1
|
Gopal Krishnan PD, Lee WX, Goh KY, Choy SM, Turqueza LRR, Lim ZH, Tang HW. Transcriptional regulation of autophagy in skeletal muscle stem cells. Dis Model Mech 2025; 18:DMM052007. [PMID: 39925192 PMCID: PMC11849978 DOI: 10.1242/dmm.052007] [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] [Indexed: 02/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for the regenerative capabilities of skeletal muscles. MuSCs are maintained in a quiescent state, but, when activated, can undergo proliferation and differentiation into myocytes, which fuse and mature to generate muscle fibers. The maintenance of MuSC quiescence and MuSC activation are processes that are tightly regulated by autophagy, a conserved degradation system that removes unessential or dysfunctional cellular components via lysosomes. Both the upregulation and downregulation of autophagy have been linked to impaired muscle regeneration, causing myopathies such as cancer cachexia, sarcopenia and Duchenne muscular dystrophy. In this Review, we highlight the importance of autophagy in regulating MuSC activity during muscle regeneration. Additionally, we summarize recent studies that link the transcriptional dysregulation of autophagy to muscle atrophy, emphasizing the dominant roles that transcription factors play in myogenic programs. Deciphering and understanding the roles of these transcription factors in the regulation of autophagy during myogenesis could advance the development of regenerative medicine.
Collapse
Affiliation(s)
- Priya D. Gopal Krishnan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Wen Xing Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Kah Yong Goh
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Sze Mun Choy
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | | | - Zhuo Han Lim
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Hong-Wen Tang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| |
Collapse
|
2
|
Shi A, He C, Otten K, Wu G, Forouzanfar T, Wüst RCI, Jaspers RT. Reduced myotube diameter induced by combined inhibition of transforming growth factor-β type I receptors Acvr1b and Tgfbr1 is associated with enhanced β1-syntrophin expression. J Cell Physiol 2024; 239:e31418. [PMID: 39164996 PMCID: PMC11649968 DOI: 10.1002/jcp.31418] [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: 12/04/2023] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Simultaneous inhibition of transforming growth factor-β (TGF-β) type I receptors Acvr1b and Tgfbr1 signalling has been associated with excessive skeletal muscle hypertrophy in vivo. However, it remains unclear whether the increased muscle mass in vivo is a direct result of inhibition of intracellular TGF-β signalling or whether this is an indirect effect of an altered extracellular anabolic environment. Here, we tested whether individual or simultaneous knockdown of TGF-β type I receptors in C2C12 myotubes was sufficient to induce muscle hypertrophy. The expression levels of TGF-β type I receptors Acvr1b and Tgfbr1 in myotubes were knocked down individually or in combination in the absence or presence of TGF-β1 and myostatin. Knocking down either Acvr1b or Tgfbr1 did not significantly change cell phenotype. Unexpectedly, simultaneous knockdown of both receptors reduced C2C12 myotube diameter, mRNA expression levels of Hgf, Ccn2 and Mymx with or without TGF-β1 and myostatin administration. In spite of decreased phosphorylation of Smad2/3, phosphorylation of P70S6K was reduced. In addition, the gene expression level of β1-syntrophin (Sntb1), which encodes a protein associated with the dystrophin-glycoprotein complex, was increased. Parallel experiments where Sntb1 gene expression was reduced showed an increase in myotube diameter and fusion of C2C12 myoblasts. Together, these results indicate that the knockdown of both TGF-β type I receptors reduced myotube diameter. This atrophic effect was attributed to reduced protein synthesis signalling and an increased expression of β1-syntrophin. These results have implications for our fundamental understanding of how TGF-β signalling regulates skeletal muscle size.
Collapse
Affiliation(s)
- Andi Shi
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of ProsthodonticsAffiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and ReconstructionGuangzhouChina
| | - Chuqi He
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Kirsten Otten
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Gang Wu
- Department of Oral Cell BiologyAcademic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU)AmsterdamThe Netherlands
| | - Tymour Forouzanfar
- Department of Oral Cell BiologyAcademic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU)AmsterdamThe Netherlands
- Department of Oral and Maxillofacial SurgeryLeiden University Medical CenterLeidenThe Netherlands
| | - Rob C. I. Wüst
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Richard T. Jaspers
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of ProsthodonticsAffiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and ReconstructionGuangzhouChina
| |
Collapse
|
3
|
Wang X, Zhou X, Li C, Qu C, Shi Y, Li CJ, Kang X. Integrative analysis of whole genome bisulfite and transcriptome sequencing reveals the effect of sodium butyrate on DNA methylation in the differentiation of bovine skeletal muscle satellite cells. Genomics 2024; 116:110959. [PMID: 39521294 DOI: 10.1016/j.ygeno.2024.110959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Butyric acid as a short-chain fatty acid (SCFA) is one of the key microbial metabolites of ruminants. Numerous studies indicate that butyrate is crucial in muscle growth and development, and plays an important molecular regulatory role mainly by inhibiting histone deacetylation. DNA methylation, a major epigenetic modification, is involved in cell differentiation. Butyrate, in addition to its role in acetylation modifications, can alter the DNA methylation status of cells. However, the impact of butyrate on the DNA methylation of bovine skeletal muscle satellite cells (SMSCs) remains unclear. In this study, we developed a differentiation model of SMSCs and employed RNA sequencing (RNA-seq) alongside whole genome bisulfite sequencing (WGBS) to explore the effects of butyrate treatment on DNA methylation status and its relationship with gene expression. Treatment of SMSCs with sodium butyrate (NaB) at 1.0 mM for 2 days significantly inhibited the expression of DNA methyltransferases (DNMT1, DNMT2, DNMT3A) at the mRNA and protein levels while promoting the expression of demethylases (TET1, TET2, TET3) at mRNA levels. WGBS identified 4292 differentially methylated regions (DMRs), comprising 2294 hypermethylated and 1998 hypomethylated regions. These DMRs were significantly enriched in the MAPK, cAMP, and Wnt signaling pathways, all of which are implicated in myogenesis and development. Combining RNA-seq and WGBS data revealed a total of 130 overlapping genes, including MDFIC, CREBBP, DMD, LTBP2 and KLF4. These genes are predominantly involved in regulating the FoxO, MAPK, PI3K-Akt, and Wnt signaling pathways. This study provides new insights into the effects of butyrate-mediated DNA methylation on SMSC development and enhances our understanding of butyrate as an epigenetic modifier beyond its role in acetylation.
Collapse
Affiliation(s)
- Xiaowei Wang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China; Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Ningxia Yinchuan 750002, China
| | - Xiaonan Zhou
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Chenglong Li
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Chang Qu
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Yuangang Shi
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Cong-Jun Li
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD 20705, USA.
| | - Xiaolong Kang
- Key Laboratory of Ruminant Molecular and Cellular Breeding, College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
4
|
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, Treebak JT. Impaired skeletal muscle regeneration in diabetes: From cellular and molecular mechanisms to novel treatments. Cell Metab 2024; 36:1204-1236. [PMID: 38490209 DOI: 10.1016/j.cmet.2024.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Diabetes represents a major public health concern with a considerable impact on human life and healthcare expenditures. It is now well established that diabetes is characterized by a severe skeletal muscle pathology that limits functional capacity and quality of life. Increasing evidence indicates that diabetes is also one of the most prevalent disorders characterized by impaired skeletal muscle regeneration, yet underlying mechanisms and therapeutic treatments remain poorly established. In this review, we describe the cellular and molecular alterations currently known to occur during skeletal muscle regeneration in people with diabetes and animal models of diabetes, including its associated comorbidities, e.g., obesity, hyperinsulinemia, and insulin resistance. We describe the role of myogenic and non-myogenic cell types on muscle regeneration in conditions with or without diabetes. Therapies for skeletal muscle regeneration and gaps in our knowledge are also discussed, while proposing future directions for the field.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
5
|
Nielsen SDH, Sahebekhtiari N, Huang Z, Young JF, Rasmussen MK. Comparison of secreted miRNAs and proteins during proliferation and differentiation of bovine satellite cells in culture implies potential roles in regulating myogenesis. Gene 2024; 894:147979. [PMID: 37952749 DOI: 10.1016/j.gene.2023.147979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Cultivated meat is an emerging new technology to produce sustainable meat for the future. The common approach for cultivated meat, is the isolation of satellite cells from donor animals, followed by in vitro proliferation and differentiation into primitive muscle fibers. The transformation of satellite cells into myofibers is tightly orchestrated by intra-cellular signaling, while the inter-cellular signaling is less well understood. Thus, the current study was conducted to map the secretion of potential signaling molecules (MicroRNAs and proteins) during proliferation and differentiation. Primary cultures of satellite cells were grown to 50% and 80% confluence, representing the proliferative phase or serum-starved for 1 and 3 days to induce differentiation. Post incubation in FBS-free media, the media were collected and analyzed for miRNA and protein content using gene-arrays and LC-MS/MS, respectively. When comparing the miRNA secretome at 50% and 80% confluence, we observed four differentially expressed miRNA, while only five were differentially expressed when comparing Day 1 to Day 3. A subsequent in silico analysis suggested that pathways of importance for myogenesis, e.g., MAPK and AMPK signaling, could be regulated by the secreted miRNAs. In addition, >300 proteins were secreted, including insulin-like growth factor 1 binding proteins 2, 3, 4, 5 and 6. In conclusion, this study demonstrated differential secretion of several miRNAs and proteins during both proliferation and differentiation of bovine satellite cells in vitro.
Collapse
Affiliation(s)
| | - Navid Sahebekhtiari
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | - Ziyu Huang
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | - Jette Feveile Young
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | | |
Collapse
|
6
|
Scala P, Manzo P, Longo R, Giudice V, Ciardulli MC, Serio B, Selleri C, Guadagno L, Rehak L, Maffulli N, Della Porta G. Contribution of peripheral blood mononuclear cells isolated by advanced filtration system to myogenesis of human bone marrow mesenchymal stem cells co-cultured with myoblasts. Heliyon 2023; 9:e17141. [PMID: 37484299 PMCID: PMC10361327 DOI: 10.1016/j.heliyon.2023.e17141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 07/25/2023] Open
Abstract
Background Contribution of peripheral blood mononuclear cells (PBMCs) in myogenesis is still under debate, even though blood filtration systems are commonly used in clinical practice for successfully management of critic limb ischemia. Objectives A commercial blood filter used for autologous human PBMC transplantation procedures is characterized and used to collect PBMCs, that are then added to well-established 2D in vitro myogenic models assembled with a co-culture of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and skeletal myoblasts (hSkMs) whit the aim of investigating their potential contribution to stem cell myogenic commitment. Methods A commercial blood filter was physically and chemically studied to understand its morphological characteristics and composition. PBMCs were concentrated using this system, further isolated by Ficoll-Paque density gradient centrifugation, and then added in an upper transwell chamber to a 2D co-culture of hBM-MSCs and hSkMs. Myogenic commitment was investigated by RT-PCR, immunofluorescence, and flow cytometry immunophenotyping. Cytokine levels were monitored by ELISA assay in culture media. Results The blood filtration system was disassembled and appeared to be formed by twelve membranes of poly-butylene terephthalate fibers (diameters, 0.9-4.0 μm) with pore size distribution of 1-20 μm. Filter functional characterization was achieved by characterizing collected cells by flow cytometry. Subsequently, collected PBMCs fraction was added to an in-vitro model of hBM-MSC myogenic commitment. In the presence of PBMCs, stem cells significantly upregulated myogenic genes, such as Desmin and MYH2, as confirmed by qRT-PCR and expressed related proteins by immunofluorescence (IF) assay, while downregulated pro-inflammatory cytokines (IL12A at day 14) along the 21 days of culture. Novelty Our work highlights chemical-physical properties of commercial blood filter and suggests that blood filtrated fraction of PBMC might modulate cytokine expression in response to muscle injury and promote myogenic events, supporting their clinical use in autologous transplantation.
