51
|
Milanesi A, Lee JW, Kim NH, Liu YY, Yang A, Sedrakyan S, Kahng A, Cervantes V, Tripuraneni N, Cheng SY, Perin L, Brent GA. Thyroid Hormone Receptor α Plays an Essential Role in Male Skeletal Muscle Myoblast Proliferation, Differentiation, and Response to Injury. Endocrinology 2016; 157:4-15. [PMID: 26451739 PMCID: PMC4701883 DOI: 10.1210/en.2015-1443] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Thyroid hormone plays an essential role in myogenesis, the process required for skeletal muscle development and repair, although the mechanisms have not been established. Skeletal muscle develops from the fusion of precursor myoblasts into myofibers. We have used the C2C12 skeletal muscle myoblast cell line, primary myoblasts, and mouse models of resistance to thyroid hormone (RTH) α and β, to determine the role of thyroid hormone in the regulation of myoblast differentiation. T3, which activates thyroid hormone receptor (TR) α and β, increased myoblast differentiation whereas GC1, a selective TRβ agonist, was minimally effective. Genetic approaches confirmed that TRα plays an important role in normal myoblast proliferation and differentiation and acts through the Wnt/β-catenin signaling pathway. Myoblasts with TRα knockdown, or derived from RTH-TRα PV (a frame-shift mutation) mice, displayed reduced proliferation and myogenic differentiation. Moreover, skeletal muscle from the TRα1PV mutant mouse had impaired in vivo regeneration after injury. RTH-TRβ PV mutant mouse model skeletal muscle and derived primary myoblasts did not have altered proliferation, myogenic differentiation, or response to injury when compared with control. In conclusion, TRα plays an essential role in myoblast homeostasis and provides a potential therapeutic target to enhance skeletal muscle regeneration.
Collapse
Affiliation(s)
- Anna Milanesi
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Jang-Won Lee
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Nam-Ho Kim
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Yan-Yun Liu
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - An Yang
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Sargis Sedrakyan
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Andrew Kahng
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Vanessa Cervantes
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Nikita Tripuraneni
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Sheue-yann Cheng
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Laura Perin
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| | - Gregory A Brent
- Department of Medicine (A.M., Y.-Y.L., A.Y., G.A.B.), Veterans Affairs Greater Los Angeles Healthcare System, and Departments of Medicine and Physiology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California 90073; Department of Neurosurgery (J.-W.L., N.-H.K., A.K., V.C.), Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Urology (S.S., N.T., L.P.), Children's Hospital Los Angeles, University of Southern California, Los Angeles, California 90027; and National Cancer Institute (S.C.), Bethesda, Maryland 20892
| |
Collapse
|
52
|
Joanisse S, Parise G. Cytokine Mediated Control of Muscle Stem Cell Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:27-44. [DOI: 10.1007/978-3-319-27511-6_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
53
|
Fu X, Zhu M, Zhang S, Foretz M, Viollet B, Du M. Obesity Impairs Skeletal Muscle Regeneration Through Inhibition of AMPK. Diabetes 2016; 65:188-200. [PMID: 26384382 PMCID: PMC4686944 DOI: 10.2337/db15-0647] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/10/2015] [Indexed: 12/18/2022]
Abstract
Obesity is increasing rapidly worldwide and is accompanied by many complications, including impaired muscle regeneration. The obese condition is known to inhibit AMPK activity in multiple tissues. We hypothesized that the loss of AMPK activity is a major reason for hampered muscle regeneration in obese subjects. We found that obesity inhibits AMPK activity in regenerating muscle, which was associated with impeded satellite cell activation and impaired muscle regeneration. To test the mediatory role of AMPKα1, we knocked out AMPKα1 and found that both proliferation and differentiation of satellite cells are reduced after injury and that muscle regeneration is severely impeded, reminiscent of hampered muscle regeneration seen in obese subjects. Transplanted satellite cells with AMPKα1 deficiency had severely impaired myogenic capacity in regenerating muscle fibers. We also found that attenuated muscle regeneration in obese mice is rescued by AICAR, a drug that specifically activates AMPK, but AICAR treatment failed to improve muscle regeneration in obese mice with satellite cell-specific AMPKα1 knockout, demonstrating the importance of AMPKα1 in satellite cell activation and muscle regeneration. In summary, AMPKα1 is a key mediator linking obesity and impaired muscle regeneration, providing a convenient drug target to facilitate muscle regeneration in obese populations.
Collapse
Affiliation(s)
- Xing Fu
- Washington Center for Muscle Biology, Department of Animal Sciences and Department of Pharmaceutical Sciences, Washington State University, Pullman, WA
| | - Meijun Zhu
- School of Food Science, Washington State University, Pullman, WA
| | - Shuming Zhang
- School of Food Science, Washington State University, Pullman, WA
| | - Marc Foretz
- INSERM U1016, Institut Cochin, Paris, France Université Paris Descartes, Sorbonne Paris Cité, Paris, France CNRS UMR 8104, Paris, France
| | - Benoit Viollet
- INSERM U1016, Institut Cochin, Paris, France Université Paris Descartes, Sorbonne Paris Cité, Paris, France CNRS UMR 8104, Paris, France
| | - Min Du
- Washington Center for Muscle Biology, Department of Animal Sciences and Department of Pharmaceutical Sciences, Washington State University, Pullman, WA
| |
Collapse
|
54
|
Carvajal Monroy PL, Grefte S, Kuijpers-Jagtman AM, Helmich MP, Wagener FA, Von den Hoff JW. Fibrosis impairs the formation of new myofibers in the soft palate after injury. Wound Repair Regen 2015. [DOI: 10.1111/wrr.12345] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Paola L. Carvajal Monroy
- Department of Orthodontics and Craniofacial Biology; Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center; Nijmegen The Netherlands
| | - Sander Grefte
- Department of Human and Animal Physiology; Wageningen University; Wageningen The Netherlands
| | - Anne Marie Kuijpers-Jagtman
- Department of Orthodontics and Craniofacial Biology; Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center; Nijmegen The Netherlands
| | - Maria P.A.C. Helmich
- Department of Orthodontics and Craniofacial Biology; Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center; Nijmegen The Netherlands
| | - Frank A.D.T.G. Wagener
- Department of Orthodontics and Craniofacial Biology; Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center; Nijmegen The Netherlands
| | - Johannes W. Von den Hoff
- Department of Orthodontics and Craniofacial Biology; Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center; Nijmegen The Netherlands
| |
Collapse
|
55
|
Piccoli M, Urbani L, Alvarez-Fallas ME, Franzin C, Dedja A, Bertin E, Zuccolotto G, Rosato A, Pavan P, Elvassore N, De Coppi P, Pozzobon M. Improvement of diaphragmatic performance through orthotopic application of decellularized extracellular matrix patch. Biomaterials 2015; 74:245-55. [PMID: 26461117 DOI: 10.1016/j.biomaterials.2015.10.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 12/15/2022]
Abstract
Muscle tissue engineering can provide support to large congenital skeletal muscle defects using scaffolds able to allow cell migration, proliferation and differentiation. Acellular extracellular matrix (ECM) scaffold can generate a positive inflammatory response through the activation of anti-inflammatory T-cell populations and M2 polarized macrophages that together lead to a local pro-regenerative environment. This immunoregulatory effect is maintained when acellular matrices are transplanted in a xenogeneic setting, but it remains unclear whether it can be therapeutic in a model of muscle diseases. We demonstrated here for the first time that orthotopic transplantation of a decellularized diaphragmatic muscle from wild animals promoted tissue functional recovery in an established atrophic mouse model. In particular, ECM supported a local immunoresponse activating a pro-regenerative environment and stimulating host muscle progenitor cell activation and migration. These results indicate that acellular scaffolds may represent a suitable regenerative medicine option for improving performance of diseased muscles.
Collapse
Affiliation(s)
- M Piccoli
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.
| | - L Urbani
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Institute of Child Health and Great Ormond Street Hospital, London, United Kingdom.
| | - M E Alvarez-Fallas
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - C Franzin
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - A Dedja
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - E Bertin
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - G Zuccolotto
- Department of Medicine, University of Padua, Padua, Italy
| | - A Rosato
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - P Pavan
- Department of Industrial Engineering, University of Padua, Padua, Italy; Centre for Mechanics of Biological Materials, University of Padua, Padua, Italy
| | - N Elvassore
- Department of Industrial Engineering, University of Padua, and Venetian Institute of Molecular Medicine, Padua, Italy
| | - P De Coppi
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Institute of Child Health and Great Ormond Street Hospital, London, United Kingdom.
| | - M Pozzobon
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.
| |
Collapse
|
56
|
Abstract
UNLABELLED Skeletal muscle satellite cells (SCs) are involved in muscle growth and repair. However, clarification of their behavior in hibernating mammals is lacking. The aim of this study was to quantify SCs and total myonuclei in hibernator muscle during different phases of the torpor-arousal cycle. Skeletal muscle was collected from thirteen-lined ground squirrels, Ictidomys tridecemlineatus, at five timepoints during hibernation: control euthermic [CON, stable body temperature (Tb)], early torpor (ET, within 24h), late torpor (LT, 5+ consecutive days), early arousal (EA, increased respiratory rate >60 breaths/min, Tb 9-12°C) and interbout arousal (IA, euthermic Tb). Protein levels of p21, Myf5, Wnt4, and β-catenin were determined by western blotting. SCs (Pax7(+)) and myonuclei were identified using immunohistochemistry. Over the torpor-arousal cycle, myonuclei/fiber remained unchanged. However, the percentage of SCs increased significantly during ET (7.35±1.04% vs. CONTROL 4.18±0.58%; p<0.05) and returned to control levels during LT. This coincided with a 224% increase in p21 protein during ET. Protein levels of Wnt4 did not change throughout, whereas Myf5 was lower during EA (p<0.08) and IA (p<0.05). Compared to torpor, β-catenin increased by 247% and 279% during EA and IA, respectively (p<0.05). In conclusion, SCs were not dormant during hibernation and increased numbers of SC during ET corresponded with elevated amounts of p21 suggesting that cell cycle control may explain the SC return to baseline levels during late torpor. Despite relatively low Tb during early arousal, active control of quiescence by Myf5 is reduced.
