301
|
Darabi R, Baik J, Clee M, Kyba M, Tupler R, Perlingeiro RCR. Engraftment of embryonic stem cell-derived myogenic progenitors in a dominant model of muscular dystrophy. Exp Neurol 2009; 220:212-6. [PMID: 19682990 PMCID: PMC2761496 DOI: 10.1016/j.expneurol.2009.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 07/24/2009] [Accepted: 08/06/2009] [Indexed: 02/01/2023]
Abstract
Muscular dystrophies (MDs) consist of a genetically heterogeneous group of disorders, recessive or dominant, characterized by progressive skeletal muscle weakening. To date, no effective treatment is available. Experimental strategies pursuing muscle regeneration through the transplantation of stem cell preparations have brought hope to patients affected by this disorder. Efficacy has been demonstrated in recessive MD models through contribution of wild-type nuclei to the muscle fiber heterokaryon; however, to date, there has been no study investigating the efficacy of a cell therapy in a dominant model of MD. We have recently demonstrated that Pax3-induced embryonic stem (ES) cell-derived myogenic progenitors are able to engraft and improve muscle function in mdx mice, a recessive mouse model for Duchenne MD. To assess whether this therapeutic effect can be extended to a dominant type of muscle disorder, here we transplanted these cells into FRG1 transgenic mice, a dominant model that has been associated with facioscapulohumeral muscular dystrophy. Our results show that Pax3-induced ES-derived myogenic progenitors are capable of significant engraftment after intramuscular or systemic transplantation into Frg1 mice. Analyses of contractile parameters revealed functional improvement in treated muscles of male mice, but not females, which are less severely affected. This study is the first to use Frg1 transgenic mice to assess muscle regeneration as well as to support the use of a cell-based therapy for autosomal dominant types of MD.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Disease Models, Animal
- Female
- Genes, Dominant/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Intercellular Signaling Peptides and Proteins/pharmacology
- Intercellular Signaling Peptides and Proteins/therapeutic use
- Male
- Mice
- Mice, Transgenic
- Microfilament Proteins
- Muscle Development/genetics
- Muscle Weakness/genetics
- Muscle Weakness/metabolism
- Muscle Weakness/surgery
- Muscle, Skeletal/cytology
- Muscle, Skeletal/physiology
- Muscle, Skeletal/surgery
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/surgery
- Nuclear Proteins/genetics
- PAX3 Transcription Factor
- Paired Box Transcription Factors/metabolism
- Paired Box Transcription Factors/pharmacology
- Paired Box Transcription Factors/therapeutic use
- RNA-Binding Proteins
- Regeneration/genetics
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/transplantation
- Sex Characteristics
- Stem Cell Transplantation/methods
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Radbod Darabi
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | |
Collapse
|
302
|
Quattrocelli M, Cassano M, Crippa S, Perini I, Sampaolesi M. Cell therapy strategies and improvements for muscular dystrophy. Cell Death Differ 2009; 17:1222-9. [DOI: 10.1038/cdd.2009.160] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
303
|
Pallafacchina G, François S, Regnault B, Czarny B, Dive V, Cumano A, Montarras D, Buckingham M. An adult tissue-specific stem cell in its niche: a gene profiling analysis of in vivo quiescent and activated muscle satellite cells. Stem Cell Res 2009; 4:77-91. [PMID: 19962952 DOI: 10.1016/j.scr.2009.10.003] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 10/21/2009] [Accepted: 10/21/2009] [Indexed: 01/15/2023] Open
Abstract
The satellite cell of skeletal muscle provides a paradigm for quiescent and activated tissue stem cell states. We have carried out transcriptome analyses on satellite cells purified by flow cytometry from Pax3(GFP/+) mice. We compared samples from adult skeletal muscles where satellite cells are mainly quiescent, with samples from growing muscles or regenerating (mdx) muscles, where they are activated. Analysis of regulation that is shared by both activated states avoids other effects due to immature or pathological conditions. This in vivo profile differs from that of previously analyzed satellite cells activated after cell culture. It reveals how the satellite cell protects itself from damage and maintains quiescence, while being primed for activation on receipt of the appropriate signal. This is illustrated by manipulation of the corepressor Dach1, and by the demonstration that quiescent satellite cells are better protected from oxidative stress than those from mdx or 1-week-old muscles. The quiescent versus in vivo activated comparison also gives new insights into how the satellite cell controls its niche on the muscle fiber through cell adhesion and matrix remodeling. The latter also potentiates growth factor activity through proteoglycan modification. Dismantling the extracellular matrix is important for satellite cell activation when the expression of proteinases is up-regulated, whereas transcripts for their inhibitors are high in quiescent cells. In keeping with this, we demonstrate that metalloproteinase function is required for efficient regeneration in vivo.