Collapse
Affiliation(s)
- Pasqualina Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
| | - Paola Manzo
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D'Aragona”, Largo Città d'Ippocrate, 1, 84131 Salerno SA, Italy
| | - Raffaele Longo
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D'Aragona”, Largo Città d'Ippocrate, 1, 84131 Salerno SA, Italy
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
| | - Bianca Serio
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D'Aragona”, Largo Città d'Ippocrate, 1, 84131 Salerno SA, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D'Aragona”, Largo Città d'Ippocrate, 1, 84131 Salerno SA, Italy
| | - Liberata Guadagno
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy
| | - Laura Rehak
- Athena Biomedical Innovations, Viale Europa 139, Florence, 50126, Italy
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 275 Bancroft Road, London E1 4DG, UK
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 43, 84081 Baronissi SA, Italy
- Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo II, 132, 84084 Fisciano SA, Italy
| |
Collapse
|
7
|
Zhao Y, Ding Y, He L, Zhou Q, Chen X, Li Y, Alfonsi MV, Wu Z, Sun H, Wang H. Multiscale 3D genome reorganization during skeletal muscle stem cell lineage progression and aging. SCIENCE ADVANCES 2023; 9:eabo1360. [PMID: 36800432 PMCID: PMC9937580 DOI: 10.1126/sciadv.abo1360] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/17/2023] [Indexed: 06/13/2023]
Abstract
Little is known about three-dimensional (3D) genome organization in skeletal muscle stem cells [also called satellite cells (SCs)]. Here, we comprehensively map the 3D genome topology reorganization during mouse SC lineage progression. Specifically, rewiring at the compartment level is most pronounced when SCs become activated. Marked loss in topologically associating domain (TAD) border insulation and chromatin looping also occurs during early activation process. Meanwhile, TADs can form TAD clusters and super-enhancer-containing TAD clusters orchestrate stage-specific gene expression. Furthermore, we uncover that transcription factor PAX7 is pivotal in enhancer-promoter (E-P) loop formation. We also identify cis-regulatory elements that are crucial for local chromatin organization at Pax7 locus and Pax7 expression. Lastly, we unveil that geriatric SC displays a prominent gain in long-range contacts and loss of TAD border insulation. Together, our results uncover that 3D chromatin extensively reorganizes at multiple architectural levels and underpins the transcriptome remodeling during SC lineage development and SC aging.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yingzhe Ding
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Liangqiang He
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Qin Zhou
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaona Chen
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuying Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Maria Vittoria Alfonsi
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhenguo Wu
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hao Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Huating Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
8
|
Scala P, Manzo P, Lamparelli EP, Lovecchio J, Ciardulli MC, Giudice V, Selleri C, Giordano E, Rehak L, Maffulli N, Della Porta G. Peripheral blood mononuclear cells contribute to myogenesis in a 3D bioengineered system of bone marrow mesenchymal stem cells and myoblasts. Front Bioeng Biotechnol 2023; 10:1075715. [PMID: 36704300 PMCID: PMC9871311 DOI: 10.3389/fbioe.2022.1075715] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
In this work, a 3D environment obtained using fibrin scaffold and two cell populations, such as bone marrow-derived mesenchymal stem cells (BM-MSCs), and primary skeletal muscle cells (SkMs), was assembled. Peripheral blood mononuclear cells (PBMCs) fraction obtained after blood filtration with HemaTrate® filter was then added to the 3D culture system to explore their influence on myogenesis. The best cell ratio into a 3D fibrin hydrogel was 1:1 (BM-MSCs plus SkMs:PBMCs) when cultured in a perfusion bioreactor; indeed, excellent viability and myogenic event induction were observed. Myogenic genes were significantly overexpressed when cultured with PBMCs, such as MyoD1 of 118-fold at day 14 and Desmin 6-fold at day 21. Desmin and Myosin Heavy Chain were also detected at protein level by immunostaining along the culture. Moreover, the presence of PBMCs in 3D culture induced a significant downregulation of pro-inflammatory cytokine gene expression, such as IL6. This smart biomimetic environment can be an excellent tool for investigation of cellular crosstalk and PBMC influence on myogenic processes.
Collapse
Affiliation(s)
- Pasqualina Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Paola Manzo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | | | - Joseph Lovecchio
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Bologna, Italy
| | | | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Bologna, Italy
| | - Laura Rehak
- Athena Biomedical innovations, Florence, Italy
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, England
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Interdepartment Centre BIONAM, University of Salerno, Fisciano, Italy,*Correspondence: Giovanna Della Porta,
| |
Collapse
|
9
|
Enhanced Muscle Fibers of Epinephelus coioides by Myostatin Autologous Nucleic Acid Vaccine. Int J Mol Sci 2022; 23:ijms23136997. [PMID: 35805999 PMCID: PMC9266527 DOI: 10.3390/ijms23136997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
Epinephelus coioides is a fish species with high economic value due to its delicious meat, high protein content, and rich fatty acid nutrition. It has become a high-economic fish in southern parts of China and some other Southeast Asian countries. In this study, the myostatin nucleic acid vaccine was constructed and used to immunize E. coioides. The results from body length and weight measurements indicated the myostatin nucleic acid vaccine promoted E. coioides growth performance by increasing muscle fiber size. The results from RT-qPCR analysis showed that myostatin nucleic acid vaccine upregulated the expression of myod, myog and p21 mRNA, downregulated the expression of smad3 and mrf4 mRNA. This preliminary study is the first report that explored the role of myostatin in E. coioides and showed positive effects of autologous nucleic acid vaccine on the muscle growth of E. coioides. Further experiments with increased numbers of animals and different doses are needed for its application to E. coiodes aquaculture production.
Collapse
|
10
|
Rodgers BD, Ward CW. Myostatin/Activin Receptor Ligands in Muscle and the Development Status of Attenuating Drugs. Endocr Rev 2022; 43:329-365. [PMID: 34520530 PMCID: PMC8905337 DOI: 10.1210/endrev/bnab030] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Muscle wasting disease indications are among the most debilitating and often deadly noncommunicable disease states. As a comorbidity, muscle wasting is associated with different neuromuscular diseases and myopathies, cancer, heart failure, chronic pulmonary and renal diseases, peripheral neuropathies, inflammatory disorders, and, of course, musculoskeletal injuries. Current treatment strategies are relatively ineffective and can at best only limit the rate of muscle degeneration. This includes nutritional supplementation and appetite stimulants as well as immunosuppressants capable of exacerbating muscle loss. Arguably, the most promising treatments in development attempt to disrupt myostatin and activin receptor signaling because these circulating factors are potent inhibitors of muscle growth and regulators of muscle progenitor cell differentiation. Indeed, several studies demonstrated the clinical potential of "inhibiting the inhibitors," increasing muscle cell protein synthesis, decreasing degradation, enhancing mitochondrial biogenesis, and preserving muscle function. Such changes can prevent muscle wasting in various disease animal models yet many drugs targeting this pathway failed during clinical trials, some from serious treatment-related adverse events and off-target interactions. More often, however, failures resulted from the inability to improve muscle function despite preserving muscle mass. Drugs still in development include antibodies and gene therapeutics, all with different targets and thus, safety, efficacy, and proposed use profiles. Each is unique in design and, if successful, could revolutionize the treatment of both acute and chronic muscle wasting. They could also be used in combination with other developing therapeutics for related muscle pathologies or even metabolic diseases.
Collapse
Affiliation(s)
| | - Christopher W Ward
- Department of Orthopedics and Center for Biomedical Engineering and Technology (BioMET), University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Garcia LA, Zapata-Bustos R, Day SE, Campos B, Hamzaoui Y, Wu L, Leon AD, Krentzel J, Coletta RL, De Filippis E, Roust LR, Mandarino LJ, Coletta DK. Can Exercise Training Alter Human Skeletal Muscle DNA Methylation? Metabolites 2022; 12:metabo12030222. [PMID: 35323665 PMCID: PMC8953782 DOI: 10.3390/metabo12030222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 12/26/2022] Open
Abstract
Skeletal muscle is highly plastic and dynamically regulated by the body’s physical demands. This study aimed to determine the plasticity of skeletal muscle DNA methylation in response to 8 weeks of supervised exercise training in volunteers with a range of insulin sensitivities. We studied 13 sedentary participants and performed euglycemic hyperinsulinemic clamps with basal vastus lateralis muscle biopsies and peak aerobic activity (VO2 peak) tests before and after training. We extracted DNA from the muscle biopsies and performed global methylation using Illumina’s Methylation EPIC 850K BeadChip. Training significantly increased peak aerobic capacity and insulin-stimulated glucose disposal. Fasting serum insulin and insulin levels during the steady state of the clamp were significantly lower post-training. Insulin clearance rates during the clamp increased following the training. We identified 13 increased and 90 decreased differentially methylated cytosines (DMCs) in response to 8 weeks of training. Of the 13 increased DMCs, 2 were within the following genes, FSTL3, and RP11-624M8.1. Of the 90 decreased DMCs, 9 were within the genes CNGA1, FCGR2A, KIF21A, MEIS1, NT5DC1, OR4D1, PRPF4B, SLC26A7, and ZNF280C. Moreover, pathway analysis showed an enrichment in metabolic and actin-cytoskeleton pathways for the decreased DMCs, and for the increased DMCs, an enrichment in signal-dependent regulation of myogenesis, NOTCH2 activation and transmission, and SMAD2/3: SMAD4 transcriptional activity pathways. Our findings showed that 8 weeks of exercise training alters skeletal muscle DNA methylation of specific genes and pathways in people with varying degrees of insulin sensitivity.
Collapse
Affiliation(s)
- Luis A. Garcia
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ 85724, USA; (L.A.G.); (R.Z.-B.); (B.C.); (A.D.L.); (J.K.); (L.J.M.)
- Center for Disparities in Diabetes Obesity and Metabolism, University of Arizona, Tucson, AZ 85724, USA;
| | - Rocio Zapata-Bustos
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ 85724, USA; (L.A.G.); (R.Z.-B.); (B.C.); (A.D.L.); (J.K.); (L.J.M.)
- Center for Disparities in Diabetes Obesity and Metabolism, University of Arizona, Tucson, AZ 85724, USA;
| | - Samantha E. Day
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ 85004, USA;
| | - Baltazar Campos
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ 85724, USA; (L.A.G.); (R.Z.-B.); (B.C.); (A.D.L.); (J.K.); (L.J.M.)
- Center for Disparities in Diabetes Obesity and Metabolism, University of Arizona, Tucson, AZ 85724, USA;
| | - Yassin Hamzaoui
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA; (Y.H.); (L.W.)
| | - Linda Wu
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA; (Y.H.); (L.W.)
| | - Alma D. Leon
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ 85724, USA; (L.A.G.); (R.Z.-B.); (B.C.); (A.D.L.); (J.K.); (L.J.M.)
- Center for Disparities in Diabetes Obesity and Metabolism, University of Arizona, Tucson, AZ 85724, USA;
| | - Judith Krentzel
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ 85724, USA; (L.A.G.); (R.Z.-B.); (B.C.); (A.D.L.); (J.K.); (L.J.M.)
- Center for Disparities in Diabetes Obesity and Metabolism, University of Arizona, Tucson, AZ 85724, USA;
| | - Richard L. Coletta
- Center for Disparities in Diabetes Obesity and Metabolism, University of Arizona, Tucson, AZ 85724, USA;
| | - Eleanna De Filippis
- Department of Endocrinology, Metabolism and Diabetes, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA; (E.D.F.); (L.R.R.)
| | - Lori R. Roust
- Department of Endocrinology, Metabolism and Diabetes, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA; (E.D.F.); (L.R.R.)
| | - Lawrence J. Mandarino
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ 85724, USA; (L.A.G.); (R.Z.-B.); (B.C.); (A.D.L.); (J.K.); (L.J.M.)
- Center for Disparities in Diabetes Obesity and Metabolism, University of Arizona, Tucson, AZ 85724, USA;
| | - Dawn K. Coletta
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ 85724, USA; (L.A.G.); (R.Z.-B.); (B.C.); (A.D.L.); (J.K.); (L.J.M.)
- Center for Disparities in Diabetes Obesity and Metabolism, University of Arizona, Tucson, AZ 85724, USA;
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA; (Y.H.); (L.W.)
- Correspondence: ; Tel.: +1-(520)-626-9316
| |
Collapse
|
12
|
Esposito P, Picciotto D, Battaglia Y, Costigliolo F, Viazzi F, Verzola D. Myostatin: Basic biology to clinical application. Adv Clin Chem 2021; 106:181-234. [PMID: 35152972 DOI: 10.1016/bs.acc.2021.09.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Myostatin is a member of the transforming growth factor (TGF)-β superfamily. It is expressed by animal and human skeletal muscle cells where it limits muscle growth and promotes protein breakdown. Its effects are influenced by complex mechanisms including transcriptional and epigenetic regulation and modulation by extracellular binding proteins. Due to its actions in promoting muscle atrophy and cachexia, myostatin has been investigated as a promising therapeutic target to counteract muscle mass loss in experimental models and patients affected by different muscle-wasting conditions. Moreover, growing evidence indicates that myostatin, beyond to regulate skeletal muscle growth, may have a role in many physiologic and pathologic processes, such as obesity, insulin resistance, cardiovascular and chronic kidney disease. In this chapter, we review myostatin biology, including intracellular and extracellular regulatory pathways, and the role of myostatin in modulating physiologic processes, such as muscle growth and aging. Moreover, we discuss the most relevant experimental and clinical evidence supporting the extra-muscle effects of myostatin. Finally, we consider the main strategies developed and tested to inhibit myostatin in clinical trials and discuss the limits and future perspectives of the research on myostatin.
Collapse
Affiliation(s)
- Pasquale Esposito
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Daniela Picciotto
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Yuri Battaglia
- Nephrology and Dialysis Unit, St. Anna University Hospital, Ferrara, Italy
| | - Francesca Costigliolo
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Viazzi
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Verzola
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
13
|
Nair VD, Vasoya M, Nair V, Smith GR, Pincas H, Ge Y, Douglas CM, Esser KA, Sealfon SC. Differential analysis of chromatin accessibility and gene expression profiles identifies cis-regulatory elements in rat adipose and muscle. Genomics 2021; 113:3827-3841. [PMID: 34547403 DOI: 10.1016/j.ygeno.2021.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 01/04/2023]
Abstract
Chromatin accessibility is a key factor influencing gene expression. We optimized the Omni-ATAC-seq protocol and used it together with RNA-seq to investigate cis-regulatory elements in rat white adipose and skeletal muscle, two tissues with contrasting metabolic functions. While promoter accessibility correlated with RNA expression, integration of the two datasets identified tissue-specific differentially accessible regions (DARs) that predominantly localized in intergenic and intron regions. DARs were mapped to differentially expressed (DE) genes enriched in distinct biological processes in each tissue. Randomly selected DE genes were validated by qPCR. Top enriched motifs in DARs predicted binding sites for transcription factors (TFs) showing tissue-specific up-regulation. The correlation between differential chromatin accessibility at a given TF binding motif and differential expression of target genes further supported the functional relevance of that motif. Our study identified cis-regulatory regions that likely play a major role in the regulation of tissue-specific gene expression in adipose and muscle.