Collapse
|
57
|
Carvajal Monroy PL, Yablonka-Reuveni Z, Grefte S, Kuijpers-Jagtman AM, Wagener FADTG, Von den Hoff JW. Isolation and Characterization of Satellite Cells from Rat Head Branchiomeric Muscles. J Vis Exp 2015:e52802. [PMID: 26274878 PMCID: PMC4544364 DOI: 10.3791/52802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Fibrosis and defective muscle regeneration can hamper the functional recovery of the soft palate muscles after cleft palate repair. This causes persistent problems in speech, swallowing, and sucking. In vitro culture systems that allow the study of satellite cells (myogenic stem cells) from head muscles are crucial to develop new therapies based on tissue engineering to promote muscle regeneration after surgery. These systems will offer new perspectives for the treatment of cleft palate patients. A protocol for the isolation, culture and differentiation of satellite cells from head muscles is presented. The isolation is based on enzymatic digestion and trituration to release the satellite cells. In addition, this protocol comprises an innovative method using extracellular matrix gel coatings of millimeter size, which requires only low numbers of satellite cells for differentiation assays.
Collapse
Affiliation(s)
- Paola L Carvajal Monroy
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center
| | | | - Sander Grefte
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center
| | - Anne Marie Kuijpers-Jagtman
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center
| | - Frank A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center
| | - Johannes W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center;
| |
Collapse
|
58
|
Yablonka-Reuveni Z, Danoviz ME, Phelps M, Stuelsatz P. Myogenic-specific ablation of Fgfr1 impairs FGF2-mediated proliferation of satellite cells at the myofiber niche but does not abolish the capacity for muscle regeneration. Front Aging Neurosci 2015; 7:85. [PMID: 26074812 PMCID: PMC4446549 DOI: 10.3389/fnagi.2015.00085] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 04/30/2015] [Indexed: 11/13/2022] Open
Abstract
Skeletal muscle satellite cells (SCs) are Pax7+ myogenic stem cells that reside between the basal lamina and the plasmalemma of the myofiber. In mature muscles, SCs are typically quiescent, but can be activated in response to muscle injury. Depending on the magnitude of tissue trauma, SCs may divide minimally to repair subtle damage within individual myofibers or produce a larger progeny pool that forms new myofibers in cases of overt muscle injury. SC transition through proliferation, differentiation and renewal is governed by the molecular blueprint of the cells as well as by the extracellular milieu at the SC niche. In particular, the role of the fibroblast growth factor (FGF) family in regulating SCs during growth and aging is well recognized. Of the several FGFs shown to affect SCs, FGF1, FGF2, and FGF6 proteins have been documented in adult skeletal muscle. These prototypic paracrine FGFs transmit their mitogenic effect through the FGFRs, which are transmembrane tyrosine kinase receptors. Using the mouse model, we show here that of the four Fgfr genes, only Fgfr1 and Fgfr4 are expressed at relatively high levels in quiescent SCs and their proliferating progeny. To further investigate the role of FGFR1 in adult myogenesis, we have employed a genetic (Cre/loxP) approach for myogenic-specific (MyoDCre-driven) ablation of Fgfr1. Neither muscle histology nor muscle regeneration following cardiotoxin-induced injury were overtly affected in Fgfr1-ablated mice. This suggests that FGFR1 is not obligatory for SC performance in this acute muscle trauma model, where compensatory growth factor/cytokine regulatory cascades may exist. However, the SC mitogenic response to FGF2 is drastically repressed in isolated myofibers prepared from Fgfr1-ablated mice. Collectively, our study indicates that FGFR1 is important for FGF-mediated proliferation of SCs and its mitogenic role is not compensated by FGFR4 that is also highly expressed in SCs.
Collapse
Affiliation(s)
- Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle WA, USA
| | - Maria E Danoviz
- Department of Biological Structure, University of Washington School of Medicine, Seattle WA, USA
| | - Michael Phelps
- Department of Biological Structure, University of Washington School of Medicine, Seattle WA, USA
| | - Pascal Stuelsatz
- Department of Biological Structure, University of Washington School of Medicine, Seattle WA, USA
| |
Collapse
|
59
|
Dai JM, Yu MX, Shen ZY, Guo CY, Zhuang SQ, Qiu XS. Leucine Promotes Proliferation and Differentiation of Primary Preterm Rat Satellite Cells in Part through mTORC1 Signaling Pathway. Nutrients 2015; 7:3387-400. [PMID: 26007333 PMCID: PMC4446757 DOI: 10.3390/nu7053387] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 12/19/2022] Open
Abstract
Signaling through the mammalian target of rapamycin (mTOR) in response to leucine modulates many cellular and developmental processes. However, in the context of satellite cell proliferation and differentiation, the role of leucine and mTORC1 is less known. This study investigates the role of leucine in the process of proliferation and differentiation of primary preterm rat satellite cells, and the relationship with mammalian target of rapamycin complex 1 (mTORC1) activation. Dissociation of primary satellite cells occurred with type I collagenase and trypsin, and purification, via different speed adherence methods. Satellite cells with positive expression of Desmin were treated with leucine and rapamycin. We observed that leucine promoted proliferation and differentiation of primary satellite cells and increased the phosphorylation of mTOR. Rapamycin inhibited proliferation and differentiation, as well as decreased the phosphorylation level of mTOR. Furthermore, leucine increased the expression of MyoD and myogenin while the protein level of MyoD decreased due to rapamycin. However, myogenin expressed no affect by rapamycin. In conclusion, leucine may up-regulate the activation of mTORC1 to promote proliferation and differentiation of primary preterm rat satellite cells. We have shown that leucine promoted the differentiation of myotubes in part through the mTORC1-MyoD signal pathway.
Collapse
Affiliation(s)
- Jie-Min Dai
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China.
| | - Mu-Xue Yu
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China.
| | - Zhen-Yu Shen
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China.
| | - Chu-Yi Guo
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China.
| | - Si-Qi Zhuang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China.
| | - Xiao-Shan Qiu
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China.
| |
Collapse
|
60
|
Abstract
Skeletal muscles in vertebrates have a phenomenal regenerative capacity. A muscle that has been crushed can regenerate fully both structurally and functionally within a month. Remarkably, efficient regeneration continues to occur following repeated injuries. Thousands of muscle precursor cells are needed to accomplish regeneration following acute injury. The differentiated muscle cells, the multinucleated contractile myofibers, are terminally withdrawn from mitosis. The source of the regenerative precursors is the skeletal muscle stem cells-the mononucleated cells closely associated with myofibers, which are known as satellite cells. Satellite cells are mitotically quiescent or slow-cycling, committed to myogenesis, but undifferentiated. Disruption of the niche after muscle damage results in their exit from quiescence and progression towards commitment. They eventually arrest proliferation, differentiate, and fuse to damaged myofibers or make de novo myofibers. Satellite cells are one of the well-studied adult tissue-specific stem cells and have served as an excellent model for investigating adult stem cells. They have also emerged as an important standard in the field of ageing and stem cells. Several recent reviews have highlighted the importance of these cells as a model to understand stem cell biology. This chapter begins with the discovery of satellite cells as skeletal muscle stem cells and their developmental origin. We discuss transcription factors and signalling cues governing stem cell function of satellite cells and heterogeneity in the satellite cell pool. Apart from satellite cells, a number of other stem cells have been shown to make muscle and are being considered as candidate stem cells for amelioration of muscle degenerative diseases. We discuss these "offbeat" muscle stem cells and their status as adult skeletal muscle stem cells vis-a-vis satellite cells. The ageing context is highlighted in the concluding section.
Collapse
Affiliation(s)
- Ramkumar Sambasivan
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK, Bellary Road, Bangalore, 560065, India,
| | | |
Collapse
|
61
|
De Luca G, Ferretti R, Bruschi M, Mezzaroma E, Caruso M. Cyclin D3 critically regulates the balance between self-renewal and differentiation in skeletal muscle stem cells. Stem Cells 2014; 31:2478-91. [PMID: 23897741 PMCID: PMC3963451 DOI: 10.1002/stem.1487] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/26/2013] [Accepted: 06/21/2013] [Indexed: 12/28/2022]
Abstract
Satellite cells are mitotically quiescent myogenic stem cells resident beneath the basal lamina surrounding adult muscle myofibers. In response to injury, multiple extrinsic signals drive the entry of satellite cells into the cell cycle and then to proliferation, differentiation, and self-renewal of their downstream progeny. Because satellite cells must endure for a lifetime, their cell cycle activity must be carefully controlled to coordinate proliferative expansion and self-renewal with the onset of the differentiation program. In this study, we find that cyclin D3, a member of the family of mitogen-activated D-type cyclins, is critically required for proper developmental progression of myogenic progenitors. Using a cyclin D3-knockout mouse we determined that cyclin D3 deficiency leads to reduced myofiber size and impaired establishment of the satellite cell population within the adult muscle. Cyclin D3-null myogenic progenitors, studied ex vivo on isolated myofibers and in vitro, displayed impaired cell cycle progression, increased differentiation potential, and reduced self-renewal capability. Similarly, silencing of cyclin D3 in C2 myoblasts caused anticipated exit from the cell cycle and precocious onset of terminal differentiation. After induced muscle damage, cyclin D3-null myogenic progenitors exhibited proliferation deficits, a precocious ability to form newly generated myofibers and a reduced capability to repopulate the satellite cell niche at later stages of the regeneration process. These results indicate that cyclin D3 plays a cell-autonomous and nonredundant function in regulating the dynamic balance between proliferation, differentiation, and self-renewal that normally establishes an appropriate pool size of adult satellite cells.