Collapse
Affiliation(s)
- Giorgia Pallafacchina
- Molecular Genetics of Development Unit, Department of Developmental Biology, URA CNRS 2578, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
304
|
Albrecht DE, Garg N, Rufibach LE, Williams BA, Monnier N, Hwang E, Mittal P. 3rd Annual Dysferlin Conference 2-5 June 2009, Boston, Massachusetts, USA. Neuromuscul Disord 2009; 19:867-73. [PMID: 19781937 DOI: 10.1016/j.nmd.2009.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Indexed: 11/26/2022]
Affiliation(s)
- Douglas E Albrecht
- Jain Foundation Inc., 2310 130th Ave. NE, Suite B101, Bellevue, Washington 98005, USA
| | | | | | | | | | | | | |
Collapse
|
305
|
Yang Z, Zeng Q, Ma Z, Wang Y, Xu X. Tracking dynamics of muscle engraftment in small animals by in vivo fluorescent imaging. J Vis Exp 2009:1388. [PMID: 19770816 PMCID: PMC3150052 DOI: 10.3791/1388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Muscular dystrophies are a group of degenerative muscle diseases characterized by progressive loss of contractile muscle cells. Currently, there is no curative treatment available. Recent advances in stem cell biology have generated new hopes for the development of effective cell based therapies to treat these diseases. Transplantation of various types of stem cells labeled with fluorescent proteins into muscles of dystrophic animal models has been used broadly in the field. A non-invasive technique with the capability to track the transplanted cell fate longitudinally can further our ability to evaluate muscle engraftment by transplanted cells more accurately and efficiently. In the present study, we demonstrate that in vivo fluorescence imaging is a sensitive and reliable method for tracking transplanted GFP (Green Fluorescent Protein)-labeled cells in mouse skeletal muscles. Despite the concern about background due to the use of an external light necessary for excitation of fluorescent protein, we found that by using either nude mouse or eliminating hair with hair removal reagents much of this problem is eliminated. Using a CCD camera, the fluorescent signal can be detected in the tibialis anterior (TA) muscle after injection of 5 x 10(5) cells from either GFP transgenic mice or eGFP transduced myoblast culture. For more superficial muscles such as the extensor digitorum longus (EDL), injection of fewer cells produces a detectable signal. Signal intensity can be measured and quantified as the number of emitted photons per second in a region of interest (ROI). Since the acquired images show clear boundaries demarcating the engrafted area, the size of the ROI can be measured as well. If the legs are positioned consistently every time, the changes in total number of photons per second per muscle and the size of the ROI reflect the changes in the number of engrafted cells and the size of the engrafted area. Therefore the changes in the same muscle over time are quantifiable. In vivo fluorescent imaging technique has been used primarily to track the growth of tumorogenic cells, our study shows that it is a powerful tool that enables us to track the fate of transplanted stem cells.
Collapse
Affiliation(s)
- Zhong Yang
- Department of Anesthesia, Brigham and Woman's Hospital, USA
| | | | | | | | | |
Collapse
|
306
|
Le Grand F, Jones AE, Seale V, Scimè A, Rudnicki MA. Wnt7a activates the planar cell polarity pathway to drive the symmetric expansion of satellite stem cells. Cell Stem Cell 2009; 4:535-47. [PMID: 19497282 DOI: 10.1016/j.stem.2009.03.013] [Citation(s) in RCA: 363] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 02/12/2009] [Accepted: 03/19/2009] [Indexed: 12/18/2022]
Abstract
Satellite cells in skeletal muscle are a heterogeneous population of stem cells and committed progenitors. We found that quiescent satellite stem cells expressed the Wnt receptor Fzd7 and that its candidate ligand Wnt7a was upregulated during regeneration. Wnt7a markedly stimulated the symmetric expansion of satellite stem cells but did not affect the growth or differentiation of myoblasts. Silencing of Fzd7 abrogated Wnt7a binding and stimulation of stem cell expansion. Wnt7a signaling induced the polarized distribution of the planar cell polarity effector Vangl2. Silencing of Vangl2 inhibited Wnt7a action on satellite stem cell expansion. Wnt7a overexpression enhanced muscle regeneration and increased both satellite cell numbers and the proportion of satellite stem cells. Muscle lacking Wnt7a exhibited a marked decrease in satellite cell number following regeneration. Therefore, Wnt7a signaling through the planar cell polarity pathway controls the homeostatic level of satellite stem cells and hence regulates the regenerative potential of muscle.