Collapse
Affiliation(s)
- Venugopalan D Nair
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Mital Vasoya
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vishnu Nair
- Department of Computer Sciences, Columbia University, New York, NY 10027, USA
| | - Gregory R Smith
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hanna Pincas
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yongchao Ge
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Collin M Douglas
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - Stuart C Sealfon
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
14
|
Shen X, Liu Z, Wang C, Xu F, Zhang J, Li M, Lei Y, Wang A, Bi C, Zhu G. Inhibition of Postn Rescues Myogenesis Defects in Myotonic Dystrophy Type 1 Myoblast Model. Front Cell Dev Biol 2021; 9:710112. [PMID: 34490258 PMCID: PMC8417118 DOI: 10.3389/fcell.2021.710112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/30/2021] [Indexed: 12/27/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is an inherited neuromuscular disease caused by expanded CTG repeats in the 3' untranslated region (3'UTR) of the DMPK gene. The myogenesis process is defective in DM1, which is closely associated with progressive muscle weakness and wasting. Despite many proposed explanations for the myogenesis defects in DM1, the underlying mechanism and the involvement of the extracellular microenvironment remained unknown. Here, we constructed a DM1 myoblast cell model and reproduced the myogenesis defects. By RNA sequencing (RNA-seq), we discovered that periostin (Postn) was the most significantly upregulated gene in DM1 myogenesis compared with normal controls. This difference in Postn was confirmed by real-time quantitative PCR (RT-qPCR) and western blotting. Moreover, Postn was found to be significantly upregulated in skeletal muscle and myoblasts of DM1 patients. Next, we knocked down Postn using a short hairpin RNA (shRNA) in DM1 myoblast cells and found that the myogenesis defects in the DM1 group were successfully rescued, as evidenced by increases in the myotube area, the fusion index, and the expression of myogenesis regulatory genes. Similarly, Postn knockdown in normal myoblast cells enhanced myogenesis. As POSTN is a secreted protein, we treated the DM1 myoblast cells with a POSTN-neutralizing antibody and found that DM1 myogenesis defects were successfully rescued by POSTN neutralization. We also tested the myogenic ability of myoblasts in the skeletal muscle injury mouse model and found that Postn knockdown improved the myogenic ability of DM1 myoblasts. The activity of the TGF-β/Smad3 pathway was upregulated during DM1 myogenesis but repressed when inhibiting Postn with a Postn shRNA or a POSTN-neutralizing antibody, which suggested that the TGF-β/Smad3 pathway might mediate the function of Postn in DM1 myogenesis. These results suggest that Postn is a potential therapeutical target for the treatment of myogenesis defects in DM1.
Collapse
Affiliation(s)
- Xiaopeng Shen
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Zhongxian Liu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Chunguang Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Feng Xu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Jingyi Zhang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Meng Li
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Yang Lei
- Wuhu Center for Disease Control and Prevention, Wuhu, China
| | - Ao Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Chao Bi
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Guoping Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, China
| |
Collapse
|
15
|
Lamarche É, AlSudais H, Rajgara R, Fu D, Omaiche S, Wiper-Bergeron N. SMAD2 promotes myogenin expression and terminal myogenic differentiation. Development 2021; 148:dev.195495. [PMID: 33462116 DOI: 10.1242/dev.195495] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/24/2020] [Indexed: 11/20/2022]
Abstract
SMAD2 is a transcription factor, the activity of which is regulated by members of the transforming growth factor β (TGFβ) superfamily. Although activation of SMAD2 and SMAD3 downstream of TGFβ or myostatin signaling is known to inhibit myogenesis, we found that SMAD2 in the absence of TGFβ signaling promotes terminal myogenic differentiation. We found that, during myogenic differentiation, SMAD2 expression is induced. Knockout of SMAD2 expression in primary myoblasts did not affect the efficiency of myogenic differentiation but produced smaller myotubes with reduced expression of the terminal differentiation marker myogenin. Conversely, overexpression of SMAD2 stimulated myogenin expression, and enhanced both differentiation and fusion, and these effects were independent of classical activation by the TGFβ receptor complex. Loss of Smad2 in muscle satellite cells in vivo resulted in decreased muscle fiber caliber and impaired regeneration after acute injury. Taken together, we demonstrate that SMAD2 is an important positive regulator of myogenic differentiation, in part through the regulation of Myog.
Collapse
Affiliation(s)
- Émilie Lamarche
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Hamood AlSudais
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Rashida Rajgara
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Dechen Fu
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Saadeddine Omaiche
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Nadine Wiper-Bergeron
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
16
|
Skoglund E, Grönholdt-Klein M, Rullman E, Thornell LE, Strömberg A, Hedman A, Cederholm T, Ulfhake B, Gustafsson T. Longitudinal Muscle and Myocellular Changes in Community-Dwelling Men Over Two Decades of Successful Aging-The ULSAM Cohort Revisited. J Gerontol A Biol Sci Med Sci 2021; 75:654-663. [PMID: 31002330 DOI: 10.1093/gerona/glz068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
Participants of the population-based Uppsala longitudinal study of adult men (ULSAM) cohort reaching more than 88 years of age (survivors, S) were investigated at age 70, 82, and 88-90 and compared at 70 years with non-survivors (NS) not reaching 82 years. Body composition, muscle mass and muscle histology were remarkably stable over 18 years of advanced aging in S. Analysis of genes involved in muscle remodeling showed that S had higher mRNA levels of myogenic differentiation factors (Myogenin, MyoD), embryonic myosin (eMyHC), enzymes involved in regulated breakdown of myofibrillar proteins (Smad2, Trim32, MuRF1,) and NCAM compared with healthy adult men (n = 8). S also had higher mRNA levels of eMyHC, Smad 2, MuRF1 compared with NS. At 88 years, S expressed decreased levels of Myogenin, MyoD, eMyHC, NCAM and Smad2 towards those seen in NS at 70 years. The gene expression pattern of S at 70 years was likely beneficial since they maintained muscle fiber histology and appendicular lean body mass until advanced age. The expression pattern at 88 years may indicate a diminished muscle remodeling coherent with a decline of reinnervation capacity and/or plasticity at advanced age.
Collapse
Affiliation(s)
- Elisabeth Skoglund
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden.,Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Sweden.,Department of Integrative Medical Biology, Umeå University, Sweden
| | | | - Eric Rullman
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Anna Strömberg
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Anu Hedman
- Heart Centre East-Tallinn Central Hospital, Estonia
| | - Tommy Cederholm
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Sweden
| | - Brun Ulfhake
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Gustafsson
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Grönholdt‐Klein M, Altun M, Becklén M, Dickman Kahm E, Fahlström A, Rullman E, Ulfhake B. Muscle atrophy and regeneration associated with behavioural loss and recovery of function after sciatic nerve crush. Acta Physiol (Oxf) 2019; 227:e13335. [PMID: 31199566 DOI: 10.1111/apha.13335] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/31/2019] [Accepted: 06/11/2019] [Indexed: 12/22/2022]
Abstract
AIM To resolve timing and coordination of denervation atrophy and the re-innervation recovery process to discern correlations indicative of common programs governing these processes. METHODS Female Sprague-Dawley (SD) rats had a unilateral sciatic nerve crush. Based on longitudinal behavioural observations, the triceps surae muscle was analysed at different time points post-lesion. RESULTS Crush results in a loss of muscle function and mass (-30%) followed by a recovery to almost pre-lesion status at 30 days post-crush (dpc). There was no loss of fibres nor any significant change in the number of nuclei per fibre but a shift in fibres expressing myosins I and II that reverted back to control levels at 30 dpc. A residual was the persistence of hybrid fibres. Early on a CHNR -ε to -γ switch and a re-expression of embryonic MyHC showed as signs of denervation. Foxo1, Smad3, Fbxo32 and Trim63 transcripts were upregulated but not Myostatin, InhibinA and ActivinR2B. Combined this suggests that the mechanism instigating atrophy provides a selectivity of pathway(s) activated. The myogenic differentiation factors (MDFs: Myog, Myod1 and Myf6) were upregulated early on suggesting a role also in the initial atrophy. The regulation of these transcripts returned towards baseline at 30 dpc. The examined genes showed a strong baseline covariance in transcript levels which dissolved in the response to crush driven mainly by the MDFs. At 30 dpc the naïve expression pattern was re-established. CONCLUSION Peripheral nerve crush offers an excellent model to assess and interfere with muscle adaptions to denervation and re-innervation.
Collapse
Affiliation(s)
| | - Mikael Altun
- Department of Laboratory Medicine Karolinska Institutet Huddinge Sweden
| | - Meneca Becklén
- Department of Neuroscience Karolinska Institutet Stockholm Sweden
| | | | - Andreas Fahlström
- Department of Neuroscience Karolinska Institutet Stockholm Sweden
- Department of Neuroscience, Neurosurgery Uppsala University Uppsala Sweden
| | - Eric Rullman
- Department of Laboratory Medicine Karolinska Institutet Huddinge Sweden
| | - Brun Ulfhake
- Department of Neuroscience Karolinska Institutet Stockholm Sweden
| |
Collapse
|
18
|
Gene expression profiling of skeletal myogenesis in human embryonic stem cells reveals a potential cascade of transcription factors regulating stages of myogenesis, including quiescent/activated satellite cell-like gene expression. PLoS One 2019; 14:e0222946. [PMID: 31560727 PMCID: PMC6764674 DOI: 10.1371/journal.pone.0222946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/10/2019] [Indexed: 01/05/2023] Open
Abstract
Human embryonic stem cell (hESC)-derived skeletal muscle progenitors (SMP)—defined as PAX7-expressing cells with myogenic potential—can provide an abundant source of donor material for muscle stem cell therapy. As in vitro myogenesis is decoupled from in vivo timing and 3D-embryo structure, it is important to characterize what stage or type of muscle is modeled in culture. Here, gene expression profiling is analyzed in hESCs over a 50 day skeletal myogenesis protocol and compared to datasets of other hESC-derived skeletal muscle and adult murine satellite cells. Furthermore, day 2 cultures differentiated with high or lower concentrations of CHIR99021, a GSK3A/GSK3B inhibitor, were contrasted. Expression profiling of the 50 day time course identified successively expressed gene subsets involved in mesoderm/paraxial mesoderm induction, somitogenesis, and skeletal muscle commitment/formation which could be regulated by a putative cascade of transcription factors. Initiating differentiation with higher CHIR99021 concentrations significantly increased expression of MSGN1 and TGFB-superfamily genes, notably NODAL, resulting in enhanced paraxial mesoderm and reduced ectoderm/neuronal gene expression. Comparison to adult satellite cells revealed that genes expressed in 50-day cultures correlated better with those expressed by quiescent or early activated satellite cells, which have the greatest therapeutic potential. Day 50 cultures were similar to other hESC-derived skeletal muscle and both expressed known and novel SMP surface proteins. Overall, a putative cascade of transcription factors has been identified which regulates four stages of myogenesis. Subsets of these factors were upregulated by high CHIR99021 or their binding sites were significantly over-represented during SMP activation, ranging from quiescent to late-activated stages. This analysis serves as a resource to further study the progression of in vitro skeletal myogenesis and could be mined to identify novel markers of pluripotent-derived SMPs or regulatory transcription/growth factors. Finally, 50-day hESC-derived SMPs appear similar to quiescent/early activated satellite cells, suggesting they possess therapeutic potential.
Collapse
|
19
|
Rao VV, Sangiah U, Mary KA, Akira S, Mohanty A. Role of Akirin1 in the regulation of skeletal muscle fiber-type switch. J Cell Biochem 2019; 120:11284-11304. [PMID: 30746755 DOI: 10.1002/jcb.28406] [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: 08/31/2018] [Revised: 12/19/2018] [Accepted: 01/07/2019] [Indexed: 01/24/2023]
Abstract
Akirin1 is a highly conserved ubiquitously expressed nuclear protein. Owing to its strong nuclear localization signal and protein-protein interaction properties, Akirin1 has been speculated to regulate transcription of target genes as a cofactor. Previous studies have reported Akirin1 as a downstream target of myostatin, a potent negative regulator of myogenesis. Mice lacking myostatin displayed enhanced Akirin1 gene expression. Further, in vitro evidence has shown Akirin1 overexpression leads to hypertrophy in C2 C 12 myotubes. In this study, we used Akirin1 knockout mice as a model system to further elucidate the function of Akirin1 in fully differentiated skeletal muscle. Akirin1 knockout mice did not show any obvious phenotypic difference when compared with wild type. However, promoter-reporter assay suggested that Akirin1 regulated the transcription of muscle-specific RING finger 1 (MuRF-1), an important E3 ubiquitin ligase in skeletal muscle. Furthermore, ablation of Akirin1 resulted in increased type IIa and decreased type I muscle fibers, which was further supported by an increase in Myh2 and decrease in Myh7 gene expression. Also, histochemical studies for succinate dehydrogenase activity revealed a less oxidative muscle in the absence of Akirin1. Together, our study suggests a novel role of Akirin1 in maintaining the muscle fiber type and regulation of the metabolic activity of the skeletal muscle.
