SIV animals compared with controls. SIV and CBA decreased muscle-specific miR-206 in plasma and muscle and SIV decreased miR-206 expression in myoblasts, with no statistically significant changes in other muscle-specific miRs. These findings were associated with a significant increase in histone deacetylase 4 (HDAC4) and decrease in myogenic enhancer factor 2C (MEF2C) expression in CBA/SIV muscle. Transfection with miR-206 inhibitor decreased myotube differentiation, increased expression of HDAC4, and decreased MEF2C, suggesting a critical role of miR-206 in myogenesis. Moreover, HDAC4 was confirmed to be a direct miR-206 target. These results support a mechanistic role for decreased miR-206 in suppression of myoblast differentiation resulting from chronic alcohol and SIV infection. The parallel changes in skeletal muscle and circulating levels of miR-206 warrant studies to establish the possible use of plasma miR-206 as an indicator of impaired muscle function.
Collapse
Affiliation(s)
- L Simon
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana;
- Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| | - S M Ford
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - K Song
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - P Berner
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - C Vande Stouwe
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - S Nelson
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - G J Bagby
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - P E Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
- Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| |
Collapse
|
35
|
D'Souza RF, Bjørnsen T, Zeng N, Aasen KMM, Raastad T, Cameron-Smith D, Mitchell CJ. MicroRNAs in Muscle: Characterizing the Powerlifter Phenotype. Front Physiol 2017. [PMID: 28638346 PMCID: PMC5461344 DOI: 10.3389/fphys.2017.00383] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Powerlifters are the epitome of muscular adaptation and are able to generate extreme forces. The molecular mechanisms underpinning the significant capacity for force generation and hypertrophy are not fully elucidated. MicroRNAs (miRs) are short non-coding RNA sequences that control gene expression via promotion of transcript breakdown and/or translational inhibition. Differences in basal miR expression may partially account for phenotypic differences in muscle mass and function between powerlifters and untrained age-matched controls. Muscle biopsies were obtained from m. vastus lateralis of 15 national level powerlifters (25.1 ± 5.8 years) and 13 untrained controls (24.1 ± 2.0 years). The powerlifters were stronger than the controls (isokinetic knee extension at 60°/s: 307.8 ± 51.6 Nm vs. 211.9 ± 41.9 Nm, respectively P < 0.001), and also had larger muscle fibers (type I CSA 9,122 ± 1,238 vs. 4,511 ± 798 μm2p < 0.001 and type II CSA 11,100 ± 1,656 vs. 5,468 ± 1,477 μm2p < 0.001). Of the 17 miRs species analyzed, 12 were differently expressed (p < 0.05) between groups with 7 being more abundant in powerlifters and five having lower expression. Established transcriptionally regulated miR downstream gene targets involved in muscle mass regulation, including myostatin and MyoD, were also differentially expressed between groups. Correlation analysis demonstrates the abundance of eight miRs was correlated to phenotype including peak strength, fiber size, satellite cell abundance, and fiber type regardless of grouping. The unique miR expression profiles between groups allow for categorization of individuals as either powerlifter or healthy controls based on a five miR signature (miR-126, -23b, -16, -23a, -15a) with considerable accuracy (100%). Thus, this unique miR expression may be important to the characterization of the powerlifter phenotype.
Collapse
Affiliation(s)
| | - Thomas Bjørnsen
- Department of Public Health, Sport and Nutrition, Faculty of Health and Sport Sciences, University of AgderKristiansand, Norway
| | - Nina Zeng
- Liggins Institute, University of AucklandAuckland, New Zealand
| | | | - Truls Raastad
- Department of Physical Performance, Norwegian School of Sport SciencesOslo, Norway
| | | | | |
Collapse
|
36
|
McCormick R, Goljanek-Whysall K. MicroRNA Dysregulation in Aging and Pathologies of the Skeletal Muscle. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:265-308. [PMID: 28838540 DOI: 10.1016/bs.ircmb.2017.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Skeletal muscle is one of the biggest organs of the body with important mechanistic and metabolic functions. Muscle homeostasis is controlled by environmental, genetic, and epigenetic factors. Indeed, MiRNAs, small noncoding RNAs robust regulators of gene expression, have and have been shown to regulate muscle homeostasis on several levels: through controlling myogenesis, muscle growth (hypertrophy) and atrophy, as well as interactions of muscle with other tissues. Given the large number of MiRNA target genes and the important role of MiRNAs in most physiological processes and various diseases, MiRNAs may have an enormous potential as therapeutic targets against numerous disorders, including pathologies of muscle. The purpose of this review is to present the current knowledge of the role of MiRNAs in skeletal muscle homeostasis and pathologies and the potential of MiRNAs as therapeutics for skeletal muscle wasting, with particular focus on the age- and disease-related loss of muscle mass and function.