Collapse
Affiliation(s)
- Fabien Le Grand
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | | | | | | | | |
Collapse
|
307
|
Cosgrove BD, Sacco A, Gilbert PM, Blau HM. A home away from home: challenges and opportunities in engineering in vitro muscle satellite cell niches. Differentiation 2009; 78:185-94. [PMID: 19751902 PMCID: PMC2801624 DOI: 10.1016/j.diff.2009.08.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 08/11/2009] [Indexed: 12/24/2022]
Abstract
Satellite cells are skeletal muscle stem cells with a principal role in postnatal skeletal muscle regeneration. Satellite cells, like many tissue-specific adult stem cells, reside in a quiescent state in an instructive, anatomically defined niche. The satellite cell niche constitutes a distinct membrane-enclosed compartment within the muscle fiber, containing a diversity of biochemical and biophysical signals that influence satellite cell function. A major limitation to the study and clinical utility of satellite cells is that upon removal from the muscle fiber and plating in traditional plastic tissue culture platforms, their muscle stem cell properties are rapidly lost. Clearly, the maintenance of stem cell function is critically dependent on in vivo niche signals, highlighting the need to create novel in vitro microenvironments that allow for the maintenance and propagation of satellite cells while retaining their potential to function as muscle stem cells. Here, we discuss how emerging biomaterials technologies offer great promise for engineering in vitro microenvironments to meet these challenges. In engineered biomaterials, signaling molecules can be presented in a manner that more closely mimics cell-cell and cell-matrix interactions, and matrices can be fabricated with diverse rigidities that approximate in vivo tissues. The development of in vitro microenvironments in which niche features can be systematically modulated will be instrumental not only to future insights into muscle stem cell biology and therapeutic approaches to muscle diseases and muscle wasting with aging, but also will provide a paradigm for the analysis of numerous adult tissue-specific stem cells.
Collapse
Affiliation(s)
- Benjamin D. Cosgrove
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Alessandra Sacco
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Penney M. Gilbert
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen M. Blau
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
308
|
Lepper C, Conway SJ, Fan CM. Adult satellite cells and embryonic muscle progenitors have distinct genetic requirements. Nature 2009; 460:627-31. [PMID: 19554048 DOI: 10.1038/nature08209] [Citation(s) in RCA: 412] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Accepted: 06/16/2009] [Indexed: 11/09/2022]
Abstract
Myogenic potential, survival and expansion of mammalian muscle progenitors depend on the myogenic determinants Pax3 and Pax7 embryonically, and Pax7 alone perinatally. Several in vitro studies support the critical role of Pax7 in these functions of adult muscle stem cells (satellite cells), but a formal demonstration has been lacking in vivo. Here we show, through the application of inducible Cre/loxP lineage tracing and conditional gene inactivation to the tibialis anterior muscle regeneration paradigm, that, unexpectedly, when Pax7 is inactivated in adult mice, mutant satellite cells are not compromised in muscle regeneration, they can proliferate and reoccupy the sublaminal satellite niche, and they are able to support further regenerative processes. Dual adult inactivation of Pax3 and Pax7 also results in normal muscle regeneration. Multiple time points of gene inactivation reveal that Pax7 is only required up to the juvenile period when progenitor cells make the transition into quiescence. Furthermore, we demonstrate a cell-intrinsic difference between neonatal progenitor and adult satellite cells in their Pax7-dependency. Our finding of an age-dependent change in the genetic requirement for muscle stem cells cautions against inferring adult stem-cell biology from embryonic studies, and has direct implications for the use of stem cells from hosts of different ages in transplantation-based therapy.