Collapse
Affiliation(s)
- Giulia De Luca
- National Research Council, Institute of Cell Biology and Neurobiology, Fondazione Santa Lucia, Roma, Italy
| | | | | | | | | |
Collapse
|
62
|
Schöneich C, Dremina E, Galeva N, Sharov V. Apoptosis in differentiating C2C12 muscle cells selectively targets Bcl-2-deficient myotubes. Apoptosis 2014; 19:42-57. [PMID: 24129924 DOI: 10.1007/s10495-013-0922-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Muscle cell apoptosis accompanies normal muscle development and regeneration, as well as degenerative diseases and aging. C2C12 murine myoblast cells represent a common model to study muscle differentiation. Though it was already shown that myogenic differentiation of C2C12 cells is accompanied by enhanced apoptosis in a fraction of cells, either the cell population sensitive to apoptosis or regulatory mechanisms for the apoptotic response are unclear so far. In the current study we characterize apoptotic phenotypes of different types of C2C12 cells at all stages of differentiation, and report here that myotubes of differentiated C2C12 cells with low levels of anti-apoptotic Bcl-2 expression are particularly vulnerable to apoptosis even though they are displaying low levels of pro-apoptotic proteins Bax, Bak and Bad. In contrast, reserve cells exhibit higher levels of Bcl-2 and high resistance to apoptosis. The transfection of proliferating myoblasts with Bcl-2 prior to differentiation did not protect against spontaneous apoptosis accompanying differentiation of C2C12 cells but led to Bcl-2 overexpression in myotubes and to significant protection from apoptotic cell loss caused by exposure to hydrogen peroxide. Overall, our data advocate for a Bcl-2-dependent mechanism of apoptosis in differentiated muscle cells. However, downstream processes for spontaneous and hydrogen peroxide induced apoptosis are not completely similar. Apoptosis in differentiating myoblasts and myotubes is regulated not through interaction of Bcl-2 with pro-apoptotic Bcl-2 family proteins such as Bax, Bak, and Bad.
Collapse
|
63
|
Abstract
Muscle regeneration recapitulates many aspects of embryonic myogenesis and is an important homeostatic process of the adult skeletal muscle, which, after development, retains the capacity to regenerate in response to appropriate stimuli, activating the muscle compartment of stem cells, namely, satellite cells, as well as other precursor cells. Moreover, significant evidence suggests that while stem cells represent an important determinant for tissue regeneration, a “qualified” environment is necessary to guarantee and achieve functional results. It is therefore plausible that the loss of control over these cell fate decisions could lead to a pathological transdifferentiation, leading to pathologic defects in the regenerative process. This review provides an overview about the general aspects of muscle development and discusses the cellular and molecular aspects that characterize the five interrelated and time-dependent phases of muscle regeneration, namely, degeneration, inflammation, regeneration, remodeling, and maturation/functional repair.
Collapse
|
64
|
Sondag GR, Salihoglu S, Lababidi SL, Crowder DC, Moussa FM, Abdelmagid SM, Safadi FF. Osteoactivin induces transdifferentiation of C2C12 myoblasts into osteoblasts. J Cell Physiol 2014; 229:955-66. [PMID: 24265122 DOI: 10.1002/jcp.24512] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 11/18/2013] [Indexed: 12/19/2022]
Abstract
Osteoactivin (OA) is a novel osteogenic factor important for osteoblast differentiation and function. Previous studies showed that OA stimulates matrix mineralization and transcription of osteoblast specific genes required for differentiation. OA plays a role in wound healing and its expression was shown to increase in post fracture calluses. OA expression was reported in muscle as OA is upregulated in cases of denervation and unloading stress. The regulatory mechanisms of OA in muscle and bone have not yet been determined. In this study, we examined whether OA plays a role in transdifferentiation of C2C12 myoblast into osteoblasts. Infected C2C12 with a retroviral vector overexpressing OA under the CMV promoter were able to transdifferentiate from myoblasts into osteoblasts. Immunofluorescence analysis showed that skeletal muscle marker MF-20 was severely downregulated in cells overexpressing OA and contained significantly less myotubes compared to uninfected control. C2C12 myoblasts overexpressing OA showed an increase in expression of bone specific markers such as alkaline phosphatase and alizarin red staining, and also showed an increase in Runx2 protein expression. We also detected increased levels of phosphorylated focal adhesion kinase (FAK) in C2C12 myoblasts overexpressing OA compared to control. Taken together, our results suggest that OA is able to induce transdifferentiation of myoblasts into osteoblasts through increasing levels of phosphorylated FAK.
Collapse
Affiliation(s)
- Gregory R Sondag
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, Ohio; School of Biomedical Sciences, Kent State University, Kent, Ohio
| | | | | | | | | | | | | |
Collapse
|
65
|
Froehlich JM, Seiliez I, Gabillard JC, Biga PR. Preparation of primary myogenic precursor cell/myoblast cultures from basal vertebrate lineages. J Vis Exp 2014. [PMID: 24835774 DOI: 10.3791/51354] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Due to the inherent difficulty and time involved with studying the myogenic program in vivo, primary culture systems derived from the resident adult stem cells of skeletal muscle, the myogenic precursor cells (MPCs), have proven indispensible to our understanding of mammalian skeletal muscle development and growth. Particularly among the basal taxa of Vertebrata, however, data are limited describing the molecular mechanisms controlling the self-renewal, proliferation, and differentiation of MPCs. Of particular interest are potential mechanisms that underlie the ability of basal vertebrates to undergo considerable postlarval skeletal myofiber hyperplasia (i.e. teleost fish) and full regeneration following appendage loss (i.e. urodele amphibians). Additionally, the use of cultured myoblasts could aid in the understanding of regeneration and the recapitulation of the myogenic program and the differences between them. To this end, we describe in detail a robust and efficient protocol (and variations therein) for isolating and maintaining MPCs and their progeny, myoblasts and immature myotubes, in cell culture as a platform for understanding the evolution of the myogenic program, beginning with the more basal vertebrates. Capitalizing on the model organism status of the zebrafish (Danio rerio), we report on the application of this protocol to small fishes of the cyprinid clade Danioninae. In tandem, this protocol can be utilized to realize a broader comparative approach by isolating MPCs from the Mexican axolotl (Ambystoma mexicanum) and even laboratory rodents. This protocol is now widely used in studying myogenesis in several fish species, including rainbow trout, salmon, and sea bream(1-4).
Collapse
Affiliation(s)
| | | | | | - Peggy R Biga
- Department of Biology, University of Alabama at Birmingham;
| |
Collapse
|
66
|
Abstract
Since the seminal discovery of the cell-fate regulator Myod, studies in skeletal myogenesis have inspired the search for cell-fate regulators of similar potential in other tissues and organs. It was perplexing that a similar transcription factor for other tissues was not found; however, it was later discovered that combinations of molecular regulators can divert somatic cell fates to other cell types. With the new era of reprogramming to induce pluripotent cells, the myogenesis paradigm can now be viewed under a different light. Here, we provide a short historical perspective and focus on how the regulation of skeletal myogenesis occurs distinctly in different scenarios and anatomical locations. In addition, some interesting features of this tissue underscore the importance of reconsidering the simple-minded view that a single stem cell population emerges after gastrulation to assure tissuegenesis. Notably, a self-renewing long-term Pax7+ myogenic stem cell population emerges during development only after a first wave of terminal differentiation occurs to establish a tissue anlagen in the mouse. How the future stem cell population is selected in this unusual scenario will be discussed. Recently, a wealth of information has emerged from epigenetic and genome-wide studies in myogenic cells. Although key transcription factors such as Pax3, Pax7, and Myod regulate only a small subset of genes, in some cases their genomic distribution and binding are considerably more promiscuous. This apparent nonspecificity can be reconciled in part by the permissivity of the cell for myogenic commitment, and also by new roles for some of these regulators as pioneer transcription factors acting on chromatin state.
Collapse
Affiliation(s)
- Glenda Comai
- Stem Cells and Development, CNRS URA 2578, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, CNRS URA 2578, Department of Developmental & Stem Cell Biology, Institut Pasteur, Paris, France.
| |
Collapse
|
67
|
Wu MP, Doyle JR, Barry B, Beauvais A, Rozkalne A, Piao X, Lawlor MW, Kopin AS, Walsh CA, Gussoni E. G-protein coupled receptor 56 promotes myoblast fusion through serum response factor- and nuclear factor of activated T-cell-mediated signalling but is not essential for muscle development in vivo. FEBS J 2013; 280:6097-113. [PMID: 24102982 PMCID: PMC3877849 DOI: 10.1111/febs.12529] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/24/2013] [Accepted: 09/04/2013] [Indexed: 12/28/2022]
Abstract
Mammalian muscle cell differentiation is a complex process of multiple steps for which many of the factors involved have not yet been defined. In a screen to identify the regulators of myogenic cell fusion, we found that the gene for G-protein coupled receptor 56 (GPR56) was transiently up-regulated during the early fusion of human myoblasts. Human mutations in the gene for GPR56 cause the disease bilateral frontoparietal polymicrogyria; however, the consequences of receptor dysfunction on muscle development have not been explored. Using knockout mice, we defined the role of GPR56 in skeletal muscle. GPR56(-/-) myoblasts have decreased fusion and smaller myotube sizes in culture. In addition, a loss of GPR56 expression in muscle cells results in decreases or delays in the expression of myogenic differentiation 1, myogenin and nuclear factor of activated T-cell (NFAT)c2. Our data suggest that these abnormalities result from decreased GPR56-mediated serum response element and NFAT signalling. Despite these changes, no overt differences in phenotype were identified in the muscle of GPR56 knockout mice, which presented only a mild but statistically significant elevation of serum creatine kinase compared to wild-type. In agreement with these findings, clinical data from 13 bilateral frontoparietal polymicrogyria patients revealed mild serum creatine kinase increase in only two patients. In summary, targeted disruption of GPR56 in mice results in myoblast abnormalities. The absence of a severe muscle phenotype in GPR56 knockout mice and human patients suggests that other factors may compensate for the lack of this G-protein coupled receptor during muscle development and that the motor delay observed in these patients is likely not a result of primary muscle abnormalities.