Collapse
Affiliation(s)
- Rachel McCormick
- Musculoskeletal Biology II, Centre for Integrated Research into Musculoskeletal Aging, Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.
| | - Katarzyna Goljanek-Whysall
- Musculoskeletal Biology II, Centre for Integrated Research into Musculoskeletal Aging, Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
37
|
Amirouche A, Jahnke VE, Lunde JA, Koulmann N, Freyssenet DG, Jasmin BJ. Muscle-specific microRNA-206 targets multiple components in dystrophic skeletal muscle representing beneficial adaptations. Am J Physiol Cell Physiol 2017; 312:C209-C221. [DOI: 10.1152/ajpcell.00185.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 12/06/2016] [Accepted: 12/21/2016] [Indexed: 01/31/2023]
Abstract
Over the last several years, converging lines of evidence have indicated that miR-206 plays a pivotal role in promoting muscle differentiation and regeneration, thereby potentially impacting positively on the progression of neuromuscular disorders, including Duchenne muscular dystrophy (DMD). Despite several studies showing the regulatory function of miR-206 on target mRNAs in skeletal muscle cells, the effects of overexpression of miR-206 in dystrophic muscles remain to be established. Here, we found that miR-206 overexpression in mdx mouse muscles simultaneously targets multiple mRNAs and proteins implicated in satellite cell differentiation, muscle regeneration, and at the neuromuscular junction. Overexpression of miR-206 also increased the levels of several muscle-specific mRNAs/proteins, while enhancing utrophin A expression at the sarcolemma. Finally, we also observed that the increased expression of miR-206 in dystrophin-deficient mouse muscle decreased the production of proinflammatory cytokines and infiltration of macrophages. Taken together, our results show that miR-206 acts as a pleiotropic regulator that targets multiple key mRNAs and proteins expected to provide beneficial adaptations in dystrophic muscle, thus highlighting its therapeutic potential for DMD.
Collapse
Affiliation(s)
- Adel Amirouche
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Vanessa E. Jahnke
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
| | - John A. Lunde
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Nathalie Koulmann
- Institut de Recherche Biomédicale des Armées, Département Environnements Opérationnels, Bretigny-sur-Orge, France
| | - Damien G. Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
| | - Bernard J. Jasmin
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| |
Collapse
|
38
|
Graham ZA, De Gasperi R, Bauman WA, Cardozo CP. Recombinant myostatin reduces highly expressed microRNAs in differentiating C2C12 cells. Biochem Biophys Rep 2017; 9:273-280. [PMID: 28691106 PMCID: PMC5500170 DOI: 10.1016/j.bbrep.2017.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/20/2022] Open
Abstract
Myostatin is small glycopeptide that is produced and secreted by skeletal muscle. It is a potent negative regulator of muscle growth that has been associated with conditions of frailty. In C2C12 cells, myostatin limits cell differentiation. Myostatin acts through activin receptor IIB, activin receptor-like kinase (ALK) and Smad transcription factors. microRNAs (miRNA) are short, 22 base pair nucleotides that bind to the 3' UTR of target mRNA to repress translation or reduce mRNA stability. In the present study, expression in differentiating C2C12 cells of the myomiRs miR-1 and 133a were down-regulated following treatment with 1 μg of recombinant myostatin at 1 d post-induction of differentiation while all myomiRs (miR-1, 133a/b and 206) were upregulated by SB431542, a potent ALK4/5/7 inhibitor which reduces Smad2 signaling, at 1 d and all, with the exception of miR-206, were upregulated by SB431542 at 3 d. The expression of the muscle-enriched miR-486 was greater following treatment with SB431542 but not altered by myostatin. Other highly expressed miRNAs in skeletal muscle, miR-23a/b and 145, were altered only at 1 d post-induction of differentiation. miR-27b responded differently to treatments at 1 d, where it was upregulated, as compared to 3 d, where it was downregulated. Neither myostatin nor SB431542 altered cell size or cell morphology. The data indicate that myostatin represses myomiR expression in differentiating C2C12 cells and that inhibition of Smad signaling with SB431542 can result in large changes in highly expressed miRNAs in differentiating myoblasts.