Collapse
Affiliation(s)
- Christoph Lepper
- Department of Embryology, Carnegie Institution, 3520 San Martin Drive, Baltimore, Maryland 21218, USA.
| | | | | |
Collapse
|
309
|
Abstract
The last 20 years have witnessed major advances in the understanding of muscle diseases and significant inroads are being made to treat muscular dystrophy. However, no curative therapy is currently available for any of the muscular dystrophies, despite the immense progress made using several approaches and only palliative and symptomatic treatment is available for patients. The discovery of miRNAs as new and important regulators of gene expression is expected to broaden our biological understanding of the regulatory mechanism in muscle by adding another dimension of regulation to the diversity and complexity of gene-regulatory networks. As important regulators of muscle development, unravelling the regulatory circuits involved may be challenging, given that a single miRNA can regulate the expression of many mRNA targets. Although the identification of the regulatory targets of miRNAs in muscle is a challenge, it will be critical for placing them in genetic pathways and biological contexts. Therefore, combining informatics, biochemical and genetic approaches will not only expected to reveal the elucidation of the miRNA regulatory network in skeletal muscle and to bring a better knowledge on muscle tissue regulation but will also raise new opportunities for therapeutic intervention in muscular dystrophies by identifying candidate miRNAs as potential targets for clinical application.
Collapse
Affiliation(s)
- Iris Eisenberg
- Howard Hughes Medical Institute, Childrens Hospital Boston, Boston, MA 02115, USA
| | | | | |
Collapse
|
310
|
Wang Z, Chamberlain JS, Tapscott SJ, Storb R. Gene therapy in large animal models of muscular dystrophy. ILAR J 2009; 50:187-98. [PMID: 19293461 DOI: 10.1093/ilar.50.2.187] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The muscular dystrophies are a group of genetically and phenotypically heterogeneously inherited diseases characterized by progressive muscle wasting, which can lead to premature death in severe forms such as Duchenne muscular dystrophy (DMD). In many cases they are caused by the absence of proteins that are critical components of the dystrophin-glycoprotein complex, which links the cytoskeleton and the basal lamina. There is no effective treatment for these disorders at present, but several novel strategies for replacing or repairing the defective gene are in development, with early encouraging results from animal models. We review these strategies, which include the use of stem cells of different tissue origins, gene replacement therapies mediated by various viral vectors, and transcript repair treatments using exon skipping strategies. We comment on their advantages and on limitations that must be overcome before successful application to human patients. Our focus is on studies in a clinically relevant large canine model of DMD. Recent advances in the field suggest that effective therapies for muscular dystrophies are on the horizon. Because of the complex nature of these diseases, it may be necessary to combine multiple approaches to achieve a successful treatment.
Collapse
Affiliation(s)
- Zejing Wang
- Division of Clinical Research, Fred Hutchinson Cancer Research Center in Seattle, Washington 98109, USA
| | | | | | | |
Collapse
|
311
|
Jørgensen LH, Larochelle N, Orlopp K, Dunant P, Dudley RW, Stucka R, Thirion C, Walter MC, Laval SH, Lochmüller H. Efficient and Fast Functional Screening of Microdystrophin ConstructsIn VivoandIn Vitrofor Therapy of Duchenne Muscular Dystrophy. Hum Gene Ther 2009; 20:641-50. [DOI: 10.1089/hum.2008.162] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Louise H. Jørgensen
- Institute of Human Genetics, University of Newcastle, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Nancy Larochelle
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Kristian Orlopp
- Friedrich Baur Institute and Department of Neurology, Ludwig Maximilians University, Munich 81377, Germany
| | - Patrick Dunant
- Friedrich Baur Institute and Department of Neurology, Ludwig Maximilians University, Munich 81377, Germany
| | - Roy W.R. Dudley
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Rolf Stucka
- Friedrich Baur Institute and Department of Neurology, Ludwig Maximilians University, Munich 81377, Germany
| | - Christian Thirion
- Friedrich Baur Institute and Department of Neurology, Ludwig Maximilians University, Munich 81377, Germany
| | - Maggie C. Walter
- Friedrich Baur Institute and Department of Neurology, Ludwig Maximilians University, Munich 81377, Germany
| | - Steven H. Laval
- Institute of Human Genetics, University of Newcastle, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Hanns Lochmüller
- Institute of Human Genetics, University of Newcastle, Newcastle upon Tyne NE1 3BZ, United Kingdom
- Friedrich Baur Institute and Department of Neurology, Ludwig Maximilians University, Munich 81377, Germany
| |