Collapse
Affiliation(s)
- Melissa P. Wu
- Biological and Biomedical Sciences, Harvard Medical School, Boston MA 02115, USA
- Division of Genetics, Boston Children’s Hospital, Boston MA 02115, USA
| | - Jamie R. Doyle
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Brenda Barry
- Division of Genetics, Boston Children’s Hospital, Boston MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston MA 02115, USA
| | - Ariane Beauvais
- Division of Genetics, Boston Children’s Hospital, Boston MA 02115, USA
| | - Anete Rozkalne
- Division of Genetics, Boston Children’s Hospital, Boston MA 02115, USA
| | - Xianhua Piao
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA 02115, USA
| | - Michael W. Lawlor
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Wisconsin and Medical College of Wisconsin, Milwaukee WI 53226, USA
| | - Alan S. Kopin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Christopher A. Walsh
- Division of Genetics, Boston Children’s Hospital, Boston MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston MA 02115, USA
| | - Emanuela Gussoni
- Division of Genetics, Boston Children’s Hospital, Boston MA 02115, USA
| |
Collapse
|
68
|
Duchesne E, Bouchard P, Roussel MP, Côté CH. Mast cells can regulate skeletal muscle cell proliferation by multiple mechanisms. Muscle Nerve 2013; 48:403-14. [DOI: 10.1002/mus.23758] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2012] [Indexed: 12/16/2022]
Affiliation(s)
- Elise Duchesne
- CHUQ Research Center and Faculty of Medicine; Laval University; 2705 Boulevard Laurier, RC-9800 Québec G1V 4G2 Canada
| | - Patrice Bouchard
- CHUQ Research Center and Faculty of Medicine; Laval University; 2705 Boulevard Laurier, RC-9800 Québec G1V 4G2 Canada
| | - Marie-Pier Roussel
- CHUQ Research Center and Faculty of Medicine; Laval University; 2705 Boulevard Laurier, RC-9800 Québec G1V 4G2 Canada
| | - Claude H. Côté
- CHUQ Research Center and Faculty of Medicine; Laval University; 2705 Boulevard Laurier, RC-9800 Québec G1V 4G2 Canada
| |
Collapse
|
69
|
Carvajal Monroy PL, Grefte S, Kuijpers-Jagtman AM, Helmich MPAC, Ulrich DJO, Von den Hoff JW, Wagener FADTG. A rat model for muscle regeneration in the soft palate. PLoS One 2013; 8:e59193. [PMID: 23554995 PMCID: PMC3598650 DOI: 10.1371/journal.pone.0059193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/12/2013] [Indexed: 11/21/2022] Open
Abstract
Background Children with a cleft in the soft palate have difficulties with speech, swallowing, and sucking. Despite successful surgical repositioning of the muscles, optimal function is often not achieved. Scar formation and defective regeneration may hamper the functional recovery of the muscles after cleft palate repair. Therefore, the aim of this study is to investigate the anatomy and histology of the soft palate in rats, and to establish an in vivo model for muscle regeneration after surgical injury. Methods Fourteen adult male Sprague Dawley rats were divided into four groups. Groups 1 (n = 4) and 2 (n = 2) were used to investigate the anatomy and histology of the soft palate, respectively. Group 3 (n = 6) was used for surgical wounding of the soft palate, and group 4 (n = 2) was used as unwounded control group. The wounds (1 mm) were evaluated by (immuno)histochemistry (AZAN staining, Pax7, MyoD, MyoG, MyHC, and ASMA) after 7 days. Results The present study shows that the anatomy and histology of the soft palate muscles of the rat is largely comparable with that in humans. All wounds showed clinical evidence of healing after 7 days. AZAN staining demonstrated extensive collagen deposition in the wound area, and initial regeneration of muscle fibers and salivary glands. Proliferating and differentiating satellite cells were identified in the wound area by antibody staining. Conclusions This model is the first, suitable for studying muscle regeneration in the rat soft palate, and allows the development of novel adjuvant strategies to promote muscle regeneration after cleft palate surgery.
Collapse
Affiliation(s)
- Paola L Carvajal Monroy
- Department of Orthodontics and Craniofacial Biology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
70
|
Abstract
Adult skeletal muscle in mammals is a stable tissue under normal circumstances but has remarkable ability to repair after injury. Skeletal muscle regeneration is a highly orchestrated process involving the activation of various cellular and molecular responses. As skeletal muscle stem cells, satellite cells play an indispensible role in this process. The self-renewing proliferation of satellite cells not only maintains the stem cell population but also provides numerous myogenic cells, which proliferate, differentiate, fuse, and lead to new myofiber formation and reconstitution of a functional contractile apparatus. The complex behavior of satellite cells during skeletal muscle regeneration is tightly regulated through the dynamic interplay between intrinsic factors within satellite cells and extrinsic factors constituting the muscle stem cell niche/microenvironment. For the last half century, the advance of molecular biology, cell biology, and genetics has greatly improved our understanding of skeletal muscle biology. Here, we review some recent advances, with focuses on functions of satellite cells and their niche during the process of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Hang Yin
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
71
|
Yuan J, Tang Z, Yang S, Li K. CRABP2 promotes myoblast differentiation and is modulated by the transcription factors MyoD and Sp1 in C2C12 cells. PLoS One 2013; 8:e55479. [PMID: 23383201 PMCID: PMC3561243 DOI: 10.1371/journal.pone.0055479] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 12/23/2012] [Indexed: 11/19/2022] Open
Abstract
Cellular retinoic acid binding protein 2 (CRABP2), a member of a family of specific carrier proteins for Vitamin A, belongs to a family of small cytosolic lipid binding proteins. Our previous study suggested that CRABP2 was involved in skeletal muscle development; however, the molecular function and regulatory mechanism of CRABP2 in myogenesis remained unclear. In this study, we found that the expression of the CRABP2 gene was upregulated during C2C12 differentiation. An over-expression assay revealed that CRABP2 promotes myogenic transformation by regulating the cell cycle during C2C12 differentiation. The region from -459 to -4 bp was identified as the core promoter and contains a TATA box, a GC box and binding sites for the transcription factors MyoD and Sp1. Over-expression, site-directed mutagenesis and EMSA assays indicated that the transcription factors MyoD and Sp1 regulate CRABP2 expression and promote myoblast differentiation in C2C12 cells.
Collapse
Affiliation(s)
- Jing Yuan
- State Key Laboratory for Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
- College of Animal Science, Yangtze University, Jingzhou, People's Republic of China
| | - Zhonglin Tang
- State Key Laboratory for Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
- * E-mail: (ZT); (KL)
| | - Shulin Yang
- State Key Laboratory for Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Kui Li
- State Key Laboratory for Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
- * E-mail: (ZT); (KL)
| |
Collapse
|
72
|
Parker MH, Loretz C, Tyler AE, Duddy WJ, Hall JK, Olwin BB, Bernstein ID, Storb R, Tapscott SJ. Activation of Notch signaling during ex vivo expansion maintains donor muscle cell engraftment. Stem Cells 2013; 30:2212-20. [PMID: 22865615 PMCID: PMC3448880 DOI: 10.1002/stem.1181] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Transplantation of myogenic stem cells possesses great potential for long‐term repair of dystrophic muscle. However, a single donor muscle biopsy is unlikely to provide enough cells to effectively transplant the muscle mass of a patient affected by muscular dystrophy. Expansion of cells ex vivo using traditional culture techniques significantly reduces engraftment potential. We hypothesized that activation of Notch signaling during ex vivo expansion would maintain donor cell engraftment potential. In this study, we expanded freshly isolated canine muscle‐derived cells on tissue culture plates coated with Delta‐1ext‐IgG to activate Notch signaling or with human IgG as a control. A model of canine‐to‐murine xenotransplantation was used to quantitatively compare canine muscle cell engraftment and determine whether engrafted donor cells could function as satellite cells in vivo. We show that Delta‐1ext‐IgG inhibited differentiation of canine muscle‐derived cells and increased the level of genes normally expressed in myogenic precursors. Moreover, cells expanded on Delta‐1ext‐IgG resulted in a significant increase in the number of donor‐derived fibers, as compared to cells expanded on human IgG, reaching engraftment levels similar to freshly isolated cells. Importantly, cells expanded on Delta‐1ext‐IgG engrafted to the recipient satellite cell niche and contributed to further regeneration. A similar strategy of expanding human muscle‐derived cells on Notch ligand might facilitate engraftment and muscle regeneration for patients affected with muscular dystrophy. Stem Cells2012;30:2212–2220
Collapse
Affiliation(s)
- Maura H Parker
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109-1024, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Abstract
Multinucleated myofibers are the functional contractile units of skeletal muscle. In adult muscle, mononuclear satellite cells, located between the basal lamina and the plasmalemma of the myofiber, are the primary myogenic stem cells. This chapter describes protocols for isolation, culturing, and immunostaining of myofibers from mouse skeletal muscle. Myofibers are isolated intact and retain their associated satellite cells. The first protocol discusses myofiber isolation from the flexor digitorum brevis (FDB) muscle. These short myofibers are cultured in dishes coated with PureCol collagen (formerly known as Vitrogen) using a serum replacement medium. Employing such culture conditions, satellite cells remain associated with the myofibers, undergoing proliferation and differentiation on the myofiber surface. The second protocol discusses the isolation of longer myofibers from the extensor digitorum longus (EDL) muscle. Different from the FDB preparation, where multiple myofibers are processed together, the longer EDL myofibers are typically processed and cultured individually in dishes coated with Matrigel using a growth factor rich medium. Under these conditions, satellite cells initially remain associated with the parent myofiber and later migrate away, giving rise to proliferating and differentiating progeny. Myofibers from other types of muscles, such as diaphragm, masseter, and extraocular muscles can also be isolated and analyzed using protocols described herein. Overall, cultures of isolated myofibers provide essential tools for studying the interplay between the parent myofiber and its associated satellite cells. The current chapter provides background, procedural, and reagent updates, and step-by-step images of FDB and EDL muscle isolations, not included in our 2005 publication in this series.