Collapse
Affiliation(s)
- Zachary A. Graham
- National Center for the Medical Consequences of Spinal Cord Injury, Bronx, NY, USA
- Departments of Medicine, Icahn School of Medicine at Mt. Sinai, NY, USA
| | - Rita De Gasperi
- National Center for the Medical Consequences of Spinal Cord Injury, Bronx, NY, USA
- Departments of Psychiatry, Icahn School of Medicine at Mt. Sinai, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mt. Sinai, NY, USA
| | - William A. Bauman
- National Center for the Medical Consequences of Spinal Cord Injury, Bronx, NY, USA
- Medical Service, James J. Peters VAMC, Bronx, NY, USA
- Departments of Medicine, Icahn School of Medicine at Mt. Sinai, NY, USA
- Departments of Rehabilitation Medicine, Icahn School of Medicine at Mt. Sinai, NY, USA
| | - Christopher P. Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, Bronx, NY, USA
- Medical Service, James J. Peters VAMC, Bronx, NY, USA
- Departments of Medicine, Icahn School of Medicine at Mt. Sinai, NY, USA
- Departments of Rehabilitation Medicine, Icahn School of Medicine at Mt. Sinai, NY, USA
- Departments of Pharmacologic Science and the Icahn School of Medicine at Mt. Sinai, NY, USA
| |
Collapse
|
39
|
Exercise Training and Epigenetic Regulation: Multilevel Modification and Regulation of Gene Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:281-322. [PMID: 29098627 DOI: 10.1007/978-981-10-4304-8_16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Exercise training elicits acute and adaptive long term changes in human physiology that mediate the improvement of performance and health state. The responses are integrative and orchestrated by several mechanisms, as gene expression. Gene expression is essential to construct the adaptation of the biological system to exercise training, since there are molecular processes mediating oxidative and non-oxidative metabolism, angiogenesis, cardiac and skeletal myofiber hypertrophy, and other processes that leads to a greater physiological status. Epigenetic is the field that studies about gene expression changes heritable by meiosis and mitosis, by changes in chromatin and DNA conformation, but not in DNA sequence, that studies the regulation on gene expression that is independent of genotype. The field approaches mechanisms of DNA and chromatin conformational changes that inhibit or increase gene expression and determine tissue specific pattern. The three major studied epigenetic mechanisms are DNA methylation, Histone modification, and regulation of noncoding RNA-associated genes. This review elucidates these mechanisms, focusing on the relationship between them and their relationship with exercise training, physical performance and the enhancement of health status. On this chapter, we clarified the relationship of epigenetic modulations and their intimal relationship with acute and chronic effect of exercise training, concentrating our effort on skeletal muscle, heart and vascular responses, that are the most responsive systems against to exercise training and play crucial role on physical performance and improvement of health state.