Collapse
|
312
|
Kumar D, Shadrach JL, Wagers AJ, Lassar AB. Id3 is a direct transcriptional target of Pax7 in quiescent satellite cells. Mol Biol Cell 2009; 20:3170-7. [PMID: 19458195 DOI: 10.1091/mbc.e08-12-1185] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Pax7 is a key regulator of skeletal muscle stem cells and is required along with Pax3 to generate skeletal muscle precursors. We have identified a collection of genes induced by either Pax3 or Pax7 in C2C12 muscle cells. Two notable Pax3/7 targets are the inhibitory helix-loop-helix (HLH) proteins inhibitor of DNA binding (Id) 2 and Id3, both of which are coordinately expressed with Pax7 in quiescent satellite cells and are induced in quiescent C2C12 myogenic cells after ectopic expression of either Pax3 or Pax7. Ectopic Pax7 activates expression of a luciferase reporter driven by the Id3 promoter, and maximal induction of this reporter requires a conserved Pax7 binding site located upstream of the Id3 gene. Chromatin immunoprecipitation indicated that Pax7 is bound upstream of the Id3 promoter in quiescent satellite cells. In addition, short hairpin RNA-mediated knockdown of Pax7 expression in cultured satellite cells coordinately decreased both Id2 and Id3 expression. Together, these findings indicate that Id3 is a direct transcriptional target for Pax7 in quiescent satellite cells, and they suggest that Pax7 acts to block premature differentiation of quiescent satellite cells by inducing the expression of Id2 and Id3, which in turn may act to block either the precocious induction of myogenic basic (b)HLH proteins, the activity of myogenic bHLH proteins, or both.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
313
|
Gang EJ, Darabi R, Bosnakovski D, Xu Z, Kamm KE, Kyba M, Perlingeiro RCR. Engraftment of mesenchymal stem cells into dystrophin-deficient mice is not accompanied by functional recovery. Exp Cell Res 2009; 315:2624-36. [PMID: 19460366 DOI: 10.1016/j.yexcr.2009.05.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 05/05/2009] [Accepted: 05/06/2009] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cell preparations have been proposed for muscle regeneration in musculoskeletal disorders. Although MSCs have great in vitro expansion potential and possess the ability to differentiate into several mesenchymal lineages, myogenesis has proven to be much more difficult to induce. We have recently demonstrated that Pax3, the master regulator of the embryonic myogenic program, enables the in vitro differentiation of a murine mesenchymal stem cell line (MSCB9-Pax3) into myogenic progenitors. Here we show that injection of these cells into cardiotoxin-injured muscles of immunodeficient mice leads to the development of muscle tumors, resembling rhabdomyosarcomas. We then extended these studies to primary human mesenchymal stem cells (hMSCs) isolated from bone marrow. Upon genetic modification with a lentiviral vector encoding PAX3, hMSCs activated the myogenic program as demonstrated by expression of myogenic regulatory factors. Upon transplantation, the PAX3-modified MSCs did not generate rhabdomyosarcomas but rather, resulted in donor-derived myofibers. These were found at higher frequency in PAX3-transduced hMSCs than in mock-transduced MSCs. Nonetheless, neither engraftment of PAX3-modified or unmodified MSCs resulted in improved contractility. Thus these findings suggest that limitations remain to be overcome before MSC preparations result in effective treatment for muscular dystrophies.
Collapse
Affiliation(s)
- Eun Ji Gang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9133, USA
| | | | | | | | | | | | | |
Collapse
|
314
|
Gayraud-Morel B, Chrétien F, Tajbakhsh S. Skeletal muscle as a paradigm for regenerative biology and medicine. Regen Med 2009; 4:293-319. [PMID: 19317647 DOI: 10.2217/17460751.4.2.293] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Tissue development and regeneration share common features, since modules of regulatory pathways and transcription factors that are crucial for prenatal development are redeployed for tissue reconstruction after trauma. Regenerative medicine has therefore gained important insights through the study of developmental and regenerative biology. Moreover, diverse experimental models have been used to investigate the regeneration process in different tissues and organs. Paradoxically, little is known regarding the relative contribution of stem cells with respect to the supporting tissue during tissue regeneration. Particular attention will be given to mouse models using distinct injury paradigms to investigate the regenerative biology of skeletal muscle. An understanding of the response of stem and parenchymal cells is crucial for the development of clinical strategies to combat the normal decline in tissue performance during aging or its reconstitution after trauma and during disease. This review addresses these issues, focusing on muscle regeneration and how different factors, including genes, cells and the environment, impinge on this process.