Collapse
|
74
|
Andres-Mateos E, Mejias R, Soleimani A, Lin BM, Burks TN, Marx R, Lin B, Zellars RC, Zhang Y, Huso DL, Marr TG, Leinwand LA, Merriman DK, Cohn RD. Impaired skeletal muscle regeneration in the absence of fibrosis during hibernation in 13-lined ground squirrels. PLoS One 2012; 7:e48884. [PMID: 23155423 PMCID: PMC3498346 DOI: 10.1371/journal.pone.0048884] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Accepted: 10/02/2012] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle atrophy can occur as a consequence of immobilization and/or starvation in the majority of vertebrates studied. In contrast, hibernating mammals are protected against the loss of muscle mass despite long periods of inactivity and lack of food intake. Resident muscle-specific stem cells (satellite cells) are known to be activated by muscle injury and their activation contributes to the regeneration of muscle, but whether satellite cells play a role in hibernation is unknown. In the hibernating 13-lined ground squirrel we show that muscles ablated of satellite cells were still protected against atrophy, demonstrating that satellite cells are not involved in the maintenance of skeletal muscle during hibernation. Additionally, hibernating skeletal muscle showed extremely slow regeneration in response to injury, due to repression of satellite cell activation and myoblast differentiation caused by a fine-tuned interplay of p21, myostatin, MAPK, and Wnt signaling pathways. Interestingly, despite long periods of inflammation and lack of efficient regeneration, injured skeletal muscle from hibernating animals did not develop fibrosis and was capable of complete recovery when animals emerged naturally from hibernation. We propose that hibernating squirrels represent a new model system that permits evaluation of impaired skeletal muscle remodeling in the absence of formation of tissue fibrosis.
Collapse
Affiliation(s)
- Eva Andres-Mateos
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Rebeca Mejias
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Arshia Soleimani
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Brian M. Lin
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Tyesha N. Burks
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ruth Marx
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Benjamin Lin
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Richard C. Zellars
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Yonggang Zhang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - David L. Huso
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Tom G. Marr
- Hiberna Corporation, Boulder, Colorado, United States of America
| | - Leslie A. Leinwand
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Dana K. Merriman
- Department of Biology and Microbiology, University of Wisconsin, Oshkosh, Wisconsin, United States of America
| | - Ronald D. Cohn
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
75
|
Qin LL, Li XK, Xu J, Mo DL, Tong X, Pan ZC, Li JQ, Chen YS, Zhang Z, Wang C, Long QM. Mechano growth factor (MGF) promotes proliferation and inhibits differentiation of porcine satellite cells (PSCs) by down-regulation of key myogenic transcriptional factors. Mol Cell Biochem 2012; 370:221-30. [PMID: 22875667 DOI: 10.1007/s11010-012-1413-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 07/25/2012] [Indexed: 11/27/2022]
Abstract
Porcine satellite cells represent an ideal model system for studying the cellular and molecular basis regulating myogenic stem cell proliferation and differentiation and for exploring the experimental conditions for myoblast transplantation. Here, we investigated the effects of mechano growth factor (MGF), a spliced variant of the IGF-1 gene, on porcine satellite cells. We show that MGF potently stimulated proliferation while inhibited differentiation of porcine satellite cells. MGF-treatment acutely down-regulates the expression of myogenic determination factor (MyoD) and the cyclin-dependent kinase inhibitor p21. MGF-treatment also markedly reduced the overall expression of cyclin B1 and key factors of the myogenic regulatory and myocyte enhancer families, including Myogenein and MEF2A. Taken together, the gene expression data from MGF-treated porcine satellite cells are in favor of a molecular model in which MGF inhibits porcine satellite cell differentiation by down-regulating either the activity or expression of MyoD, which, in turn, suppresses the expression of key genes required for cell cycle progression and differentiation, such as p21, Myogenin, and MEF2. Overall, our findings are in support of the previous suggestion that MGF may be used in vivo and in vitro to promote proliferation of myogenic stem cells to prevent and treat age-related muscle degenerative diseases.
Collapse
Affiliation(s)
- Li-Li Qin
- College of Animal Science/Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Fatima S, Zhou S, Sorrentino BP. Abcg2 expression marks tissue-specific stem cells in multiple organs in a mouse progeny tracking model. Stem Cells 2012; 30:210-21. [PMID: 22134889 DOI: 10.1002/stem.1002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The side population phenotype is associated with the Hoechst dye efflux activity of the Abcg2 transporter and identifies hematopoietic stem cells (HSCs) in the bone marrow. This association suggests the direct use of Abcg2 expression to identify adult stem cells in various other organs. We have generated a lineage tracing mouse model based on an allele that coexpresses both Abcg2 and a CreERT2 expression cassette. By crossing these mice with lox-STOP-lox reporter lines (LacZ or YFP), cells that express Abcg2 and their progeny were identified following treatment with tamoxifen (Tam). In the liver and kidney, in which mature cells express Abcg2, reporter gene expression verified the expected physiologic expression pattern of the recombinant allele. Long-term marking of HSCs was seen in multiple peripheral blood lineages from adult mice, demonstrating that Abcg2(+) bone marrow HSCs contribute to steady-state hematopoiesis. Stem cell tracing patterns were seen in the small intestine and in seminiferous tubules in the testis 20 months after Tam treatment, proving that stem cells from these organs express Abcg2. Interstitial cells from skeletal and cardiac muscle were labeled, and some cells were costained with endothelial markers, raising the possibility that these cells may function in the repair response to muscle injury. Altogether, these studies prove that Abcg2 is a stem cell marker for blood, small intestine, testicular germ cells, and possibly for injured skeletal and/or cardiac muscle and provide a new model for studying stem cell activity that does not require transplant-based assays.
Collapse
Affiliation(s)
- Soghra Fatima
- Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | |
Collapse
|
77
|
Parker MH, von Maltzahn J, Bakkar N, Al-Joubori B, Ishibashi J, Guttridge D, Rudnicki MA. MyoD-dependent regulation of NF-κB activity couples cell-cycle withdrawal to myogenic differentiation. Skelet Muscle 2012; 2:6. [PMID: 22541644 PMCID: PMC3356597 DOI: 10.1186/2044-5040-2-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 04/27/2012] [Indexed: 01/19/2023] Open
Abstract
Background Mice lacking MyoD exhibit delayed skeletal muscle regeneration and markedly enhanced numbers of satellite cells. Myoblasts isolated from MyoD-/- myoblasts proliferate more rapidly than wild type myoblasts, display a dramatic delay in differentiation, and continue to incorporate BrdU after serum withdrawal. Methods Primary myoblasts isolated from wild type and MyoD-/- mutant mice were examined by microarray analysis and further characterized by cell and molecular experiments in cell culture. Results We found that NF-κB, a key regulator of cell-cycle withdrawal and differentiation, aberrantly maintains nuclear localization and transcriptional activity in MyoD-/- myoblasts. As a result, expression of cyclin D is maintained during serum withdrawal, inhibiting expression of muscle-specific genes and progression through the differentiation program. Sustained nuclear localization of cyclin E, and a concomitant increase in cdk2 activity maintains S-phase entry in MyoD-/- myoblasts even in the absence of mitogens. Importantly, this deficit was rescued by forced expression of IκBαSR, a non-degradable mutant of IκBα, indicating that inhibition of NF-κB is sufficient to induce terminal myogenic differentiation in the absence of MyoD. Conclusion MyoD-induced cytoplasmic relocalization of NF-κB is an essential step in linking cell-cycle withdrawal to the terminal differentiation of skeletal myoblasts. These results provide important insight into the unique functions of MyoD in regulating the switch from progenitor proliferation to terminal differentiation.
Collapse
Affiliation(s)
- Maura H Parker
- Ottawa Hospital Research Institute, 501 Smyth Rd, Ottawa, ON, K1H 8L6, Canada.
| | | | | | | | | | | | | |
Collapse
|
78
|
Anderson JE, Wozniak AC, Mizunoya W. Single muscle-fiber isolation and culture for cellular, molecular, pharmacological, and evolutionary studies. Methods Mol Biol 2012; 798:85-102. [PMID: 22130833 DOI: 10.1007/978-1-61779-343-1_6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The technique of single muscle-fiber cultures has already proven valuable in extending knowledge of myogenesis, stem cell heterogeneity, the stem cell niche in skeletal muscle, and satellite cell activation. This report reviews the background of the model and applications, and details the procedures of muscle dissection, fiber digestion and isolation, cleaning the fiber preparation, plating fibers, and extensions of the technique for studying activation from stable quiescence of satellite cells, mRNA expression by in situ hybridization and regulation of satellite cell activation in zebrafish muscle by nitric oxide, hepatocyte growth factor.
Collapse
Affiliation(s)
- Judy E Anderson
- Department of Biological Sciences, Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada.
| | | | | |
Collapse
|
79
|
Mokalled MH, Johnson AN, Creemers EE, Olson EN. MASTR directs MyoD-dependent satellite cell differentiation during skeletal muscle regeneration. Genes Dev 2012; 26:190-202. [PMID: 22279050 DOI: 10.1101/gad.179663.111] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In response to skeletal muscle injury, satellite cells, which function as a myogenic stem cell population, become activated, expand through proliferation, and ultimately fuse with each other and with damaged myofibers to promote muscle regeneration. Here, we show that members of the Myocardin family of transcriptional coactivators, MASTR and MRTF-A, are up-regulated in satellite cells in response to skeletal muscle injury and muscular dystrophy. Global and satellite cell-specific deletion of MASTR in mice impairs skeletal muscle regeneration. This impairment is substantially greater when MRTF-A is also deleted and is due to aberrant differentiation and excessive proliferation of satellite cells. These abnormalities mimic those associated with genetic deletion of MyoD, a master regulator of myogenesis, which is down-regulated in the absence of MASTR and MRTF-A. Consistent with an essential role of MASTR in transcriptional regulation of MyoD expression, MASTR activates a muscle-specific postnatal MyoD enhancer through associations with MEF2 and members of the Myocardin family. Our results provide new insights into the genetic circuitry of muscle regeneration and identify MASTR as a central regulator of this process.