Collapse
|
40
|
Histone Deacetylase Inhibitor Phenylbutyrate Exaggerates Heart Failure in Pressure Overloaded Mice independently of HDAC inhibition. Sci Rep 2016; 6:34036. [PMID: 27667442 PMCID: PMC5036044 DOI: 10.1038/srep34036] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 09/05/2016] [Indexed: 12/18/2022] Open
Abstract
4-Sodium phenylbutyrate (PBA) has been reported to inhibit endoplasmic reticulum stress and histone deacetylation (HDAC), both of which are novel therapeutic targets for cardiac hypertrophy and heart failure. However, it is unclear whether PBA can improve heart function. Here, we tested the effects of PBA and some other HDAC inhibitors on cardiac dysfunction induced by pressure overload. Transverse aortic constriction (TAC) was performed on male C57BL/6 mice. PBA treatment (100 mg/kg, 6 weeks) unexpectedly led to a higher mortality, exacerbated cardiac remodelling and dysfunction. Similar results were noted in TAC mice treated with butyrate sodium (BS), a PBA analogue. In contrast, other HDAC inhibitors, valproic acid (VAL) and trichostatin A (TSA), improved the survival. All four HDAC inhibitors induced histone H3 acetylation and inhibited HDAC total activity. An individual HDAC activity assay showed that rather than class IIa members (HDAC4 and 7), PBA and BS predominantly inhibited class I members (HDAC2 and 8), whereas VAL and TSA inhibited all of them. These findings indicate that PBA and BS accelerate cardiac hypertrophy and dysfunction, whereas VAL and TSA have opposing effects.
Collapse
|
41
|
Wei X, Li H, Zhang B, Li C, Dong D, Lan X, Huang Y, Bai Y, Lin F, Zhao X, Chen H. miR-378a-3p promotes differentiation and inhibits proliferation of myoblasts by targeting HDAC4 in skeletal muscle development. RNA Biol 2016; 13:1300-1309. [PMID: 27661135 DOI: 10.1080/15476286.2016.1239008] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Muscle development, or myogenesis, is a highly regulated, complex process. A subset of microRNAs (miRNAs) have been identified as critical regulators of myogenesis. Recently, miR-378a was found to be involved in myogenesis, but the mechanism of how miR-378a regulates the proliferation and differentiation of myoblasts has not been determined. We found that miR-378a-3p expression in muscle was significantly higher than in other tissues, suggesting an important effect on muscle development. Overexpression of miR-378a-3p increased the expression of MyoD and MHC in C2C12 myoblasts both at the level of mRNA and protein, confirming that miR-378a-3p promoted muscle cell differentiation. The forced expression of miR-378a-3p promoted apoptosis of C2C12 cells as evidenced by CCK-8 assay and Annexin V-FITC/PI staining results. Through TargetScan, histone acetylation enzyme 4 (HDAC4) was identified as a potential target of miR-378a-3p. We confirmed targeting of HDAC4 by miR-378a-3p using a dual luciferase assay and western blotting. Our RNAi analysis results also showed that HDAC4 significantly promoted differentiation of C2C12 cells and inhibited cell survival through Bcl-2. Therefore, we conclude that miR-378a-3p regulates skeletal muscle growth and promotes the differentiation of myoblasts through the post-transcriptional down-regulation of HDAC4.
Collapse
Affiliation(s)
- Xuefeng Wei
- a Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , China
| | - Hui Li
- a Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , China
| | - Bowen Zhang
- a Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , China
| | - Caixia Li
- a Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , China
| | - Dong Dong
- a Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , China
| | - Xianyong Lan
- a Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , China
| | - Yongzhen Huang
- a Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , China
| | - Yueyu Bai
- b Animal Health Supervision in Henan Province , Zhengzhou , Henan , China
| | - Fengpeng Lin
- c Bureau of Animal Husbandry of Biyang County , Biyang , Henan , China
| | - Xue Zhao
- d Bureau of Animal Husbandry of Suibin County , Suibin , Heilongjiang , China
| | - Hong Chen
- a Shaanxi Key Laboratory of Agricultural Molecular Biology, College of Animal Science and Technology, Northwest A&F University , Yangling , Shaanxi , China
| |
Collapse
|
42
|
Ciesla M, Marona P, Kozakowska M, Jez M, Seczynska M, Loboda A, Bukowska-Strakova K, Szade A, Walawender M, Kusior M, Stepniewski J, Szade K, Krist B, Yagensky O, Urbanik A, Kazanowska B, Dulak J, Jozkowicz A. Heme Oxygenase-1 Controls an HDAC4-miR-206 Pathway of Oxidative Stress in Rhabdomyosarcoma. Cancer Res 2016; 76:5707-5718. [PMID: 27488535 DOI: 10.1158/0008-5472.can-15-1883] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 07/16/2016] [Indexed: 11/16/2022]
Abstract