Collapse
Affiliation(s)
- Barbara Gayraud-Morel
- Stem Cells & Development, Department of Developmental Biology, Pasteur Institute, CNRS URA 2578, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | | | | |
Collapse
|
315
|
Tanaka KK, Hall JK, Troy AA, Cornelison DDW, Majka SM, Olwin BB. Syndecan-4-expressing muscle progenitor cells in the SP engraft as satellite cells during muscle regeneration. Cell Stem Cell 2009; 4:217-25. [PMID: 19265661 DOI: 10.1016/j.stem.2009.01.016] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2008] [Revised: 12/03/2008] [Accepted: 01/21/2009] [Indexed: 10/21/2022]
Abstract
Skeletal muscle satellite cells, located between the basal lamina and plasma membrane of myofibers, are required for skeletal muscle regeneration. The capacity of satellite cells as well as other cell lineages including mesoangioblasts, mesenchymal stem cells, and side population (SP) cells to contribute to muscle regeneration has complicated the identification of a satellite stem cell. We have characterized a rare subset of the muscle SP that efficiently engrafts into the host satellite cell niche when transplanted into regenerating muscle, providing 75% of the satellite cell population and 30% of the myonuclear population, respectively. These cells are found in the satellite cell position, adhere to isolated myofibers, and spontaneously undergo myogenesis in culture. We propose that this subset of SP cells (satellite-SP cells), characterized by ABCG2, Syndecan-4, and Pax7 expression, constitutes a self-renewing muscle stem cell capable of generating both satellite cells and their myonuclear progeny in vivo.
Collapse
|
316
|
Abstract
Multiple myogenic populations have been highlighted in past publications. In this issue of Cell Stem Cell, Tanaka et al. (2009) advance our understanding of the cells that contribute to muscle regeneration by identifying an ABCG2-expressing population that exhibits excellent engraftment potential, particularly within the satellite cell niche.
Collapse
Affiliation(s)
- Stefani Fontana
- Department of Pediatrics and Neurology, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, BRB 529 Baltimore, MD 21205, USA
| | | |
Collapse
|
317
|
Jia Y, Warin R, Yu X, Epstein R, Noguchi CT. Erythropoietin signaling promotes transplanted progenitor cell survival. FASEB J 2009; 23:3089-99. [PMID: 19417086 DOI: 10.1096/fj.09-130237] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We examine the potential for erythropoietin signaling to promote donor cell survival in a model of myoblast transplantation. Expression of a truncated erythropoietin receptor in hematopoietic stem cells has been shown to promote selective engraftment in mice. We previously demonstrated expression of endogenous erythropoietin receptor on murine myoblasts, and erythropoietin treatment can stimulate myoblast proliferation and delay differentiation. Here, we report that enhanced erythropoietin receptor expression, as well as exogenous erythropoietin treatment in myoblasts, provided a survival advantage and protection against apoptosis under serum-starvation conditions. When cultured in differentiation medium, expression of the myogenic regulatory proteins shifted toward early differentiation with increased erythropoietin receptor. Expression of early myogenic differentiation proteins Myf-5 and MyoD increased, while later stage protein myogenin decreased. Transplantation of C2C12 myoblasts overexpressing truncated erythropoietin receptor showed more transplanted cell incorporation into muscle fibers in muscular dystrophy mdx mice. These cells also restored dystrophin protein expression in mdx mice at 6 wk after cell treatment that was further increased with exogenous erythropoietin administration. In summary, enhanced erythropoietin receptor expression promotes transplanted cell survival in a mouse model for myoblast transplantation and provides dystrophin expression in mice with muscular dystrophy.
Collapse
Affiliation(s)
- Yi Jia
- Molecular Medicine Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1822, USA
| | | | | | | | | |
Collapse
|
318
|
Arnett ALH, Chamberlain JR, Chamberlain JS. Therapy for neuromuscular disorders. Curr Opin Genet Dev 2009; 19:290-7. [PMID: 19411172 DOI: 10.1016/j.gde.2009.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 03/17/2009] [Accepted: 03/20/2009] [Indexed: 12/21/2022]
Abstract
Research into therapeutic approaches for both recessive and dominant neuromuscular disorders has made great progress over the past few years. In the field of gene therapy, antisense-mediated exon skipping is being applied to bypass deleterious mutations in the dystrophin gene and restore dystrophin expression in animal models of muscular dystrophy. Approaches for the dominant genetic muscle diseases have turned toward elimination of the mutant gene product with anti-sense oligonucleotide therapy and RNA interference techniques. Refinements of adeno-associated viral vectors and strategies for their delivery are also leading towards future clinical trials. The discovery of new, multipotent cell lineages, some of which possess the ability to successfully engraft muscle following vascular delivery, presents exciting prospects for the field of stem cell therapy. These discoveries represent steady progress towards the development of effective therapies for a wide range of neuromuscular disorders.