Collapse
Affiliation(s)
- Mayssa H Mokalled
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, TX 75390, USA
| | | | | | | |
Collapse
|
80
|
Ma X, Zhang S, Zhou J, Chen B, Shang Y, Gao T, Wang X, Xie H, Chen F. Clone-derived human AF-amniotic fluid stem cells are capable of skeletal myogenic differentiation in vitro and in vivo. J Tissue Eng Regen Med 2012; 6:598-613. [PMID: 22396316 DOI: 10.1002/term.462] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 05/02/2011] [Accepted: 07/01/2011] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | | | | | - Xue Wang
- Department of Urology; Shanghai Children's Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai; People's Republic of China
| | - Hua Xie
- Department of Urology; Shanghai Children's Hospital; Shanghai Jiao Tong University School of Medicine; Shanghai; People's Republic of China
| | | |
Collapse
|
81
|
Yablonka-Reuveni Z. The skeletal muscle satellite cell: still young and fascinating at 50. J Histochem Cytochem 2012; 59:1041-59. [PMID: 22147605 DOI: 10.1369/0022155411426780] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The skeletal muscle satellite cell was first described and named based on its anatomic location between the myofiber plasma and basement membranes. In 1961, two independent studies by Alexander Mauro and Bernard Katz provided the first electron microscopic descriptions of satellite cells in frog and rat muscles. These cells were soon detected in other vertebrates and acquired candidacy as the source of myogenic cells needed for myofiber growth and repair throughout life. Cultures of isolated myofibers and, subsequently, transplantation of single myofibers demonstrated that satellite cells were myogenic progenitors. More recently, satellite cells were redefined as myogenic stem cells given their ability to self-renew in addition to producing differentiated progeny. Identification of distinctively expressed molecular markers, in particular Pax7, has facilitated detection of satellite cells using light microscopy. Notwithstanding the remarkable progress made since the discovery of satellite cells, researchers have looked for alternative cells with myogenic capacity that can potentially be used for whole body cell-based therapy of skeletal muscle. Yet, new studies show that inducible ablation of satellite cells in adult muscle impairs myofiber regeneration. Thus, on the 50th anniversary since its discovery, the satellite cell's indispensable role in muscle repair has been reaffirmed.
Collapse
Affiliation(s)
- Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle, Washington 98195, USA.
| |
Collapse
|
82
|
Fan GC. Role of heat shock proteins in stem cell behavior. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:305-22. [PMID: 22917237 DOI: 10.1016/b978-0-12-398459-3.00014-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stress response is well appreciated to induce the expression of heat shock proteins (Hsps) in the cell. Numerous studies have demonstrated that Hsps function as molecular chaperones in the stabilization of intracellular proteins, repairing damaged proteins, and assisting in protein translocation. Various kinds of stem cells (embryonic stem cells, adult stem cells, or induced pluripotent stem cells) have to maintain their stemness and, under certain circumstances, undergo stress. Therefore, Hsps should have an important influence on stem cells. Actually, numerous studies have indicated that some Hsps physically interact with a number of transcription factors as well as intrinsic and extrinsic signaling pathways. Importantly, alterations in Hsp expression have been demonstrated to affect stem cell behavior including self-renewal, differentiation, sensitivity to environmental stress, and aging. This chapter summarizes recent findings related to (1) the roles of Hsps in maintenance of stem cell dormancy, proliferation, and differentiation; (2) the expression signature of Hsps in embryonic/adult stem cells and differentiated stem cells; (3) the protective roles of Hsps in transplanted stem cells; and (4) the possible roles of Hsps in stem cell aging.
Collapse
Affiliation(s)
- Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
83
|
Danoviz ME, Yablonka-Reuveni Z. Skeletal muscle satellite cells: background and methods for isolation and analysis in a primary culture system. Methods Mol Biol 2012; 798:21-52. [PMID: 22130829 PMCID: PMC3325159 DOI: 10.1007/978-1-61779-343-1_2] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Repair of adult skeletal muscle depends on satellite cells, myogenic stem cells located between the basal lamina and the plasmalemma of the myofiber. Standardized protocols for the isolation and culture of satellite cells are key tools for understanding cell autonomous and extrinsic factors that regulate their performance. Knowledge gained from such studies can contribute important insights to developing strategies for the improvement of muscle repair following trauma and in muscle wasting disorders. This chapter provides an introduction to satellite cell biology and further describes the basic protocol used in our laboratory to isolate and culture satellite cells from adult skeletal muscle. The cell culture conditions detailed herein support proliferation and differentiation of satellite cell progeny and the development of reserve cells, which are thought to reflect the in vivo self-renewal ability of satellite cells. Additionally, this chapter describes our standard immunostaining protocol that allows the characterization of satellite cell progeny by the temporal expression of characteristic transcription factors and structural proteins associated with different stages of myogenic progression. Although emphasis is given here to the isolation and characterization of satellite cells from mouse hindlimb muscles, the protocols are suitable for other muscle types (such as diaphragm and extraocular muscles) and for muscles from other species, including chicken and rat. Altogether, the basic protocols described are straightforward and facilitate the study of diverse aspects of skeletal muscle stem cells.
Collapse
Affiliation(s)
- Maria Elena Danoviz
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Zipora Yablonka-Reuveni
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
84
|
Alter J, Bengal E. Stress-induced C/EBP homology protein (CHOP) represses MyoD transcription to delay myoblast differentiation. PLoS One 2011; 6:e29498. [PMID: 22242125 PMCID: PMC3248460 DOI: 10.1371/journal.pone.0029498] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Accepted: 11/29/2011] [Indexed: 11/23/2022] Open
Abstract
When mouse myoblasts or satellite cells differentiate in culture, the expression of myogenic regulatory factor, MyoD, is downregulated in a subset of cells that do not differentiate. The mechanism involved in the repression of MyoD expression remains largely unknown. Here we report that a stress-response pathway repressing MyoD transcription is transiently activated in mouse-derived C2C12 myoblasts growing under differentiation-promoting conditions. We show that phosphorylation of the α subunit of the translation initiation factor 2 (eIF2α) is followed by expression of C/EBP homology protein (CHOP) in some myoblasts. ShRNA-driven knockdown of CHOP expression caused earlier and more robust differentiation, whereas its constitutive expression delayed differentiation relative to wild type myoblasts. Cells expressing CHOP did not express the myogenic regulatory factors MyoD and myogenin. These results indicated that CHOP directly repressed the transcription of the MyoD gene. In support of this view, CHOP associated with upstream regulatory region of the MyoD gene and its activity reduced histone acetylation at the enhancer region of MyoD. CHOP interacted with histone deacetylase 1 (HDAC1) in cells. This protein complex may reduce histone acetylation when bound to MyoD regulatory regions. Overall, our results suggest that the activation of a stress pathway in myoblasts transiently downregulate the myogenic program.
Collapse
Affiliation(s)
- Joel Alter
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eyal Bengal
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- * E-mail:
| |
Collapse
|
85
|
Averous J, Gabillard JC, Seiliez I, Dardevet D. Leucine limitation regulates myf5 and myoD expression and inhibits myoblast differentiation. Exp Cell Res 2011; 318:217-27. [PMID: 22079119 DOI: 10.1016/j.yexcr.2011.10.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 10/25/2011] [Accepted: 10/27/2011] [Indexed: 01/27/2023]
Abstract
Satellite cells are the major pool of muscle stem cells after birth; they represent an important component required to maintain muscle mass and functionality during life. The molecular mechanisms involved in myogenic differentiation are relatively well-known. However, the role of extracellular stimulus in the control of differentiation remains largely unresolved. Notably little is known about the impact of nutrients on this process. Here we have studied the role of leucine, an essential amino acid, in the control of myogenic differentiation. Leucine is a well-known regulator of muscle protein synthesis. It acts not only as a substrate for translation but also as a regulator of gene expression and signaling pathways such as those involving mTOR and GCN2. In this study we demonstrated that the lack of leucine abolishes the differentiation of both C2C12 myoblasts and primary satellite cells. This effect is associated with a modification of the pattern of expression of the myogenic regulatory factors (MRF) myf5 and myoD. We report an up-regulation of myf5 mRNA and a decrease of myoD protein level during leucine starvation. This study demonstrates the importance of a nutrient, leucine, in the control of the myogenic differentiation program.
Collapse
Affiliation(s)
- J Averous
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 CLERMONT-FERRAND, France.
| | | | | | | |
Collapse
|
86
|
Johnston APW, Bellamy LM, Lisio MD, Parise G. Captopril treatment induces hyperplasia but inhibits myonuclear accretion following severe myotrauma in murine skeletal muscle. Am J Physiol Regul Integr Comp Physiol 2011; 301:R363-9. [DOI: 10.1152/ajpregu.00766.2010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of ANG II in skeletal muscle and satellite cell regulation is largely unknown. Cardiotoxin (CTX) was used to investigate whether muscle injury activates a local ANG II signaling system. Following injury, immunohistochelmistry (IHC) analysis revealed a robust increase in the intensity of angiotensinogen and angiotensin type 1 (AT1) receptor expression. As regeneration proceeded, however, AT1 and angiotensinogen were downregulated. Nuclear accretion and fiber formation were also assessed during muscle regeneration in mice treated with captopril (an angiotensin-converting enzyme inhibitor). When ANG II formation was blocked through the use of captopril, we observed a significantly reduced accretion of nuclei into myofibers (−25%), while tibialis anterior total fiber number was significantly increased +37%. This phenotype appeared to be due to alterations in satellite cell differentiation kinetics; captopril treatment led to sustained mRNA expression of markers associated with quiescence and proliferation (Myf5, Pax7) and simultaneously delayed or inhibited the expression of myogenin. IHC staining supported these findings, revealing that captopril treatment resulted in a strong trend ( P = 0.06) for a decrease in the proportion of myogenin-positive myoblasts. Furthermore, these observations were associated with a delay in muscle fiber maturation; captopril treatment resulted in sustained expression of embryonic myosin heavy chain. Collectively, these findings demonstrate that localized skeletal muscle angiotensin signaling is important to muscle fiber formation, myonuclear accretion, and satellite cell function.