Collapse
Affiliation(s)
- Vanitha Venkoba Rao
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Umamaheswari Sangiah
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kavitha Arockia Mary
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shizuo Akira
- Department of Host Defense, Osaka University, Suita, Japan
| | - Abhishek Mohanty
- Department of Molecular Oncology, MVR Cancer Center and Research Institute, Kozhikode, India
| |
Collapse
|
20
|
Zhang L, Ning Y, Li P, Zan L. Smad3 influences Smad2 expression via the transcription factor C/EBPα and C/EBPβ during bovine myoblast differentiation. Arch Biochem Biophys 2019; 671:235-244. [PMID: 31071302 DOI: 10.1016/j.abb.2019.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/17/2019] [Accepted: 05/05/2019] [Indexed: 01/14/2023]
Abstract
Transforming growth factor β (TGFβ) has participated in a variety of cellular biological processes. Smad2 and Smad3 are equally important TGFβ downstream effectors in mediating TGFβ signals. However, genes involved in controlling the balance between these two signaling pathways are unknown. In this study, we showed that although Smad2 and Smad3 are structurally similar, with 89% amino acid sequence similarity in bovine, Smad3 significantly decreased Smad2 mRNA and protein expression during bovine myoblast differentiation, but not by binding on its promoter. Luciferase assays and electrophoretic mobility shift assays (EMSA) demonstrated that the transcription factors C/EBPα and C/EBPβ activate Smad2 promoter activity and expression under high serum medium (GM), whereas the opposite was observed under low serum medium (DM). Moreover, over-expression and interference assays revealed that Smad3 has a different effect on C/EBPα and C/EBPβ expression under GM versus DM conditions. After mutation of the C/EBPα and C/EBPβ binding sites, Smad3 had a reduced effect on Smad2 promoter activity. Therefore, these results demonstrated that Smad3 inhibits Smad2 expression via its transcription factors C/EBPα and C/EBPβ during bovine myoblast differentiation. This novel mechanism of the Smad2/3 genes may offer clues for further investigation of TGFβ signal function.
Collapse
Affiliation(s)
- Le Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China; School of Physical Education, Yan'an University, Yan'an, Shaanxi, China
| | - Yue Ning
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Peiwei Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China; National Beef Cattle Improvement Center, Yangling, Shaanxi, China.
| |
Collapse
|
21
|
Kim J, Wellmann KB, Smith ZK, Johnson BJ. All-trans retinoic acid increases the expression of oxidative myosin heavy chain through the PPARδ pathway in bovine muscle cells derived from satellite cells. J Anim Sci 2018; 96:2763-2776. [PMID: 29688535 DOI: 10.1093/jas/sky155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/19/2018] [Indexed: 11/15/2022] Open
Abstract
All-trans retinoic acid (ATRA) has been associated with various physiological phenomenon in mammalian adipose tissue and skeletal muscle. We hypothesized that ATRA may affect skeletal muscle fiber type in bovine satellite cell culture through various transcriptional processes. Bovine primary satellite cell (BSC) culture experiments were conducted to determine dose effects of ATRA on expression of genes and protein levels related to skeletal muscle fiber type and metabolism. The semimembranosus from crossbred steers (n = 2 steers), aged approximately 24 mo, were used to isolate BSC for 3 separate assays. Myogenic differentiation was induced using 3% horse serum upon cultured BSC with increasing doses (0, 1, 10, 100, and 1,000 nM) of ATRA. After 96 h of incubation, cells were harvested and used to measure the gene expression of protein kinase B (Akt), AMP-activated protein kinase alpha (AMPK), glucose transporter 4 (GLUT4), myogenin, lipoprotein lipase (LPL), myosin heavy chain (MHC) I, MHC IIA, MHC IIX, insulin like growth factor-1 (IGF-1), Peroxisome proliferator activated receptor gamma (PPARγ), PPARδ, and Smad transcription factor 3 (SMAD3) mRNA relative to ribosomal protein subunit 9 (RPS9). The mRNA expression of LPL was increased (P < 0.05) with 100 and 1,000 nM of ATRA. Expression of GLUT4 was altered (P < 0.05) by ATRA. The treatment of ATRA (1,000 nM) also increased (P < 0.05) mRNA gene expression of SMAD3. The gene expression of both PPARδ and PPARγ were increased (P < 0.05) with 1,000 nM of ATRA. Protein level of PPARδ was also affected (P < 0.05) by 1,000 nM of ATRA and resulted in a greater (P < 0.05) protein level of PPARδ compared to CON. All-trans retinoic acid (10 nM) increased gene expression of MHC I (P < 0.05) compared to CON. Expression of MHC IIA was also influenced (P < 0.05) by ATRA. The mRNA expression of MHC IIX was decreased (P < 0.05) with 100 and 1,000 nM of ATRA. In muscle cells, ATRA may cause muscle fibers to transition towards the MHC isoform that prefers oxidative metabolism, as evidenced by increased expression of genes associated with the MHC I isoform. These changes in MHC isoforms appeared to be brought about by changing PPARδ gene expression and protein levels.
Collapse
Affiliation(s)
- Jongkyoo Kim
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| | | | - Zachary K Smith
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| | - Bradley J Johnson
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX
| |
Collapse
|
22
|
Islam R, Yoon H, Shin HR, Bae HS, Kim BS, Yoon WJ, Woo KM, Baek JH, Lee YS, Ryoo HM. Peptidyl-prolyl cis-trans isomerase NIMA interacting 1 regulates skeletal muscle fusion through structural modification of Smad3 in the linker region. J Cell Physiol 2018; 233:9390-9403. [PMID: 30132832 PMCID: PMC6686165 DOI: 10.1002/jcp.26774] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
Myoblast fusion is critical for muscle growth, regeneration, and repair. We previously reported that the enzyme peptidyl‐prolyl cis–trans isomerase NIMA interacting 1 (Pin1) is involved in osteoclast fusion. The objective of this study was to investigate the possibility that Pin1 also inhibits myoblast fusion. Here, we show the increased number of nuclei in the Pin1+/− mice muscle fiber compared to that in wild‐type mice. Moreover, we show that low dose of the Pin1 inhibitor dipentamethylene thiuram monosulfide treatment caused enhanced fusion in C2C12 cells. The R‐Smads are well‐known mediators of muscle hypertrophy and hyperplasia as well as being substrates of Pin1. We found that Pin1 is crucial for maintaining the stability of Smad3 (homologues of the Drosophila protein, mothers against decapentaplegic (Mad) and the Caenorhabditis elegans protein Sma). Our results show that serine 204 within Smad3 is the key Pin1‐binding site during inhibition of myoblast fusion and that both the transforming growth factor‐β receptor and extracellular signal‐regulated kinase (ERK)‐mediated phosphorylation are required for the interaction of Pin1 with Smad3. These findings suggest that a precise level of Pin1 activity is essential for regulating myoblast fusion during myogenesis and muscle regeneration.
Collapse
Affiliation(s)
- Rabia Islam
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Heein Yoon
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Hye-Rim Shin
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Han-Sol Bae
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Bong-Soo Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Won-Joon Yoon
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Mi Woo
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Jeong-Hwa Baek
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Hyun-Mo Ryoo
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
23
|
Hara M, Yokota K, Saito T, Kobayakawa K, Kijima K, Yoshizaki S, Okazaki K, Yoshida S, Matsumoto Y, Harimaya K, Nakashima Y, Okada S. Periostin Promotes Fibroblast Migration and Inhibits Muscle Repair After Skeletal Muscle Injury. J Bone Joint Surg Am 2018; 100:e108. [PMID: 30106825 DOI: 10.2106/jbjs.17.01230] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Skeletal muscle injury (SMI) can cause physical disability due to insufficient recovery of the muscle. The development of muscle fibrosis after SMI has been widely regarded as a principal cause of this failure to recover. Periostin (Postn) exacerbates tissue fibrosis in various organs. We investigated whether Postn is involved in the pathophysiology after SMI. METHODS Partial laceration injuries of the gastrocnemius were created in wild-type (WT) and Postn knockout (Postn) mice. We examined the expression of the Postn gene before and after SMI. Regeneration and fibrosis of skeletal muscle were evaluated by histological analyses, and recovery of muscle strength was measured by physiological testing. Immunohistochemistry was used to examine the number and proliferative potential of infiltrating fibroblasts in injured muscle. A trans-well migration assay was used to assess the migration capability of fibroblasts. Control immunoglobulin G (IgG) or Postn-neutralizing antibody (Postn-nAb) was injected into injured muscle at 7 and 14 days after injury (dpi). We evaluated the effects of Postn-nAb on muscle repair after SMI. RESULTS The expression of Postn was dramatically upregulated after SMI. Compared with WT mice, Postn mice had improved muscle recovery and attenuated fibrosis as well as a significantly reduced number of infiltrating fibroblasts. The proliferative potential of these fibroblasts in WT and Postn mice was comparable at 14 dpi; however, the migration capability of fibroblasts was significantly enhanced in the presence of Postn (mean, 258%; 95% confidence interval, 183% to 334%). Moreover, the administration of Postn-nAb inhibited fibroblast infiltration and promoted muscle repair after SMI. CONCLUSIONS Postn exacerbates fibrotic scar formation through the promotion of fibroblast migration into injured muscle after SMI. Treatment with Postn-nAb is effective for attenuating fibrosis and improving muscle recovery after SMI. CLINICAL RELEVANCE Our findings may provide a potential therapeutic strategy to enhance muscle repair and functional recovery after SMI.
Collapse
Affiliation(s)
- Masamitsu Hara
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Yokota
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeyuki Saito
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kijima
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Okazaki
- Department of Orthopaedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Shigeo Yoshida
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsumi Harimaya
- Department of Orthopaedic Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yasuharu Nakashima
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Okada
- Departments of Orthopaedic Surgery (M.H., K.Y., T.S., K. Kobayakawa, K. Kijima, S. Yoshizaki, Y.M., and Y.N.), Ophthalmology (S. Yoshida), and Advanced Medical Initiatives (S.O.), Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
24
|
Hostrup M, Reitelseder S, Jessen S, Kalsen A, Nyberg M, Egelund J, Kreiberg M, Kristensen CM, Thomassen M, Pilegaard H, Backer V, Jacobson GA, Holm L, Bangsbo J. Beta 2 -adrenoceptor agonist salbutamol increases protein turnover rates and alters signalling in skeletal muscle after resistance exercise in young men. J Physiol 2018; 596:4121-4139. [PMID: 29968301 DOI: 10.1113/jp275560] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/29/2018] [Indexed: 01/08/2023] Open
Abstract
KEY POINTS Animal models have shown that beta2 -adrenoceptor stimulation increases protein synthesis and attenuates breakdown processes in skeletal muscle. Thus, the beta2 -adrenoceptor is a potential target in the treatment of disuse-, disease- and age-related muscle atrophy. In the present study, we show that a few days of oral treatment with the commonly prescribed beta2 -adrenoceptor agonist, salbutamol, increased skeletal muscle protein synthesis and breakdown during the first 5 h after resistance exercise in young men. Salbutamol also counteracted a negative net protein balance in skeletal muscle after resistance exercise. Changes in protein turnover rates induced by salbutamol were associated with protein kinase A-signalling, activation of Akt2 and modulation of mRNA levels of growth-regulating proteins in skeletal muscle. These findings indicate that protein turnover rates can be augmented by beta2 -adrenoceptor agonist treatment during recovery from resistance exercise in humans. ABSTRACT The effect of beta2 -adrenoceptor stimulation on skeletal muscle protein turnover and intracellular signalling is insufficiently explored in humans, particularly in association with exercise. In a randomized, placebo-controlled, cross-over study investigating 12 trained men, the effects of beta2 -agonist (6 × 4 mg oral salbutamol) on protein turnover rates, intracellular signalling and mRNA response in skeletal muscle were investigated 0.5-5 h after quadriceps resistance exercise. Each trial was preceded by a 4-day lead-in treatment period. Leg protein turnover rates were assessed by infusion of [13 C6 ]-phenylalanine and sampling of arterial and venous blood, as well as vastus lateralis muscle biopsies 0.5 and 5 h after exercise. Furthermore, myofibrillar fractional synthesis rate, intracellular signalling and mRNA response were measured in muscle biopsies. The mean (95% confidence interval) myofibrillar fractional synthesis rate was higher for salbutamol than placebo [0.079 (95% CI, 0.064 to 0.093) vs. 0.066 (95% CI, 0.056 to 0.075%) × h-1 ] (P < 0.05). Mean net leg phenylalanine balance 0.5-5 h after exercise was higher for salbutamol than placebo [3.6 (95% CI, 1.0 to 6.2 nmol) × min-1 × 100 gLeg Lean Mass-1 ] (P < 0.01). Phosphorylation of Akt2, cAMP response element binding protein and PKA substrate 0.5 and 5 h after exercise, as well as phosphorylation of eEF2 5 h after exercise, was higher (P < 0.05) for salbutamol than placebo. Calpain-1, Forkhead box protein O1, myostatin and Smad3 mRNA content was higher (P < 0.01) for salbutamol than placebo 0.5 h after exercise, as well as Forkhead box protein O1 and myostatin mRNA content 5 h after exercise, whereas ActivinRIIB mRNA content was lower (P < 0.01) for salbutamol 5 h after exercise. These observations suggest that beta2 -agonist increases protein turnover rates in skeletal muscle after resistance exercise in humans, with concomitant cAMP/PKA and Akt2 signalling, as well as modulation of mRNA response of growth-regulating proteins.