Collapse
Affiliation(s)
- Andrea L H Arnett
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195-7720, United States.
| | | | | |
Collapse
|
319
|
Pawlikowski B, Lee L, Zuo J, Kramer RH. Analysis of human muscle stem cells reveals a differentiation-resistant progenitor cell population expressing Pax7 capable of self-renewal. Dev Dyn 2009; 238:138-49. [PMID: 19097049 DOI: 10.1002/dvdy.21833] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Studies using mouse models have established a critical role for resident satellite stem cells in skeletal muscle development and regeneration, but little is known about this paradigm in human muscle. Here, using human muscle stem cells, we address their lineage progression, differentiation, migration, and self-renewal. Isolated human satellite cells expressed alpha7-integrin and other definitive muscle markers, were highly motile on laminin substrates and could undergo efficient myotube differentiation and myofibrillogenesis. However, only a subpopulation of the myoblasts expressed Pax7 and displayed a variable lineage progression as measured by desmin and MyoD expression. Analysis identified a differentiation-resistant progenitor cell population that was Pax7+/desmin- and capable of self-renewal. This study extends our understanding of the role of Pax7 in regulating human satellite stem cell differentiation and self-renewal.
Collapse
Affiliation(s)
- Bradley Pawlikowski
- Department of Cell and Tissue Biology, University of California San Francisco, School of Dentistry, San Francisco, California 94143, USA.
| | | | | | | |
Collapse
|
320
|
Germano D, Blyszczuk P, Valaperti A, Kania G, Dirnhofer S, Landmesser U, Lüscher TF, Hunziker L, Zulewski H, Eriksson U. Prominin-1/CD133+ lung epithelial progenitors protect from bleomycin-induced pulmonary fibrosis. Am J Respir Crit Care Med 2009; 179:939-49. [PMID: 19234103 DOI: 10.1164/rccm.200809-1390oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
RATIONALE The mouse model of bleomycin-induced lung injury offers an approach to study idiopathic pulmonary fibrosis, a progressive interstitial lung disease with poor prognosis. Progenitor cell-based treatment strategies might combine antiinflammatory effects and the capacity for tissue repair. OBJECTIVES To expand progenitor cells with reparative and regenerative capacities and to evaluate their protective effects on pulmonary fibrosis in vivo. METHODS Prominin-1/CD133(+) epithelial progenitor cells (PEPs) were expanded from adult mouse lungs after digestion and culture of distal airways. Lung fibrosis was induced in C57Bl/6 mice by instillation of bleomycin. Two hours later, animals were transplanted with PEPs. Inflammation and fibrosis were assessed by immunohistochemistry, bronchoalveolar lavage fluid differentials, and real-time polymerase chain reaction. MEASUREMENTS AND MAIN RESULTS PEPs expanded from mouse lungs were of bone marrow origin, coexpressed stem and hematopoietic cell markers, and differentiated in vitro into alveolar type II surfactant protein-C(+) epithelial cells. In bleomycin-challenged mice, intratracheally injected PEPs engrafted into the lungs and differentiated into type II pneumocytes. Furthermore, PEPs suppressed proinflammatory and profibrotic gene expression, prevented the recruitment of inflammatory cells, and protected bleomycin-challenged mice from pulmonary fibrosis. Mechanistically, the protective effect depended on upregulation of inducible nitric oxide synthase in PEPs and nitric oxide-mediated suppression of alveolar macrophage proliferation. Accordingly, PEPs from iNOS(-/-) but not iNOS(+/+) mice failed to protect from bleomycin-induced lung injury. CONCLUSIONS The combined antiinflammatory and regenerative capacity of bone marrow-derived pulmonary epithelial progenitors offers a promising approach for development of cell-based therapeutic strategies against pulmonary fibrosis.
Collapse
Affiliation(s)
- Davide Germano
- Experimental Critical Care Medicine, Department of Biomedicine, University of Basel, Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
321
|
Bartel DP, Nakamura I, Roberts LR, Scimè A, Rudnicki MA. MicroRNAs: target recognition and regulatory functions. Cell 2009. [PMID: 23403079 DOI: 10.1016/j] [Citation(s) in RCA: 482] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are endogenous approximately 23 nt RNAs that play important gene-regulatory roles in animals and plants by pairing to the mRNAs of protein-coding genes to direct their posttranscriptional repression. This review outlines the current understanding of miRNA target recognition in animals and discusses the widespread impact of miRNAs on both the expression and evolution of protein-coding genes.