Collapse
Affiliation(s)
| | | | | | - Gianni Parise
- Departments of 1Kinesiology and Medical Physics and
- Applied Radiation Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
87
|
Leiter JRS, Peeler J, Anderson JE. Exercise-induced muscle growth is muscle-specific and age-dependent. Muscle Nerve 2011; 43:828-38. [PMID: 21607967 DOI: 10.1002/mus.21965] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Sarcopenia, and the importance of satellite cells (SCs) in muscle growth led us to examine the effects of exercise and age on SC activation and gene expression. METHODS Eight- and 18-month-old mice were either sedentary or underwent 3 weeks of exercise (N = 24). Body mass, distance traveled, and grip strength were recorded at weekly intervals. The extensor digitorum longus (EDL), tibialis anterior (TA), gastrocnemius (GAST), and quadriceps (QUAD) muscles were analyzed along with muscle fiber area, SC activation, neuronal nitric oxide synthase (NOS-I), MyoD, and myostatin protein content. RESULTS Older mice demonstrated decreased body mass, grip strength, and fiber area, but these changes were not affected by exercise. The QUAD muscle from young mice demonstrated an exercise-induced increase in SC activation and NOS-I and downregulation of myostatin. CONCLUSIONS Exercise-induced activation of SCs and regulation of gene expression are muscle-specific and age-dependent. Perturbed sensitivity to exercise in older mice provides insight into sarcopenia and potential treatments.
Collapse
Affiliation(s)
- Jeff R S Leiter
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
88
|
Hinits Y, Williams VC, Sweetman D, Donn TM, Ma TP, Moens CB, Hughes SM. Defective cranial skeletal development, larval lethality and haploinsufficiency in Myod mutant zebrafish. Dev Biol 2011; 358:102-12. [PMID: 21798255 DOI: 10.1016/j.ydbio.2011.07.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 07/12/2011] [Indexed: 11/18/2022]
Abstract
Myogenic regulatory factors of the myod family (MRFs) are transcription factors essential for mammalian skeletal myogenesis. Here we show that a mutation in the zebrafish myod gene delays and reduces early somitic and pectoral fin myogenesis, reduces miR-206 expression, and leads to a persistent reduction in somite size until at least the independent feeding stage. A mutation in myog, encoding a second MRF, has little obvious phenotype at early stages, but exacerbates the loss of somitic muscle caused by lack of Myod. Mutation of both myod and myf5 ablates all skeletal muscle. Haploinsufficiency of myod leads to reduced embryonic somite muscle bulk. Lack of Myod causes a severe reduction in cranial musculature, ablating most muscles including the protractor pectoralis, a putative cucullaris homologue. This phenotype is accompanied by a severe dysmorphology of the cartilaginous skeleton and failure of maturation of several cranial bones, including the opercle. As myod expression is restricted to myogenic cells, the data show that myogenesis is essential for proper skeletogenesis in the head.
Collapse
Affiliation(s)
- Yaniv Hinits
- Randall Division for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, SE1 1UL, UK
| | | | | | | | | | | | | |
Collapse
|
89
|
Starkey JD, Yamamoto M, Yamamoto S, Goldhamer DJ. Skeletal muscle satellite cells are committed to myogenesis and do not spontaneously adopt nonmyogenic fates. J Histochem Cytochem 2011; 59:33-46. [PMID: 21339173 DOI: 10.1369/jhc.2010.956995] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The developmental potential of skeletal muscle stem cells (satellite cells) remains controversial. The authors investigated satellite cell developmental potential in single fiber and clonal cultures derived from MyoD(iCre/+);R26R(EYFP/+) muscle, in which essentially all satellite cells are permanently labeled. Approximately 60% of the clones derived from cells that co-purified with muscle fibers spontaneously underwent adipogenic differentiation. These adipocytes stained with Oil-Red-O and expressed the terminal differentiation markers, adipsin and fatty acid binding protein 4, but did not express EYFP and were therefore not of satellite cell origin. Satellite cells mutant for either MyoD or Myf-5 also maintained myogenic programming in culture and did not adopt an adipogenic fate. Incorporation of additional wash steps prior to muscle fiber plating virtually eliminated the non-myogenic cells but did not reduce the number of adherent Pax7+ satellite cells. More than half of the adipocytes observed in cultures from Tie2-Cre mice were recombined, further demonstrating a non-satellite cell origin. Under adipogenesis-inducing conditions, satellite cells accumulated cytoplasmic lipid but maintained myogenic protein expression and did not fully execute the adipogenic differentiation program, distinguishing them from adipocytes observed in muscle fiber cultures. The authors conclude that skeletal muscle satellite cells are committed to myogenesis and do not spontaneously adopt an adipogenic fate.
Collapse
Affiliation(s)
- Jessica D Starkey
- Department of Molecular and Cell Biology, Center for Regenerative Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, Connecticut, USA
| | | | | | | |
Collapse
|
90
|
Stoppani E, Rossi S, Meacci E, Penna F, Costelli P, Bellucci A, Faggi F, Maiolo D, Monti E, Fanzani A. Point mutated caveolin-3 form (P104L) impairs myoblast differentiation via Akt and p38 signalling reduction, leading to an immature cell signature. Biochim Biophys Acta Mol Basis Dis 2011; 1812:468-79. [DOI: 10.1016/j.bbadis.2010.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 11/30/2010] [Accepted: 12/08/2010] [Indexed: 11/24/2022]
|
91
|
Ciemerych MA, Archacka K, Grabowska I, Przewoźniak M. Cell cycle regulation during proliferation and differentiation of mammalian muscle precursor cells. Results Probl Cell Differ 2011; 53:473-527. [PMID: 21630157 DOI: 10.1007/978-3-642-19065-0_20] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Proliferation and differentiation of muscle precursor cells are intensively studied not only in the developing mouse embryo but also using models of skeletal muscle regeneration or analyzing in vitro cultured cells. These analyses allowed to show the universality of the cell cycle regulation and also uncovered tissue-specific interplay between major cell cycle regulators and factors crucial for the myogenic differentiation. Examination of the events accompanying proliferation and differentiation leading to the formation of functional skeletal muscle fibers allows understanding the molecular basis not only of myogenesis but also of skeletal muscle regeneration. This chapter presents the basis of the cell cycle regulation in proliferating and differentiating muscle precursor cells during development and after muscle injury. It focuses at major cell cycle regulators, myogenic factors, and extracellular environment impacting on the skeletal muscle.
Collapse
Affiliation(s)
- Maria A Ciemerych
- Department of Cytology, Institute of Zoology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland.
| | | | | | | |
Collapse
|
92
|
Abstract
Muscle development, growth, and regeneration take place throughout vertebrate life. In amniotes, myogenesis takes place in four successive, temporally distinct, although overlapping phases. Understanding how embryonic, fetal, neonatal, and adult muscle are formed from muscle progenitors and committed myoblasts is an area of active research. In this review we examine recent expression, genetic loss-of-function, and genetic lineage studies that have been conducted in the mouse, with a particular focus on limb myogenesis. We synthesize these studies to present a current model of how embryonic, fetal, neonatal, and adult muscle are formed in the limb.
Collapse
Affiliation(s)
- Malea Murphy
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | | |
Collapse
|
93
|
Osborn DPS, Li K, Hinits Y, Hughes SM. Cdkn1c drives muscle differentiation through a positive feedback loop with Myod. Dev Biol 2010; 350:464-75. [PMID: 21147088 DOI: 10.1016/j.ydbio.2010.12.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 12/01/2010] [Accepted: 12/03/2010] [Indexed: 01/15/2023]
Abstract
Differentiation often requires conversion of analogue signals to a stable binary output through positive feedback. Hedgehog (Hh) signalling promotes myogenesis in the vertebrate somite, in part by raising the activity of muscle regulatory factors (MRFs) of the Myod family above a threshold. Hh is known to enhance MRF expression. Here we show that Hh is also essential at a second step that increases Myod protein activity, permitting it to promote Myogenin expression. Hh acts by inducing expression of cdkn1c (p57(Kip2)) in slow muscle precursor cells, but neither Hh nor Cdkn1c is required for their cell cycle exit. Cdkn1c co-operates with Myod to drive differentiation of several early zebrafish muscle fibre types. Myod in turn up-regulates cdkn1c, thereby providing a positive feedback loop that switches myogenic cells to terminal differentiation.
Collapse
Affiliation(s)
- Daniel P S Osborn
- King's College London, Randall Division for Cell and Molecular Biophysics, London, UK
| | | | | | | |
Collapse
|
94
|
Xynos A, Corbella P, Belmonte N, Zini R, Manfredini R, Ferrari G. Bone marrow-derived hematopoietic cells undergo myogenic differentiation following a Pax-7 independent pathway. Stem Cells 2010; 28:965-73. [PMID: 20333749 DOI: 10.1002/stem.418] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Several reports showed that hematopoietic stem cells (HSCs) participate in muscle regeneration, raising hope for their therapeutic potential for degenerative muscle diseases. However, proof that HSCs are able to reprogram their fate and enter a myogenic pathway, remains elusive. We demonstrate that murine bone marrow (BM)-derived hematopoietic cells, carrying reporter genes controlled by muscle-specific regulatory elements from the Myf5, myosin light chain (MLC3F), or MCK genes, are induced by myoblasts to activate muscle-specific genes. This potential resides in the more undifferentiated progenitors, expressing surface markers typical of HSCs. Comparative gene expression profiling of CD45(+)/Sca1(+) cells isolated from muscle or BM shows that hematopoietic cells participate to muscle regeneration, by undergoing a profound although incomplete myogenic reprogramming on interaction with the muscle microenviroment. These cells undergo specification and differentiation independently from Pax7 and MyoD, and lack Pax7-associated properties, such as self-renewal and proliferation, distinguishing from satellite cells. Our findings indicate that hematopoietic cells, on seeding in the muscle, become a distinct cell population endowed with myogenic potential.