Rhabdomyosarcoma (RMS) is an aggressive soft tissue cancer characterized by disturbed myogenic differentiation. Here we report a role for the oxidative stress response factor HO-1 in progression of RMS. We found that HO-1 was elevated and its effector target miR-206 decreased in RMS cell lines and clinical primary tumors of the more aggressive alveolar phenotype (aRMS). In embryonal RMS (eRMS), HO-1 expression was induced by Pax3/7-FoxO1, an aRMS hallmark oncogene, followed by a drop in miR-206 levels. Inhibition of HO-1 by tin protoporphyrin (SnPP) or siRNA downregulated Pax3/7-FoxO1 target genes and induced a myogenic program in RMS. These effects were not mediated by altered myoD expression; instead, cells with elevated HO-1 produced less reactive oxygen species, resulting in nuclear localization of HDAC4 and miR-206 repression. HO-1 inhibition by SnPP reduced growth and vascularization of RMS tumors in vivo accompanied by induction of miR-206. Effects of SnPP on miR-206 expression and RMS tumor growth were mimicked by pharmacologic inhibition of HDAC. Thus, HO-1 inhibition activates an miR-206-dependent myogenic program in RMS, offering a novel therapeutic strategy for treatment of this malignancy. Cancer Res; 76(19); 5707-18. ©2016 AACR.
Collapse
Affiliation(s)
- Maciej Ciesla
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Paulina Marona
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Mateusz Jez
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Marta Seczynska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Agata Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Walawender
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Kusior
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jacek Stepniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Bart Krist
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Oleksandr Yagensky
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Aleksandra Urbanik
- Department of Oncology, Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Bernarda Kazanowska
- Department of Oncology, Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland. International Associated Laboratory, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
43
|
Noncoding RNAs in the regulation of skeletal muscle biology in health and disease. J Mol Med (Berl) 2016; 94:853-66. [PMID: 27377406 DOI: 10.1007/s00109-016-1443-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/11/2016] [Accepted: 06/20/2016] [Indexed: 12/22/2022]
Abstract
Skeletal muscle is composed of multinucleated myofibers that arise from the fusion of myoblasts during development. Skeletal muscle is essential for various body functions such as maintaining posture, locomotion, breathing, and metabolism. Skeletal muscle undergoes remarkable adaptations in response to environmental stimuli leading to atrophy or hypertrophy. Moreover, degeneration of skeletal muscle is a common feature in a number of muscular disorders including muscular dystrophy. Emerging evidence suggests that noncoding RNAs, such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are critical for skeletal muscle physiology. Several miRNAs and lncRNAs have now been found to control skeletal muscle development and regeneration. Noncoding RNAs also play an important role in the regulation of skeletal muscle mass in adults. Furthermore, aberrant expression of miRNAs and lncRNAs has been observed in several muscular disorders. In this article, we discuss the mechanisms of action of miRNAs and lncRNAs in skeletal muscle formation, growth, regeneration, and disease. We further highlight potential therapeutic strategies for utilizing noncoding RNAs to improve skeletal muscle function.
Collapse
|
44
|
Sun R, Zhang S, Hu W, Lu X, Lou N, Yang Z, Chen S, Zhang X, Yang H. Valproic acid attenuates skeletal muscle wasting by inhibiting C/EBPβ-regulated atrogin1 expression in cancer cachexia. Am J Physiol Cell Physiol 2016; 311:C101-15. [DOI: 10.1152/ajpcell.00344.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/13/2016] [Indexed: 12/15/2022]
Abstract
Muscle wasting is the hallmark of cancer cachexia and is associated with poor quality of life and increased mortality. Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, has important biological effects in the treatment of muscular dystrophy. To verify whether VPA could ameliorate muscle wasting induced by cancer cachexia, we explored the role of VPA in two cancer cachectic mouse models [induced by colon-26 (C26) adenocarcinoma or Lewis lung carcinoma (LLC)] and atrophied C2C12 myotubes [induced by C26 cell conditioned medium (CCM) or LLC cell conditioned medium (LCM)]. Our data demonstrated that treatment with VPA increased the mass and cross-sectional area of skeletal muscles in tumor-bearing mice. Furthermore, treatment with VPA also increased the diameter of myotubes cultured in conditioned medium. The skeletal muscles in cachectic mice or atrophied myotubes treated with VPA exhibited reduced levels of CCAAT/enhancer binding protein beta (C/EBPβ), resulting in atrogin1 downregulation and the eventual alleviation of muscle wasting and myotube atrophy. Moreover, atrogin1 promoter activity in myotubes was stimulated by CCM via activating the C/EBPβ-responsive cis-element and subsequently inhibited by VPA. In contrast to the effect of VPA on the levels of C/EBPβ, the levels of inactivating forkhead box O3 (FoxO3a) were unaffected. In summary, VPA attenuated muscle wasting and myotube atrophy and reduced C/EBPβ binding to atrogin1 promoter locus in the myotubes. Our discoveries indicate that HDAC inhibition by VPA might be a promising new approach for the preservation of skeletal muscle in cancer cachexia.