Collapse
Affiliation(s)
- Morten Hostrup
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.,Department of Respiratory Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Søren Reitelseder
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Søren Jessen
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.,Department of Respiratory Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Anders Kalsen
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.,Department of Respiratory Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Michael Nyberg
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jon Egelund
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kreiberg
- Department of Respiratory Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | | | - Martin Thomassen
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Pilegaard
- Department of Biology, University of Copenhagen, Copenhagen, Copenhagen, Denmark
| | - Vibeke Backer
- Department of Respiratory Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Glenn A Jacobson
- Division of Pharmacy, School of Medicine, University of Tasmania, Hobart, Australia
| | - Lars Holm
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg University Hospital, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,School of Sport Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Jens Bangsbo
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Aloysius A, DasGupta R, Dhawan J. The transcription factor Lef1 switches partners from β-catenin to Smad3 during muscle stem cell quiescence. Sci Signal 2018; 11:11/540/eaan3000. [PMID: 30042129 DOI: 10.1126/scisignal.aan3000] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle stem cells (MuSCs), also known as satellite cells, persist in adult mammals by entering a state of quiescence (G0) during the early postnatal period. Quiescence is reversed during damage-induced regeneration and re-established after regeneration. Entry of cultured myoblasts into G0 is associated with a specific, reversible induction of Wnt target genes, thus implicating members of the Tcf and Lef1 (Tcf/Lef) transcription factor family, which mediate transcriptional responses to Wnt signaling, in the initiation of quiescence. We found that the canonical Wnt effector β-catenin, which cooperates with Tcf/Lef, was dispensable for myoblasts to enter quiescence. Using pharmacological and genetic approaches in cultured C2C12 myoblasts and in MuSCs, we demonstrated that Tcf/Lef activity during quiescence depended not on β-catenin but on the transforming growth factor-β (TGF-β) effector and transcriptional coactivator Smad3, which colocalized with Lef1 at canonical Wnt-responsive elements and directly interacted with Lef1 specifically in G0 Depletion of Smad3, but not β-catenin, reduced Lef1 occupancy at target promoters, Tcf/Lef target gene expression, and self-renewal of myoblasts. In vivo, MuSCs underwent a switch from β-catenin-Lef1 to Smad3-Lef1 interactions during the postnatal switch from proliferation to quiescence, with β-catenin-Lef1 interactions recurring during damage-induced reactivation. Our findings suggest that the interplay of Wnt-Tcf/Lef and TGF-β-Smad3 signaling activates canonical Wnt target promoters in a manner that depends on β-catenin during myoblast proliferation but is independent of β-catenin during MuSC quiescence.
Collapse
Affiliation(s)
- Ajoy Aloysius
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India.,Centre for Cellular and Molecular Biology, Hyderabad 500007, India.,Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India
| | | | - Jyotsna Dhawan
- Centre for Cellular and Molecular Biology, Hyderabad 500007, India. .,Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India
| |
Collapse
|
26
|
Zhang L, Ning Y, Li P, Guo H, Zan L. Tissue Expression Analysis and Characterization of Smad3 Promoter in Bovine Myoblasts and Preadipocytes. DNA Cell Biol 2018; 37:551-559. [PMID: 29672161 PMCID: PMC5985903 DOI: 10.1089/dna.2018.4152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 03/07/2018] [Accepted: 03/20/2018] [Indexed: 11/12/2022] Open
Abstract
The transforming growth factor-β (TGFβ) pathway plays many key roles in regulating numerous biological processes. In addition, the effects of TGFβ are mediated by the transcription factor Smad3. However, the regulation of Smad3 activity is not well understood. In the present study, quantitative real-time PCR revealed that the Smad3 gene was expressed ubiquitously in 11 bovine tissues and displayed different expression patterns between muscle and adipose tissue. We further explored the expression and regulation of Smad3 gene by cloning the bovine Smad3 gene promoter; a dual-luciferase reporter assay identified that the core promoter region -337 to -41 bp was located in a CpG island. In addition, mutational analyses and electrophoretic mobility shift assays provided evidence that the KLF6, KLF15, MZF1, and KLF7 binding sites within the Smad3 promoter were responsible for the regulation of Smad3 transcription. These findings were confirmed by executing further RNA interference assays in bovine myoblasts and preadipocytes, which indicated that KLF6, KLF15, MZF1, and KLF7 are important transcriptional activators of Smad3 in both adipose and muscle tissue. These results will provide an important basis for an improved understanding of the TGFβ pathway and new insights in cattle breeding.
Collapse
Affiliation(s)
- Le Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yue Ning
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Peiwei Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hongfang Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
- National Beef Cattle Improvement Center, Yangling, China
| |
Collapse
|
27
|
Grafe I, Alexander S, Peterson JR, Snider TN, Levi B, Lee B, Mishina Y. TGF-β Family Signaling in Mesenchymal Differentiation. Cold Spring Harb Perspect Biol 2018; 10:a022202. [PMID: 28507020 PMCID: PMC5932590 DOI: 10.1101/cshperspect.a022202] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) can differentiate into several lineages during development and also contribute to tissue homeostasis and regeneration, although the requirements for both may be distinct. MSC lineage commitment and progression in differentiation are regulated by members of the transforming growth factor-β (TGF-β) family. This review focuses on the roles of TGF-β family signaling in mesenchymal lineage commitment and differentiation into osteoblasts, chondrocytes, myoblasts, adipocytes, and tenocytes. We summarize the reported findings of cell culture studies, animal models, and interactions with other signaling pathways and highlight how aberrations in TGF-β family signaling can drive human disease by affecting mesenchymal differentiation.
Collapse
Affiliation(s)
- Ingo Grafe
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Stefanie Alexander
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Jonathan R Peterson
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Taylor Nicholas Snider
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Benjamin Levi
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
28
|
Zhang L, Gong H, Sun Q, Zhao R, Jia Y. Spermidine-Activated Satellite Cells Are Associated with Hypoacetylation in ACVR2B and Smad3 Binding to Myogenic Genes in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:540-550. [PMID: 29224337 DOI: 10.1021/acs.jafc.7b04482] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Spermidine is an acetyltransferase inhibitor and a specific inducer of autophagy. Recently, spermidine is identified as a potential therapeutic agent for age-related muscle atrophy and inherited myopathies. However, the effect of spermidine on nonpathological skeletal muscle remains unclear. In this study, long-term spermidine administration in mice lowered the mean cross-sectional area of the gastrocnemius muscle and reduced the expression of myosin heavy chain isoforms in the muscle, which was associated with ubiquitination. Moreover, spermidine supplementation induced autophagy in satellite cells and enhanced satellite cell proliferation. ChIP assay revealed that spermidine repressed H3K56ac in the promoter of ACVR2B and lowered the binding affinity of Smad3 to the promoters of Myf5 and MyoD. Altogether, our results indicate that long-term administration of spermidine can activate satellite cells, as well as enhance autophagy, eventually resulting in muscle atrophy. In addition, H3K56ac and Smad3 emerged as key determinants of satellite cell activation.
Collapse
Affiliation(s)
- Luchu Zhang
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing 210095, P. R. China
| | - Huiying Gong
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing 210095, P. R. China
| | - Qinwei Sun
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing 210095, P. R. China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing 210095, P. R. China
- Quality and Safety Control, Jiangsu Collaborative Innovation Center of Meat Production and Processing , Nanjing 210095, P. R. China
| | - Yimin Jia
- Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing 210095, P. R. China
- Quality and Safety Control, Jiangsu Collaborative Innovation Center of Meat Production and Processing , Nanjing 210095, P. R. China
| |
Collapse
|
29
|
Past, Present, and Future Perspective of Targeting Myostatin and Related Signaling Pathways to Counteract Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:153-206. [DOI: 10.1007/978-981-13-1435-3_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Fortes MAS, Scervino MVM, Marzuca-Nassr GN, Vitzel KF, da Justa Pinheiro CH, Curi R. Hypertrophy Stimulation at the Onset of Type I Diabetes Maintains the Soleus but Not the EDL Muscle Mass in Wistar Rats. Front Physiol 2017; 8:830. [PMID: 29123487 PMCID: PMC5662641 DOI: 10.3389/fphys.2017.00830] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/06/2017] [Indexed: 12/17/2022] Open
Abstract
Diabetes mellitus induces a reduction in skeletal muscle mass and strength. Strength training is prescribed as part of treatment since it improves glycemic control and promotes increase of skeletal muscle mass. The mechanisms involved in overload-induced muscle hypertrophy elicited at the establishment of the type I diabetic state was investigated in Wistar rats. The purpose was to examine whether the overload-induced hypertrophy can counteract the hypotrophy associated to the diabetic state. The experiments were performed in oxidative (soleus) or glycolytic (EDL) muscles. PI3K/Akt/mTOR protein synthesis pathway was evaluated 7 days after overload-induced hypertrophy of soleus and of EDL muscles. The mRNA expression of genes associated with different signaling pathways that control muscle hypertrophy was also evaluated: mechanotransduction (FAK), Wnt/β-catenin, myostatin, and follistatin. The soleus and EDL muscles when submitted to overload had similar hypertrophic responses in control and diabetic animals. The increase of absolute and specific twitch and tetanic forces had the same magnitude as muscle hypertrophic response. Hypertrophy of the EDL muscle from diabetic animals mostly involved mechanical loading-stimulated PI3K/Akt/mTOR pathway besides the reduced activation of AMP-activated protein kinase (AMPK) and decrease of myostatin expression. Hypertrophy was more pronounced in the soleus muscle of diabetic animals due to a more potent activation of rpS6 and increased mRNA expression of insulin-like growth factor-1 (IGF-1), mechano-growth factor (MGF) and follistatin, and decrease of myostatin, MuRF-1 and atrogin-1 contents. The signaling changes enabled the soleus muscle mass and force of the diabetic rats to reach the values of the control group.
Collapse
Affiliation(s)
- Marco A S Fortes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria V M Scervino
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gabriel N Marzuca-Nassr
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Kaio F Vitzel
- School of Health Sciences, College of Health, Massey University, Albany, New Zealand
| | - Carlos H da Justa Pinheiro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| |
Collapse
|
31
|
Jensen JH, Madsen LB, Panitz F, Hornshøj H, Nielsen RO, Bendixen C, Oksbjerg N, Thomsen B. Transcriptome dynamics during proliferation and differentiation of porcine primary satellite cells. GENE REPORTS 2017. [DOI: 10.1016/j.genrep.2017.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
32
|
Age and sex differences in human skeletal muscle fibrosis markers and transforming growth factor-β signaling. Eur J Appl Physiol 2017; 117:1463-1472. [PMID: 28493029 DOI: 10.1007/s00421-017-3639-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/04/2017] [Indexed: 01/01/2023]
Abstract
PURPOSE The aim of the study was to determine whether higher fibrosis markers in skeletal muscle of older adults are accompanied by increased expression of components of the canonical TGF-β signal transduction pathway. METHODS Fourteen healthy young (21-35 years; 9 males and 5 females) and seventeen older (55-75 years; 9 males and 8 females) participants underwent vastus lateralis biopsies to determine intramuscular mRNA and protein expression of fibrogenic markers and TGF-β signaling molecules related to TGF-β1 and myostatin. RESULTS Expression of mRNA encoding the pro-fibrotic factors; axin 2, collagen III, β-catenin and fibronectin, were all significantly higher (all p < 0.05) in the older participants (350, 170, 298, and 641%, respectively). Furthermore, axin 2 and β-catenin mRNA were significantly higher in older females than older males (p < 0.05). Gene expression of ActRIIB, myostatin, and TGF-β1 were higher in older adults compared to younger adults (all p < 0.05). There was, however, no difference in the total protein content of myostatin, myoD or myogenin (all p > 0.05), whereas Smad3 protein phosphorylation was 48% lower (p < 0.05) in muscle from older adults. CONCLUSIONS Increased abundance of mRNA of fibrotic markers was observed in muscle from older adults and was partly accompanied by altered abundance of pro-fibrotic ligands in a sex specific manner.
Collapse
|
33
|
Kumar R, Singh SP, Mitra A. Short-hairpin Mediated Myostatin Knockdown Resulted in Altered Expression of Myogenic Regulatory Factors with Enhanced Myoblast Proliferation in Fetal Myoblast Cells of Goats. Anim Biotechnol 2017; 29:59-67. [PMID: 28358646 DOI: 10.1080/10495398.2017.1299744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myostatin (MSTN) is a well-known negative regulator of skeletal muscle development. Reduced expression due to natural mutations in the coding region and knockout as well as knockdown of MSTN results in an increase in the muscle mass. In the present study, we demonstrated as high as 60 and 52% downregulation (p < 0.01) of MSTN mRNA and protein in the primary fetal myoblast cells of goats using synthetic shRNAs (n = 3), without any interferon response. We, for the first time, evaluated the effect of MSTN knockdown on the expression of MRFs (namely, MyoD, Myf5), follistatin (FST), and IGFs (IGF-1 & IGF-2) in goat myoblast cells. MSTN knockdown caused an upregulation (p < 0.05) of MyoD and downregulation (p < 0.01) of MYf5 and FST expression. Moreover, we report up to ∼four fold (p < 0.001) enhanced proliferation in myoblasts after four days of culture. The anti-MSTN shRNA demonstrated in the present study could be used for the production of transgenic goats to increase the muscle mass.