Collapse
Affiliation(s)
- Alexandros Xynos
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | |
Collapse
|
95
|
Pöllänen E, Fey V, Törmäkangas T, Ronkainen PHA, Taaffe DR, Takala T, Koskinen S, Cheng S, Puolakka J, Kujala UM, Suominen H, Sipilä S, Kovanen V. Power training and postmenopausal hormone therapy affect transcriptional control of specific co-regulated gene clusters in skeletal muscle. AGE (DORDRECHT, NETHERLANDS) 2010; 32:347-363. [PMID: 20640546 PMCID: PMC2926854 DOI: 10.1007/s11357-010-9140-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Accepted: 03/15/2010] [Indexed: 05/29/2023]
Abstract
At the moment, there is no clear molecular explanation for the steeper decline in muscle performance after menopause or the mechanisms of counteractive treatments. The goal of this genome-wide study was to identify the genes and gene clusters through which power training (PT) comprising jumping activities or estrogen containing hormone replacement therapy (HRT) may affect skeletal muscle properties after menopause. We used musculus vastus lateralis samples from early stage postmenopausal (50-57 years old) women participating in a yearlong randomized double-blind placebo-controlled trial with PT and HRT interventions. Using microarray platform with over 24,000 probes, we identified 665 differentially expressed genes. The hierarchical clustering method was used to assort the genes. Additionally, enrichment analysis of gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways was carried out to clarify whether assorted gene clusters are enriched with particular functional categories. The analysis revealed transcriptional regulation of 49 GO/KEGG categories. PT upregulated transcription in "response to contraction"-category revealing novel candidate genes for contraction-related regulation of muscle function while HRT upregulated gene expression related to functionality of mitochondria. Moreover, several functional categories tightly related to muscle energy metabolism, development, and function were affected regardless of the treatment. Our results emphasize that during the early stages of the postmenopause, muscle properties are under transcriptional modulation, which both PT and HRT partially counteract leading to preservation of muscle power and potentially reducing the risk for aging-related muscle weakness. More specifically, PT and HRT may function through improving energy metabolism, response to contraction as well as by preserving functionality of the mitochondria.
Collapse
Affiliation(s)
- Eija Pöllänen
- Gerontology Research Centre, University Jyväskylä, Jyväskylä, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Boldrin L, Muntoni F, Morgan JE. Are human and mouse satellite cells really the same? J Histochem Cytochem 2010; 58:941-55. [PMID: 20644208 DOI: 10.1369/jhc.2010.956201] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Satellite cells are quiescent cells located under the basal lamina of skeletal muscle fibers that contribute to muscle growth, maintenance, repair, and regeneration. Mouse satellite cells have been shown to be muscle stem cells that are able to regenerate muscle fibers and self-renew. As human skeletal muscle is also able to regenerate following injury, we assume that the human satellite cell is, like its murine equivalent, a muscle stem cell. In this review, we compare human and mouse satellite cells and highlight their similarities and differences. We discuss gaps in our knowledge of human satellite cells, compared with that of mouse satellite cells, and suggest ways in which we may advance studies on human satellite cells, particularly by finding new markers and attempting to re-create the human satellite cell niche in vitro.
Collapse
Affiliation(s)
- Luisa Boldrin
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom.
| | | | | |
Collapse
|
97
|
Macharia R, Otto A, Valasek P, Patel K. Neuromuscular junction morphology, fiber-type proportions, and satellite-cell proliferation rates are altered in MyoD(-/-) mice. Muscle Nerve 2010; 42:38-52. [PMID: 20544915 DOI: 10.1002/mus.21637] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene compensation by members of the myogenic regulatory factor (MRF) family has been proposed to explain the apparent normal adult phenotype of MyoD(-/-) mice. Nerve and field stimulation were used to investigate contraction properties of muscle from MyoD(-/-) mice, and molecular approaches were used to investigate satellite-cell behavior. We demonstrate that MyoD deletion results in major alterations in the organization of the neuromuscular junction, which have a dramatic influence on the physiological contractile properties of skeletal muscle. Second, we show that the lineage progression of satellite cells (especially initial proliferation) in the absence of MyoD is abnormal and linked to perturbations in the nuclear localization of beta-catenin, a key readout of canonical Wnt signaling. These results show that MyoD has unique functions in both developing and adult skeletal muscle that are not carried out by other members of the MRF family.
Collapse
Affiliation(s)
- Raymond Macharia
- Department of Veterinary Basic Sciences, Royal Veterinary College, London, NW1 0TU, UK
| | | | | | | |
Collapse
|
98
|
Harada A, Okada S, Odawara J, Kumamaru H, Saiwai H, Aoki M, Nakamura M, Nishiyama Y, Ohkawa Y. Production of a rat monoclonal antibody specific for Myf5. Hybridoma (Larchmt) 2010; 29:59-62. [PMID: 20199153 DOI: 10.1089/hyb.2009.0066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Myogenic regulatory factors (MRFs) are transcription factors that possess a characteristic basic helix-loop-helix domain. Myf5, MyoD, MRF4, and myogenin are well-known MRF family members that activate muscle-specific genes during differentiation. Myf5 is expressed first among MRFs at the very early phase and plays an important role in myoblast specificity and cell proliferation. Myf5 shares high homology with MyoD, and therefore some commercial Myf5 antibodies are cross-reactive for Myf5 and MyoD. To allow for detailed studies of the function of Myf5, we generated a monoclonal antibody specific for Myf5 utilizing a rat medial iliac lymph node method. Immunoblot analysis using our monoclonal antibody enabled us to identify Myf5 protein from rat myoblast L6E9 cell extract. Moreover, cell immunostaining revealed the nuclear localization of Myf5 in the L6E9 cells. This monoclonal antibody against Myf5 will allow us to perform further detailed studies of Myf5 and Myf5 function.
Collapse
Affiliation(s)
- Akihito Harada
- SSP Stem Cell Unit, Faculty of Medicine, Kyushu University , Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Gill R, Hitchins L, Fletcher F, Dhoot GK. Sulf1A and HGF regulate satellite-cell growth. J Cell Sci 2010; 123:1873-83. [PMID: 20442248 PMCID: PMC2873224 DOI: 10.1242/jcs.061242] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2010] [Indexed: 12/24/2022] Open
Abstract
The role of Sulf1A, sulfation and hepatocyte growth factor (HGF) in satellite-cell growth was examined in an in vitro model of dissociated whole skeletal muscle fibres. Pax7-positive quiescent satellite cells express little or no Sulf1A but show rapid re-expression in regenerating myoblasts and myotubes, similar to embryonic muscle and in vitro satellite cells preceding asynchronous MyoD activation. Once activated, Sulf1A and MyoD re-expression persists up to 72 hours in most satellite cells under normal culture conditions and following moderate changes in sulfation, whereas Sulf1A neutralisation by antibodies not only enhances satellite-cell proliferation but also downregulates MyoD and Pax7 expression in a large proportion of the satellite cells. The HGF exposure also induces similar but even more pronounced changes characterised by variable sulfation levels and rapid downregulation of MyoD and Pax7 without myogenin activation in a sub-set of cells. This Pax7-MyoD-myogenin-negative sub-population expresses Sulf1A and Myf5. The transfer of all such satellite-cell progenies onto gelatin-coated-substratum re-activates MyoD and Pax7 gene expression in all cells, thus detecting a distinct sub-population of satellite cells. We conclude that HGF and fine-tuned sulfation levels are major contributory factors controlling satellite-cell growth by regulating the relative activities of actively proliferating and differentiating cells.
Collapse
Affiliation(s)
- Roop Gill
- Department of Basic Sciences, The Royal Veterinary College, University of London, Royal College Street, London, NW1 0TU, UK
| | - Laura Hitchins
- Department of Basic Sciences, The Royal Veterinary College, University of London, Royal College Street, London, NW1 0TU, UK
| | - Fenella Fletcher
- Department of Basic Sciences, The Royal Veterinary College, University of London, Royal College Street, London, NW1 0TU, UK
| | - Gurtej K. Dhoot
- Department of Basic Sciences, The Royal Veterinary College, University of London, Royal College Street, London, NW1 0TU, UK
| |
Collapse
|
100
|
Tsivitse S. Notch and Wnt signaling, physiological stimuli and postnatal myogenesis. Int J Biol Sci 2010; 6:268-81. [PMID: 20567496 PMCID: PMC2878172 DOI: 10.7150/ijbs.6.268] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 05/13/2010] [Indexed: 12/18/2022] Open
Abstract
Adult skeletal muscle stem cells, termed satellite cells are imperative to muscle regeneration. Much work has been performed on satellite cell identification and the subsequent activation of the myogenic response but the regulation of satellite cells including its activation is not well elucidated. The purpose of this review article is to synthesize what the literature reveals in regards to the current understanding of satellite cells including their contribution to muscle repair and growth following physiological stimuli. In addition, this review article will describe the recent findings on the roles of the classic developmental signaling pathways, Notch and Wnt, to the myogenic response in various muscle injury models. This purpose of this summary is to bring awareness of the impact that muscle contraction models have on the local and systemic environment of adult muscle stem cells which will be beneficial for comprehending and treatment development for muscle -associated ailments and other organ diseases.
Collapse
Affiliation(s)
- Susan Tsivitse
- Department of Kinesiology, Exercise Physiology Laboratory, University North Carolina-Charlotte, NC 28223, USA.
| |
Collapse
|