Collapse
Affiliation(s)
- Rulin Sun
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Santao Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Wenjun Hu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Xing Lu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Ning Lou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China; and
| | - Zhende Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Shaoyong Chen
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China; and
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| |
Collapse
|
45
|
Xie M, Dart DA, Owen S, Wen X, Ji J, Jiang W. Insights into roles of the miR-1, -133 and -206 family in gastric cancer (Review). Oncol Rep 2016; 36:1191-8. [PMID: 27349337 DOI: 10.3892/or.2016.4908] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/27/2016] [Indexed: 11/06/2022] Open
Abstract
Gastric cancer (GC) remains the third most common cause of cancer deaths worldwide and carries a high rate of metastatic risk contributing to the main cause of treatment failure. An accumulation of data has resulted in a better understanding of the molecular network of GC, however, gaps still exist between the unique bio-resources and clinical application. MicroRNAs are an important part of non-coding RNAs and behave as major regulators of tumour biology, alongside their well-known roles as intrinsic factors of gene expression in cellular processes, via their post-transcriptional regulation of components of signalling pathways in a coordinated manner. Deregulation of the miR-1, -133 and -206 family plays a key role in tumorigenesis, progression, invasion and metastasis. This review aims to provide a summary of recent findings on the miR-1, -133 and -206 family in GC and how this knowledge might be exploited for the development of future miRNA-based therapies for the treatment of GC.
Collapse
Affiliation(s)
- Meng Xie
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Haidian, Beijing 100142, P.R. China
| | - Dafydd Alwyn Dart
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Sioned Owen
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Xianzi Wen
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Haidian, Beijing 100142, P.R. China
| | - Jiafu Ji
- Department of Gastrointestinal Translational Research, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Haidian, Beijing 100142, P.R. China
| | - Wenguo Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| |
Collapse
|
46
|
Targeting oncomiRNAs and mimicking tumor suppressor miRNAs: Νew trends in the development of miRNA therapeutic strategies in oncology (Review). Int J Oncol 2016; 49:5-32. [PMID: 27175518 PMCID: PMC4902075 DOI: 10.3892/ijo.2016.3503] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 04/29/2016] [Indexed: 12/16/2022] Open
Abstract
MicroRNA (miRNA or miR) therapeutics in cancer are based on targeting or mimicking miRNAs involved in cancer onset, progression, angiogenesis, epithelial-mesenchymal transition and metastasis. Several studies conclusively have demonstrated that miRNAs are deeply involved in tumor onset and progression, either behaving as tumor-promoting miRNAs (oncomiRNAs and metastamiRNAs) or as tumor suppressor miRNAs. This review focuses on the most promising examples potentially leading to the development of anticancer, miRNA-based therapeutic protocols. The inhibition of miRNA activity can be readily achieved by the use of miRNA inhibitors and oligomers, including RNA, DNA and DNA analogues (miRNA antisense therapy), small molecule inhibitors, miRNA sponges or through miRNA masking. On the contrary, the enhancement of miRNA function (miRNA replacement therapy) can be achieved by the use of modified miRNA mimetics, such as plasmid or lentiviral vectors carrying miRNA sequences. Combination strategies have been recently developed based on the observation that i) the combined administration of different antagomiR molecules induces greater antitumor effects and ii) some anti-miR molecules can sensitize drug-resistant tumor cell lines to therapeutic drugs. In this review, we discuss two additional issues: i) the combination of miRNA replacement therapy with drug administration and ii) the combination of antagomiR and miRNA replacement therapy. One of the solid results emerging from different independent studies is that miRNA replacement therapy can enhance the antitumor effects of the antitumor drugs. The second important conclusion of the reviewed studies is that the combination of anti-miRNA and miRNA replacement strategies may lead to excellent results, in terms of antitumor effects.