Collapse
Affiliation(s)
- Rohit Kumar
- a Genome Analysis Laboratory, Animal Genetics Division , ICAR- Indian Veterinary Research Institute , Izatnagar , Bareilly , India
| | - Satyendra Pal Singh
- a Genome Analysis Laboratory, Animal Genetics Division , ICAR- Indian Veterinary Research Institute , Izatnagar , Bareilly , India
| | - Abhijit Mitra
- a Genome Analysis Laboratory, Animal Genetics Division , ICAR- Indian Veterinary Research Institute , Izatnagar , Bareilly , India
| |
Collapse
|
34
|
Uemura K, Hayashi M, Itsubo T, Oishi A, Iwakawa H, Komatsu M, Uchiyama S, Kato H. Myostatin promotes tenogenic differentiation of C2C12 myoblast cells through Smad3. FEBS Open Bio 2017; 7:522-532. [PMID: 28396837 PMCID: PMC5377394 DOI: 10.1002/2211-5463.12200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 12/31/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
Myostatin, a member of the transforming growth factor-β (TGF-β) superfamily, is expressed in developing and adult skeletal muscle and negatively regulates skeletal muscle growth. Recently, myostatin has been found to be expressed in tendons and increases tendon fibroblast proliferation and the expression of tenocyte markers. C2C12 is a mouse myoblast cell line, which has the ability to transdifferentiate into osteoblast and adipocyte lineages. We hypothesized that myostatin is capable of inducing tenogenic differentiation of C2C12 cells. We found that the expression of scleraxis, a tendon progenitor cell marker, is much higher in C2C12 than in the multipotent mouse mesenchymal fibroblast cell line C3H10T1/2. In comparison with other growth factors, myostatin significantly up-regulated the expression of the tenogenic marker in C2C12 cells under serum-free culture conditions. Immunohistochemistry showed that myostatin inhibited myotube formation and promoted the formation of spindle-shaped cells expressing tenomodulin. We examined signaling pathways essential for tenogenic differentiation to clarify the mechanism of myostatin-induced differentiation of C2C12 into tenocytes. The expression of tenomodulin was significantly suppressed by treatment with the ALK inhibitor SB341542, in contrast to p38MAPK (SB203580) and MEK1 (PD98059) inhibitors. RNAi silencing of Smad3 significantly suppressed myostatin-induced tenomodulin expression. These results indicate that myostatin has a potential role in the induction of tenogenic differentiation of C2C12 cells, which have tendon progenitor cell characteristics, through activation of Smad3-mediated signaling.
Collapse
Affiliation(s)
- Kazutaka Uemura
- Department of Orthopaedic Surgery Shinshu University School of Medicine Matsumoto Japan
| | - Masanori Hayashi
- Department of Orthopaedic Surgery Shinshu University School of Medicine Matsumoto Japan
| | | | - Ayumu Oishi
- Department of Orthopaedic Surgery Shinshu University School of Medicine Matsumoto Japan
| | - Hiroko Iwakawa
- Department of Orthopaedic Surgery Shinshu University School of Medicine Matsumoto Japan
| | - Masatoshi Komatsu
- Department of Orthopaedic Surgery Shinshu University School of Medicine Matsumoto Japan
| | - Shigeharu Uchiyama
- Department of Orthopaedic Surgery Shinshu University School of Medicine Matsumoto Japan
| | - Hiroyuki Kato
- Department of Orthopaedic Surgery Shinshu University School of Medicine Matsumoto Japan
| |
Collapse
|
35
|
Delaney K, Kasprzycka P, Ciemerych MA, Zimowska M. The role of TGF-β1 during skeletal muscle regeneration. Cell Biol Int 2017; 41:706-715. [PMID: 28035727 DOI: 10.1002/cbin.10725] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/26/2016] [Indexed: 02/06/2023]
Abstract
The injury of adult skeletal muscle initiates series of well-coordinated events that lead to the efficient repair of the damaged tissue. Any disturbances during muscle myolysis or reconstruction may result in the unsuccessful regeneration, characterised by strong inflammatory response and formation of connective tissue, that is, fibrosis. The switch between proper regeneration of skeletal muscle and development of fibrosis is controlled by various factors. Amongst them are those belonging to the transforming growth factor β family. One of the TGF-β family members is TGF-β1, a multifunctional cytokine involved in the regulation of muscle repair via satellite cells activation, connective tissue formation, as well as regulation of the immune response intensity. Here, we present the role of TGF-β1 in myogenic differentiation and muscle repair. The understanding of the mechanisms controlling these processes can contribute to the better understanding of skeletal muscle atrophy and diseases which consequence is fibrosis disrupting muscle function.
Collapse
Affiliation(s)
- Kamila Delaney
- Faculty of Biology, Department of Cytology, Institute of Zoology, University of Warsaw, 1 Miecznikowa St., 02-096 Warsaw, Poland
| | - Paulina Kasprzycka
- Faculty of Biology, Department of Cytology, Institute of Zoology, University of Warsaw, 1 Miecznikowa St., 02-096 Warsaw, Poland
| | - Maria Anna Ciemerych
- Faculty of Biology, Department of Cytology, Institute of Zoology, University of Warsaw, 1 Miecznikowa St., 02-096 Warsaw, Poland
| | - Malgorzata Zimowska
- Faculty of Biology, Department of Cytology, Institute of Zoology, University of Warsaw, 1 Miecznikowa St., 02-096 Warsaw, Poland
| |
Collapse
|
36
|
Pasteuning-Vuhman S, Boertje-van der Meulen JW, van Putten M, Overzier M, Ten Dijke P, Kiełbasa SM, Arindrarto W, Wolterbeek R, Lezhnina KV, Ozerov IV, Aliper AM, Hoogaars WM, Aartsma-Rus A, Loomans CJM. New function of the myostatin/activin type I receptor (ALK4) as a mediator of muscle atrophy and muscle regeneration. FASEB J 2016; 31:238-255. [PMID: 27733450 PMCID: PMC5161514 DOI: 10.1096/fj.201600675r] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/22/2016] [Indexed: 12/13/2022]
Abstract
Skeletal muscle fibrosis and impaired muscle regeneration are major contributors to muscle wasting in Duchenne muscular dystrophy (DMD). Muscle growth is negatively regulated by myostatin (MSTN) and activins. Blockage of these pathways may improve muscle quality and function in DMD. Antisense oligonucleotides (AONs) were designed specifically to block the function of ALK4, a key receptor for the MSTN/activin pathway in skeletal muscle. AON-induced exon skipping resulted in specific Alk4 down-regulation, inhibition of MSTN activity, and increased myoblast differentiation in vitro. Unexpectedly, a marked decrease in muscle mass (10%) was found after Alk4 AON treatment in mdx mice. In line with in vitro results, muscle regeneration was stimulated, and muscle fiber size decreased markedly. Notably, when Alk4 was down-regulated in adult wild-type mice, muscle mass decreased even more. RNAseq analysis revealed dysregulated metabolic functions and signs of muscle atrophy. We conclude that ALK4 inhibition increases myogenesis but also regulates the tight balance of protein synthesis and degradation. Therefore, caution must be used when developing therapies that interfere with MSTN/activin pathways.—Pasteuning-Vuhman, S., Boertje-van der Meulen, J. W., van Putten, M., Overzier, M., ten Dijke, P., Kiełbasa, S. M., Arindrarto, W., Wolterbeek, R., Lezhnina, K. V., Ozerov, I. V., Aliper, A. M., Hoogaars, W. M., Aartsma-Rus, A., Loomans, C. J. M. New function of the myostatin/activin type I receptor (ALK4) as a mediator of muscle atrophy and muscle regeneration.
Collapse
Affiliation(s)
| | | | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Maurice Overzier
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular and Cell Biology Leiden University Medical Center, Leiden, The Netherlands.,Cancer Genomics Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Szymon M Kiełbasa
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Wibowo Arindrarto
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ron Wolterbeek
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ksenia V Lezhnina
- InSilico Medicine, Incorporated, Emerging Technology Centers, Johns Hopkins University, Baltimore, Maryland, USA; and
| | - Ivan V Ozerov
- InSilico Medicine, Incorporated, Emerging Technology Centers, Johns Hopkins University, Baltimore, Maryland, USA; and
| | - Aleksandr M Aliper
- InSilico Medicine, Incorporated, Emerging Technology Centers, Johns Hopkins University, Baltimore, Maryland, USA; and
| | - Willem M Hoogaars
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Move Research Institute Amsterdam, Vrije Universiteit (VU) Amsterdam, Amsterdam, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands;
| | - Cindy J M Loomans
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
37
|
Du X, Liu Y, Liu J, Zhang Q, Wang X. Evolution history of duplicated smad3 genes in teleost: insights from Japanese flounder, Paralichthys olivaceus. PeerJ 2016; 4:e2500. [PMID: 27703851 PMCID: PMC5045880 DOI: 10.7717/peerj.2500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/29/2016] [Indexed: 01/27/2023] Open
Abstract
Following the two rounds of whole-genome duplication (WGD) during deuterosome evolution, a third genome duplication occurred in the ray-fined fish lineage and is considered to be responsible for the teleost-specific lineage diversification and regulation mechanisms. As a receptor-regulated SMAD (R-SMAD), the function of SMAD3 was widely studied in mammals. However, limited information of its role or putative paralogs is available in ray-finned fishes. In this study, two SMAD3 paralogs were first identified in the transcriptome and genome of Japanese flounder (Paralichthys olivaceus). We also explored SMAD3 duplication in other selected species. Following identification, genomic structure, phylogenetic reconstruction, and synteny analyses performed by MrBayes and online bioinformatic tools confirmed that smad3a/3b most likely originated from the teleost-specific WGD. Additionally, selection pressure analysis and expression pattern of the two genes performed by PAML and quantitative real-time PCR (qRT-PCR) revealed evidence of subfunctionalization of the two SMAD3 paralogs in teleost. Our results indicate that two SMAD3 genes originate from teleost-specific WGD, remain transcriptionally active, and may have likely undergone subfunctionalization. This study provides novel insights to the evolution fates of smad3a/3b and draws attentions to future function analysis of SMAD3 gene family.
Collapse
Affiliation(s)
- Xinxin Du
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong, China
| | - Yuezhong Liu
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong, China
| | - Jinxiang Liu
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong, China
| | - Quanqi Zhang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong, China
| | - Xubo Wang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong, China
| |
Collapse
|
38
|
Marzuca-Nassr GN, Vitzel KF, De Sousa LG, Murata GM, Crisma AR, Rodrigues Junior CF, Abreu P, Torres RP, Mancini-Filho J, Hirabara SM, Newsholme P, Curi R. Effects of high EPA and high DHA fish oils on changes in signaling associated with protein metabolism induced by hindlimb suspension in rats. Physiol Rep 2016; 4:e12958. [PMID: 27650250 PMCID: PMC5037913 DOI: 10.14814/phy2.12958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 12/16/2022] Open
Abstract
The effects of either eicosapentaenoic (EPA)- or docosahexaenoic (DHA)-rich fish oils on hindlimb suspension (HS)-induced muscle disuse atrophy were compared. Daily oral supplementations (0.3 mL/100 g b.w.) with mineral oil (MO) or high EPA or high DHA fish oils were performed in adult rats. After 2 weeks, the animals were subjected to HS for further 2 weeks. The treatments were maintained alongside HS At the end of 4 weeks, we evaluated: body weight gain, muscle mass and fat depots, composition of fatty acids, cross-sectional areas (CSA) of the soleus muscle and soleus muscle fibers, activities of cathepsin L and 26S proteasome, and content of carbonylated proteins in the soleus muscle. Signaling pathway activities associated with protein synthesis (Akt, p70S6K, S6, 4EBP1, and GSK3-beta) and protein degradation (atrogin-1/MAFbx, and MuRF1) were evaluated. HS decreased muscle mass, CSA of soleus muscle and soleus muscle fibers, and altered signaling associated with protein synthesis (decreased) and protein degradation (increased). The treatment with either fish oil decreased the ratio of omega-6/omega-3 fatty acids and changed protein synthesis-associated signaling. EPA-rich fish oil attenuated the changes induced by HS on 26S proteasome activity, CSA of soleus muscle fibers, and levels of p-Akt, total p70S6K, p-p70S6K/total p70S6K, p-4EBP1, p-GSK3-beta, p-ERK2, and total ERK 1/2 proteins. DHA-rich fish oil attenuated the changes induced by HS on p-4EBP1 and total ERK1 levels. The effects of EPA-rich fish oil on protein synthesis signaling were more pronounced. Both EPA- and DHA-rich fish oils did not impact skeletal muscle mass loss induced by non-inflammatory HS.