Collapse
|
47
|
Lim HJ, Yang JL. Regulatory roles and therapeutic potential of microRNA in sarcoma. Crit Rev Oncol Hematol 2016; 97:118-30. [DOI: 10.1016/j.critrevonc.2015.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 07/15/2015] [Accepted: 08/04/2015] [Indexed: 02/01/2023] Open
|
48
|
Ono K. Functions of microRNA-33a/b and microRNA therapeutics. J Cardiol 2016; 67:28-33. [DOI: 10.1016/j.jjcc.2015.10.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 10/26/2015] [Indexed: 12/18/2022]
|
49
|
Brown DM, Goljanek-Whysall K. microRNAs: Modulators of the underlying pathophysiology of sarcopenia? Ageing Res Rev 2015; 24:263-73. [PMID: 26342566 DOI: 10.1016/j.arr.2015.08.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/15/2015] [Accepted: 08/31/2015] [Indexed: 12/25/2022]
Abstract
Skeletal muscle homeostasis depends on an intricate balance between muscle hypertrophy, atrophy and regeneration. As we age, maintenance of muscle homeostasis is perturbed, resulting in a loss of muscle mass and function, termed sarcopenia. Individuals with sarcopenia exhibit impaired balance, increased falls (leading to subsequent injury) and an overall decline in quality of life. The mechanisms mediating sarcopenia are still not fully understood but clarity in our understanding of the precise pathophysiological changes occurring during skeletal muscle ageing has improved dramatically. Advances in transcriptomics has highlighted significant deregulation in skeletal muscle gene expression with ageing, suggesting epigenetic alterations may play a crucial and potentially causative role in the skeletal muscle ageing process. microRNAs (miRNAs, miRs), novel regulators of gene expression, can modulate many processes in skeletal muscle, including myogenesis, tissue regeneration and cellular programming. Expression of numerous evolutionary conserved miRNAs is disrupted in skeletal muscle with age. Given that a single miRNA can simultaneously affect the functionality of multiple signaling pathways, miRNAs are potent modulators of pathophysiological changes. miRNA-based interventions provide a promising new therapeutic strategy against alterations in muscle homeostasis. The aim of this review is two-fold; firstly to outline the latest understanding of the pathophysiological alterations impacting the deregulation of skeletal muscle mass and function with ageing, and secondly, to highlight the mounting evidence for a role of miRNAs in modulating muscle mass, and the need to explore their specific role in sarcopenia.
Collapse
Affiliation(s)
- David M Brown
- Medical Research Council-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.
| | - Katarzyna Goljanek-Whysall
- Medical Research Council-Arthritis Research UK Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.
| |
Collapse
|
50
|
miRNA therapeutics: a new class of drugs with potential therapeutic applications in the heart. Future Med Chem 2015; 7:1771-92. [PMID: 26399457 DOI: 10.4155/fmc.15.107] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
miRNAs are small non-coding RNAs (ncRNAs), which regulate gene expression. Here, the authors describe the contribution of miRNAs to cardiac biology and disease. They discuss various strategies for manipulating miRNA activity including antisense oligonucleotides (antimiRs, blockmiRs), mimics, miRNA sponges, Tough Decoys and miRNA mowers. They review developments in chemistries (e.g., locked nucleic acid) and modifications (sugar, 'ZEN', peptide nucleic acids) and miRNA delivery tools (viral vectors, liposomes, nanoparticles, pHLIP). They summarize potential miRNA therapeutic targets for heart disease based on preclinical studies. Finally, the authors review current progress of miRNA therapeutics in clinical development for HCV and cancer, and discuss challenges that will need to be overcome for similar therapies to enter the clinic for patients with cardiac disease.
Collapse
|