Collapse
Affiliation(s)
- Gabriel Nasri Marzuca-Nassr
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Kaio Fernando Vitzel
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil Massey Institute of Food Science and Technology, College of Health Massey University, Albany, New Zealand
| | - Luís Gustavo De Sousa
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Gilson M Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Amanda Rabello Crisma
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | | | - Phablo Abreu
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| | - Rosângela Pavan Torres
- Department of Lipids Laboratory, Food Science & Nutrition, Faculty of Pharmaceutical Science University of São Paulo, São Paulo, Brazil
| | - Jorge Mancini-Filho
- Department of Lipids Laboratory, Food Science & Nutrition, Faculty of Pharmaceutical Science University of São Paulo, São Paulo, Brazil
| | - Sandro M Hirabara
- Institute of Physical Activity Sciences and Sport, Cruzeiro do Sul University, São Paulo, Brazil
| | - Philip Newsholme
- School of Biomedical Sciences, CHIRI Biosciences Curtin University, Perth, Australia
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences University of São Paulo, São Paulo, Brazil
| |
Collapse
|
39
|
Myostatin signals through miR-34a to regulate Fndc5 expression and browning of white adipocytes. Int J Obes (Lond) 2016; 41:137-148. [PMID: 27297797 PMCID: PMC5220162 DOI: 10.1038/ijo.2016.110] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND/OBJECTIVES Myostatin (Mstn) has a pivotal role in glucose and lipid metabolism. Mstn deficiency leads to the increased browning of white adipose tissue (WAT), which results in the increased energy expenditure and protection against diet-induced obesity and insulin resistance. In this study, we investigated the molecular mechanism(s) through which Mstn regulates browning of white adipocytes. METHODS Quantitative molecular analyses were performed to assess Mstn regulation of miR-34a and Fndc5 expression. miR-34a was overexpressed and repressed to investigate miR-34a regulation of Fndc5. Luciferase reporter analysis verified direct binding between miR-34a and the Fndc5 3'-untranslated region (UTR). The browning phenotype of Mstn-/- adipocytes was assessed through the analysis of brown fat marker gene expression, mitochondrial function and infrared thermography. The role of miR-34a and Fndc5 in this browning phenotype was verified through antibody-mediated neutralization of FNDC5, knockdown of Fndc5 by small interfering RNA and through miR-34a gain-of-function and loss-of-function experiments. RESULTS Mstn treatment of myoblasts inhibited Fndc5 expression, whereas the loss of Mstn increased Fndc5 levels in muscles and in circulation. Mstn inhibition of Fndc5 is miR-34a dependent. Mstn treatment of C2C12 myoblasts upregulated miR-34a expression, whereas reduced miR-34a expression was noted in Mstn-/- muscle and WAT. Subsequent overexpression of miR-34a inhibited Fndc5 expression, whereas blockade of miR-34a increased Fndc5 expression in myoblasts. Reporter analysis revealed that miR-34a directly suppresses Fndc5 expression through a miR-34a-specific binding site within the Fndc5 3'UTR. Importantly, Mstn-mediated inhibition of Fndc5 was blocked upon miR-34a inhibition. Mstn-/- adipocytes showed reduced miR-34a, enhanced Fndc5 expression and increased thermogenic gene expression, which was reversed upon either neutralization of Fndc5 or Fndc5 knockdown. In agreement, Mstn-/- adipocytes have increased mitochondria, improved mitochondrial function and increased heat production. CONCLUSIONS Mstn regulates Fndc5/Irisin expression and secretion through a novel miR-34a-dependent post-transcriptional mechanism. Loss of Mstn in mice leads to the increased Fndc5/Irisin expression, which contributes to the browning of white adipocytes.
Collapse
|
40
|
Kim CH, Shin JH, Hwang SJ, Choi YH, Kim DS, Kim CM. Schisandrae fructus enhances myogenic differentiation and inhibits atrophy through protein synthesis in human myotubes. Int J Nanomedicine 2016; 11:2407-15. [PMID: 27330287 PMCID: PMC4898430 DOI: 10.2147/ijn.s101299] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Schisandrae fructus (SF) has recently been reported to increase skeletal muscle mass and inhibit atrophy in mice. We investigated the effect of SF extract on human myotube differentiation and its acting pathway. Various concentrations (0.1–10 μg/mL) of SF extract were applied on human skeletal muscle cells in vitro. Myotube area and fusion index were measured to quantify myotube differentiation. The maximum effect was observed at 0.5 μg/mL of SF extract, enhancing differentiation up to 1.4-fold in fusion index and 1.6-fold in myotube area at 8 days after induction of differentiation compared to control. Phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1 and 70 kDa ribosomal protein S6 kinase, which initiate translation as downstream of mammalian target of rapamycin pathway, was upregulated in early phases of differentiation after SF treatment. SF also attenuated dexamethasone-induced atrophy. In conclusion, we show that SF augments myogenic differentiation and attenuates atrophy by increasing protein synthesis through mammalian target of rapamycin/70 kDa ribosomal protein S6 kinase and eukaryotic translation initiation factor 4E-binding protein 1 signaling pathway in human myotubes. SF can be a useful natural dietary supplement in increasing skeletal muscle mass, especially in the aged with sarcopenia and the patients with disuse atrophy.
Collapse
Affiliation(s)
- Cy Hyun Kim
- Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea; Center for Anti-Aging Industry, Pusan National University, Busan, Republic of Korea
| | - Jin-Hong Shin
- Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea; Department of Neurology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Sung Jun Hwang
- Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea; Center for Anti-Aging Industry, Pusan National University, Busan, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| | - Dae-Seong Kim
- Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea; Department of Neurology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Cheol Min Kim
- Center for Anti-Aging Industry, Pusan National University, Busan, Republic of Korea; Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Republic of Korea
| |
Collapse
|
41
|
Busato KC, Gomes RA, Ladeira MM, Duarte MS, Freitas NC, Rodrigues AC, Chalfun-Junior A, Paiva LV, Chizzotti ML. Expression of genes related to the regulation of muscle protein turnover in Angus and Nellore bulls. J Anim Sci 2016; 94:1472-81. [PMID: 27136006 DOI: 10.2527/jas.2015-9924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We aimed to evaluate the expression of genes related to the regulation of muscle protein turnover in the longissimus dorsi (LD) muscle of Angus and Nellore bulls and to estimate the within-breed correlations of gene expression and performance traits. Thirteen genes related to the IGF-1 and myostatin pathways were studied. Thirteen animals, with an initial average BW of 381.2 ± 11.8 kg, from each breed were used in a completely randomized 2 × 2 factorial design (2 breeds and 2 feeding levels). The diet consisted of corn silage and a corn-soybean meal concentrate in a roughage-to-concentrate ratio of 30:70. Cattle were fed ad libitum (with 9 animals from each breed) or feed restricted (a 55% restriction of total DMI of ad libitum-fed animals, calculated as percentage of metabolic BW, with 4 animals of each breed). The experimental period lasted for 82 d and it was preceded by a 28-d adaptation period. The performance traits evaluated were slaughter body weight, total ADG (from d 1 to 82 of the trial), initial ADG (from d 1 to 41 of the trial), final ADG (from d 42 to 82 of the trial), total DMI (from d 1 to 82 of the trial), initial DMI (from d 1 to 41 of the trial), final DMI (from d 42 to 82 of the trial), HCW, LD weight (LDW), and rib eye area (REA). After slaughter, samples were taken from the LD muscle between the 12th and 13th ribs for gene expression analysis by quantitative reverse transcription PCR. There was no difference ( > 0.05) in the expression of any of the genes studied between ad libitum-fed Angus and ad libitum-fed Nellore, whereas feed restriction increased the expression of (; < 0.001), (; = 0.05), and (; = 0.04) and decreased the expression of ( < 0.01). The REA was negatively correlated to (; = 0.01), (; = 0.02), and ( = 0.05). The HCW was negatively correlated to ( = 0.01) and ( = 0.01) and tended to be negatively correlated to ( = 0.07), whereas the LDW tended to be negatively correlated to ( = 0.08). The genes , , and seem to be important for muscle growth and may be worthy of further investigation as future strategies for increasing muscle in livestock.
Collapse
|
42
|
Zhang Y. Potential therapeutic targets from genetic and epigenetic approaches for asthma. World J Transl Med 2016; 5:14-25. [DOI: 10.5528/wjtm.v5.i1.14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023] Open
Abstract
Asthma is a complex disorder characterised by inflammation of airway and symptoms of wheeze and shortness of breath. Allergic asthma, atopic dermatitis and allergic rhinitis are immunoglobulin E (IgE) related diseases. Current therapies targeting asthma rely on non-specific medication to control airway inflammation and prevent symptoms. Severe asthma remains difficult to treat. Genetic and genomic approaches of asthma and IgE identified many novel loci underling the disease pathophysiology. Recent epigenetic approaches also revealed the insights of DNA methylation and chromatin modification on histones in asthma and IgE. More than 30 microRNAs have been identified to have regulating roles in asthma. Understanding the pathways of the novel genetic loci and epigenetic elements in asthma and IgE will provide new therapeutic means for clinical management of the disease in future.
Collapse
|
43
|
Huang QK, Qiao HY, Fu MH, Li G, Li WB, Chen Z, Wei J, Liang BS. MiR-206 Attenuates Denervation-Induced Skeletal Muscle Atrophy in Rats Through Regulation of Satellite Cell Differentiation via TGF-β1, Smad3, and HDAC4 Signaling. Med Sci Monit 2016; 22:1161-70. [PMID: 27054781 PMCID: PMC4829125 DOI: 10.12659/msm.897909] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Denervation-induced skeletal muscle atrophy results in significant biochemical and physiological changes potentially leading to devastating outcomes including increased mortality. Effective treatments for skeletal muscle diseases are currently not available. Muscle-specific miRNAs, such as miR-206, play an important role in the regulation of muscle regeneration. The aim of the present study was to examine the beneficial effects of miR-206 treatment during the early changes in skeletal muscle atrophy, and to study the underlying signaling pathways in a rat skeletal muscle atrophy model. Material/Methods The rat denervation-induced skeletal muscle atrophy model was established. miRNA-206 was overexpressed with or without TGF-β1 inhibitor in the rats. The mRNA and protein expression of HDAC4, TGF-β1, and Smad3 was determined by real-time PCR and western blot. The gastrocnemius muscle cross-sectional area and relative muscle mass were measured. MyoD1, TGF-β1, and Pax7 were determined by immunohistochemical staining. Results After sciatic nerve surgical transection, basic muscle characteristics, such as relative muscle weight, deteriorated continuously during a 2-week period. Injection of miR-206 (30 μg/rat) attenuated morphological and physiological deterioration of muscle characteristics, prevented fibrosis effectively, and inhibited the expression of TGF-β1 and HDAC4 as assessed 2 weeks after denervation. Moreover, miR-206 treatment increased the number of differentiating (MyoD1+/Pax7+) satellite cells, thereby protecting denervated muscles from atrophy. Interestingly, the ability of miR-206 to govern HDAC4 expression and to attenuate muscle atrophy was weakened after pharmacological blockage of the TGF-β1/Smad3 axis. Conclusions TGF-β1/Smad3 signaling pathway is one of the crucial signaling pathways by which miR-206 counteracts skeletal muscle atrophy by affecting proliferation and differentiation of satellite cells. miR-206 may be a potential target for development of a new strategy for treatment of patients with early denervation-induced skeletal muscle atrophy.
Collapse
Affiliation(s)
- Qiang Kai Huang
- Department of Orthopedics, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Hu-Yuan Qiao
- Department of Orthopedics, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Ming-Huan Fu
- Division of Cardiovascular Disease, Department of Gerontology, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China (mainland)
| | - Gang Li
- Department of Orthopedics, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Wen-Bin Li
- Department of Orthopedics, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Zhi Chen
- Department of Orthopedics, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Jian Wei
- Department of Orthopedics, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Bing-Sheng Liang
- Department of Orthopaedics, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| |
Collapse
|
44
|
Shi T, Peng W, Yan J, Cai H, Lan X, Lei C, Bai Y, Chen H. A novel 17 bp indel in the <i>SMAD3</i> gene alters transcription level, contributing to phenotypic traits in Chinese cattle. Arch Anim Breed 2016. [DOI: 10.5194/aab-59-151-2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Abstract. SMAD3, the messenger of the transforming growth factor beta (TGF-β) signaling pathway, plays essential roles in myogenesis and osteogenesis and may relate to the regulation of body weight. In this study, a 17 bp indel (NC_007308: g.101893_101909insGAGGATGAGTGCTCCAG) in intron3 of the SMAD3 gene was detected in four Chinese cattle breeds (Qinchuan, Jiaxian, Nanyang and Caoyuan) by using DNA pool sequencing, and its effects on gene expression and growth traits were analyzed in Qinchuan and Caoyuan cattle. The results showed that the indel locus was significantly associated with SMAD3 transcriptional levels where II genotypes had a higher value than DD genotypes in Qinchuan (QC) cattle muscle tissue (P < 0.05). In addition, the locus was strongly associated with chest girth, chest width, rump length, hucklebone width and body weight in 2-year-old QC cattle (P < 0.05) and body weight (12 months), body height (18 months) and chest girth (18 months) in Caoyuan cattle (P < 0.5). To the best of our knowledge, this is the first evidence of the association between SMAD3 indel and cattle phenotype, and it may contribute to understanding the function of the indel, which could be a promising marker for beef cattle breeding.
Collapse
|
45
|
Wang X, Shen QW, Wang J, Zhang Z, Feng F, Chen T, Zhang Y, Wei H, Li Z, Wang X, Wang Y. KLF7 Regulates Satellite Cell Quiescence in Response to Extracellular Signaling. Stem Cells 2016; 34:1310-20. [PMID: 26930448 DOI: 10.1002/stem.2346] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 11/12/2015] [Indexed: 11/11/2022]
Abstract
Retaining muscle stem satellite cell (SC) quiescence is important for the maintenance of stem cell population and tissue regeneration. Accumulating evidence supports the model where key extracellular signals play crucial roles in maintaining SC quiescence or activation, however, the intracellular mechanisms that mediate niche signals to control SC behavior are not fully understood. Here, we reported that KLF7 functioned as a key mediator involved in low-level TGF-β signaling and canonical Notch signaling-induced SC quiescence and myoblast arrest. The data obtained showed that KLF7 was upregulated in quiescent SCs and nonproliferating myoblasts. Silence of KLF7 promoted SCs activation and myoblasts proliferation, but overexpression of KLF7 induced myogenic cell arrest. Notably, the expression of KLF7 was regulated by TGF-β and Notch3 signaling. Knockdown of KLF7 diminished low-level TGF-β and canonical Notch signaling-induced SC quiescence. Investigation into the mechanism revealed that KLF7 regulation of SC function was dependent on p21 and acetylation of Lys227 and/or 231 in the DNA binding domain of KLF7. Our study provides new insights into the regulatory network of muscle stem cell quiescence. Stem Cells 2016;34:1310-1320.
Collapse
Affiliation(s)
- Xiaobin Wang
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Qingwu W Shen
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China.,College of Food Science and Technology, Hunan Agricultural University, Changsha, Hunan, People's Republic of China
| | - Jie Wang
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Zhiguo Zhang
- College of Food Science and Engineering, Qilu University of Technology, Jinan, Shandong, People's Republic of China
| | - Fu Feng
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Ting Chen
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Yanyan Zhang
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Huan Wei
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Zhongwen Li
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
46
|
Abrigo J, Rivera JC, Simon F, Cabrera D, Cabello-Verrugio C. Transforming growth factor type beta (TGF-β) requires reactive oxygen species to induce skeletal muscle atrophy. Cell Signal 2016; 28:366-376. [PMID: 26825874 DOI: 10.1016/j.cellsig.2016.01.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 12/28/2015] [Accepted: 01/24/2016] [Indexed: 12/13/2022]
Abstract
Transforming growth factor beta 1 (TGF-β1) is a classical modulator of skeletal muscle and regulates several processes, such as myogenesis, regeneration, and muscle function in skeletal muscle diseases. Skeletal muscle atrophy, characterised by the loss of muscle strength and mass, is one of the pathological conditions regulated by TGF-β. Atrophy also results in increased myosin heavy chain (MHC) degradation and the expression of two muscle-specific E3 ubiquitin ligases, atrogin-1 and MuRF-1. Reactive oxygen species (ROS) are modulators of muscle wasting, and NAD(P)H oxidase (NOX) is one of the main sources of ROS. While it was recently found that TGF-β1 induces atrophy in skeletal muscle, the underlying mechanism is not fully understood. In this study, the role of NOX-derived ROS in skeletal muscle atrophy induced by TGF-β was assessed. TGF-β1 induced an atrophic effect in C2C12 myotubes, as evidenced by decreased myotube diameter and MHC levels, together with increased MuRF-1 levels. Concomitantly, TGF-β increased NOX-induced ROS contents. Interestingly, NOX inhibition through apocynin and the antioxidant treatment with N-acetyl cysteine (NAC) decreased increased ROS levels in myotubes. Additionally, both apocynin and NAC completely prevented the decreased MHC, decreased myotube diameter, and increased MuRF-1 induced by TGF-β. Injection of TGF-β1 into the tibialis anterior muscle induced atrophy, as observed by decreased fibre diameter and MHC levels, together with increased MuRF-1 levels. Likewise, TGF-β increased the ROS contents in the smaller fibres of skeletal muscle. Additionally, the administration of NAC to mice prevented all atrophic effects and the increase in ROS induced by TGF-β in the tibialis anterior. This is the first study to report that TGF-β has an atrophic effect dependent on NOX-induced ROS in skeletal muscle.
Collapse
Affiliation(s)
- Johanna Abrigo
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Juan Carlos Rivera
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Laboratory of Integrative Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O Higgins, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Biology and Molecular Physiopathology, Department of Biological Sciences, Faculty of Biological Sciences, Faculty of Medicine, Universidad Andrés Bello, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| |
Collapse
|
47
|
Prmt5 is a regulator of muscle stem cell expansion in adult mice. Nat Commun 2015; 6:7140. [PMID: 26028225 PMCID: PMC4458870 DOI: 10.1038/ncomms8140] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 04/09/2015] [Indexed: 01/16/2023] Open
Abstract
Skeletal muscle stem cells (MuSC), also called satellite cells, are indispensable for maintenance and regeneration of adult skeletal muscles. Yet, a comprehensive picture of the regulatory events controlling the fate of MuSC is missing. Here, we determine the proteome of MuSC to design a loss-of-function screen, and identify 120 genes important for MuSC function including the arginine methyltransferase Prmt5. MuSC-specific inactivation of Prmt5 in adult mice prevents expansion of MuSC, abolishes long-term MuSC maintenance and abrogates skeletal muscle regeneration. Interestingly, Prmt5 is dispensable for proliferation and differentiation of Pax7+ myogenic progenitor cells during mouse embryonic development, indicating significant differences between embryonic and adult myogenesis. Mechanistic studies reveal that Prmt5 controls proliferation of adult MuSC by direct epigenetic silencing of the cell cycle inhibitor p21. We reason that Prmt5 generates a poised state that keeps MuSC in a standby mode, thus allowing rapid MuSC amplification under disease conditions. Skeletal muscle satellite cells are important for muscle regeneration, but their regulatory mechanisms are largely unknown. Here the authors identify arginine methyltransferase Prmt5 as a key regulator of satellite cell maintenance and function in adult mice, and show that Prmt5 acts mainly but not exclusively on the cell cycle inhibitor p21.
Collapse
|
48
|
Lamarche É, Lala-Tabbert N, Gunanayagam A, St-Louis C, Wiper-Bergeron N. Retinoic acid promotes myogenesis in myoblasts by antagonizing transforming growth factor-beta signaling via C/EBPβ. Skelet Muscle 2015; 5:8. [PMID: 25878769 PMCID: PMC4397812 DOI: 10.1186/s13395-015-0032-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 02/18/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The effects of transforming growth factor-beta (TGFβ) are mediated by the transcription factors Smad2 and Smad3. During adult skeletal myogenesis, TGFβ signaling inhibits the differentiation of myoblasts, and this can be reversed by treatment with retinoic acid (RA). In mesenchymal stem cells and preadipocytes, RA treatment can function in a non-classical manner by stimulating the expression of Smad3. Smad3 can bind to and prevent the bzip transcription factor CCAAT/enhancer-binding protein beta (C/EBPβ) from binding DNA response elements in target promoters, thereby affecting cell differentiation. In skeletal muscle, C/EBPβ is highly expressed in satellite cells and myoblasts and is downregulated during differentiation. Persistent expression of C/EBPβ in myoblasts inhibits their differentiation. METHODS Using both C2C12 myoblasts and primary myoblasts, we examined the regulation of C/EBPβ expression and activity following treatment with TGFβ and RA. RESULTS We demonstrate that treatment with RA upregulates Smad3, but not Smad2 expression in myoblasts, and can partially rescue the block of differentiation induced by TGFβ. RA treatment reduces C/EBPβ occupancy of the Pax7 and Smad2 promoters and decreased their expression. RA also inhibits the TGFβ-mediated phosphorylation of Smad2, which may also contribute to its pro-myogenic activities. TGFβ treatment of C2C12 myoblasts stimulates C/EBPβ expression, which in turn can stimulate Pax7 and Smad2 expression, and inhibits myogenesis. Loss of C/EBPβ expression in myoblasts partially restores differentiation in the presence of TGFβ. CONCLUSIONS TGFβ acts, at least in part, to inhibit myogenesis by upregulating the expression of C/EBPβ, as treatment with RA or loss of C/EBPβ can partially rescue differentiation in TGFβ-treated cells. This work identifies a pro-myogenic role for Smad3, through the inhibition of C/EBPβ's actions in myoblasts, and reveals mechanisms of crosstalk between RA and TGFβ signaling pathways.
Collapse
Affiliation(s)
- Émilie Lamarche
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario Canada
| | - Neena Lala-Tabbert
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario Canada
| | - Angelo Gunanayagam
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario Canada
| | - Catherine St-Louis
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario Canada
| | - Nadine Wiper-Bergeron
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario Canada
| |
Collapse
|
49
|
Parker MH. The altered fate of aging satellite cells is determined by signaling and epigenetic changes. Front Genet 2015; 6:59. [PMID: 25750654 PMCID: PMC4335604 DOI: 10.3389/fgene.2015.00059] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/07/2015] [Indexed: 01/11/2023] Open
Abstract
Skeletal muscle is a striated tissue composed of multinucleated fibers that contract under the control of the somatic nervous system to direct movement. The stem cells of skeletal muscle, known as satellite cells, are responsible for muscle fiber growth, turnover, and regeneration. Satellite cells are activated and proliferate in response to stimuli, and simplistically, have two main fates—to repopulate the satellite cell niche, or differentiate to regenerate or repair muscle fibers. However, the ability to regenerate muscle and replace lost myofibers declines with age. This loss of function may be a result of extrinsic changes in the niche, such as alterations in signaling or modifications to the extracellular matrix. However, intrinsic epigenetic changes within satellite cells may also affect cell fate and cause a decline in regenerative capacity. This review will describe the mechanisms that regulate cell fate decisions in adult skeletal muscle, and how changes during aging affect muscle fiber turnover and regeneration.
Collapse
Affiliation(s)
- Maura H Parker
- Clinical Research Division, Fred Hutchinson Cancer Research Center , Seattle, WA, USA
| |
Collapse
|
50
|
Feeney SJ, McGrath MJ, Sriratana A, Gehrig SM, Lynch GS, D’Arcy CE, Price JT, McLean CA, Tupler R, Mitchell CA. FHL1 reduces dystrophy in transgenic mice overexpressing FSHD muscular dystrophy region gene 1 (FRG1). PLoS One 2015; 10:e0117665. [PMID: 25695429 PMCID: PMC4335040 DOI: 10.1371/journal.pone.0117665] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 12/29/2014] [Indexed: 01/01/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal-dominant disease with no effective treatment. The genetic cause of FSHD is complex and the primary pathogenic insult underlying the muscle disease is unknown. Several disease candidate genes have been proposed including DUX4 and FRG1. Expression analysis studies of FSHD report the deregulation of genes which mediate myoblast differentiation and fusion. Transgenic mice overexpressing FRG1 recapitulate the FSHD muscular dystrophy phenotype. Our current study selectively examines how increased expression of FRG1 may contribute to myoblast differentiation defects. We generated stable C2C12 cell lines overexpressing FRG1, which exhibited a myoblast fusion defect upon differentiation. To determine if myoblast fusion defects contribute to the FRG1 mouse dystrophic phenotype, this strain was crossed with skeletal muscle specific FHL1-transgenic mice. We previously reported that FHL1 promotes myoblast fusion in vitro and FHL1-transgenic mice develop skeletal muscle hypertrophy. In the current study, FRG1 mice overexpressing FHL1 showed an improvement in the dystrophic phenotype, including a reduced spinal kyphosis, increased muscle mass and myofiber size, and decreased muscle fibrosis. FHL1 expression in FRG1 mice, did not alter satellite cell number or activation, but enhanced myoblast fusion. Primary myoblasts isolated from FRG1 mice showed a myoblast fusion defect that was rescued by FHL1 expression. Therefore, increased FRG1 expression may contribute to a muscular dystrophy phenotype resembling FSHD by impairing myoblast fusion, a defect that can be rescued by enhanced myoblast fusion via expression of FHL1.
Collapse
Affiliation(s)
- Sandra J. Feeney
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Meagan J. McGrath
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Absorn Sriratana
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Stefan M. Gehrig
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, 3010, Australia
| | - Gordon S. Lynch
- Basic and Clinical Myology Laboratory, Department of Physiology, The University of Melbourne, Victoria, 3010, Australia
| | - Colleen E. D’Arcy
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - John T. Price
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Victoria, 8001, Australia
| | - Catriona A. McLean
- Department of Anatomical Pathology, Alfred Hospital, Prahran, Victoria, 3004, Australia
- Department of Medicine, Central Clinical School, Monash University, Clayton, VIC, 3800, Australia
| | - Rossella Tupler
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, MA, 01655, United States of America
- Dipartimento di Scienze della Vita, Universita di Modena e Reggio Emilia, 41125, Modena, Italy
| | - Christina A. Mitchell
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
- * E-mail:
| |
Collapse
|