Collapse
Affiliation(s)
- David P Bartel
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | | | | | |
Collapse
|
322
|
|
323
|
Niyibizi C, Li F. Potential implications of cell therapy for osteogenesis imperfecta. ACTA ACUST UNITED AC 2009; 4:57-66. [PMID: 20490372 DOI: 10.2217/17584272.4.1.57] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Osteogenesis imperfecta (OI) is a brittle-bone disease whose hallmark is bone fragility. Since the disease is genetic, there is currently no available cure. Several pharmacological agents have been tried with not much success, except the recent use of bisphosphonates. Stem cells have been suggested as an alternative OI treatment, but many hurdles remain before this technology can be applied for treating patients with OI. This review summarizes what is known at present regarding the application of stem cells to treat OI using animal models, clinical trials using mesenchymal stem cells to treat patients with OI and the knowledge gained from the clinical trials. Application of gene therapy in combination with stem cells is also discussed. The hurdles to be overcome to bring stem cells close to the clinic and future perspectives are discussed.
Collapse
|
324
|
Degenerative muscle fiber accelerates adipogenesis of intramuscular cells via RhoA signaling pathway. Differentiation 2009; 77:350-9. [PMID: 19281783 DOI: 10.1016/j.diff.2008.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 11/13/2008] [Accepted: 11/14/2008] [Indexed: 11/23/2022]
Abstract
In some pathological conditions such as Duchenne muscular dystrophy, it has been known that a fatty infiltration in skeletal muscle is often observed and that is also one of primary factors to induce marked decline of muscular strength. However, the mechanism of fatty infiltration, cellular origin of accumulated adipocytes and its significance are not fully understood. The fact that persistent degenerative muscle fibers are present on dystrophic muscle leads us to hypothesize that muscle fiber condition affects fatty infiltration in skeletal muscle. We employed a single fiber culture system to determine whether fiber condition affects an appearance of adipocytes on the fibers. Artificially hyper-contracted muscle fibers (HCF), generated from isolated intact fibers (IF) of rat extensor digitrum longus muscle, were maintained as non-adherent cultures for 5-7 days. Interestingly, there appeared to be considerable numbers of mature adipocytes on HCF, whereas no adipocytes were seen on IF, indicating that cells on HCF spontaneously differentiated into mature adipocytes. Activation of RhoA signaling by the addition of thrombin decreased the number of adipocytes on HCF in a dose-dependent manner, whereas the number of MyoD-positive myoblasts increased. In contrast, Y-27632, a specific inhibitor of Rho kinases (ROCK), induced adipogenic differentiation of cells derived from IF. In addition, administration of Y-27632 into mouse regenerating muscle resulted in fat accumulation in the muscle. Taken together, the present studies clearly demonstrated that muscle fiber condition affects fat accumulation in skeletal muscle and that is possibly mediated by the RhoA signaling pathway.
Collapse
|
325
|
Multiplicity of experimental approaches to therapy for genetic muscle diseases and necessity for population screening. J Muscle Res Cell Motil 2008; 29:247-52. [PMID: 19115047 DOI: 10.1007/s10974-008-9158-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Accepted: 12/03/2008] [Indexed: 12/17/2022]
Abstract
Currently a multiplicity of experimental approaches to therapy for genetic muscle diseases is being investigated. These include replacement of the missing gene, manipulation of the gene message, repair of the mutation, upregulation of an alternative gene and pharmacological interventions targeting a number of systems. A number of these approaches are in current clinical trials. There is considerable anticipation that perhaps more than one of the approaches will finally prove of clinical benefit, but there are many voices of caution. No matter which approaches might ultimately prove effective, there is a consensus that for most benefit to the patients it will be necessary to start treatment as early as possible. A consensus is also developing that the only way to do this is to implement population-based newborn screening to identify affected children shortly after birth. Population-based newborn screening is currently practised in very few places in the world and it brings with it implications for prevention rather than cure of genetic muscle diseases.
Collapse
|
326
|
|
327
|
Tremblay JP, Skuk D. Another New “Super Muscle Stem Cell” Leaves Unaddressed the Real Problems of Cell Therapy for Duchenne Muscular Dystrophy. Mol Ther 2008; 16:1907-9. [DOI: 10.1038/mt.2008.243] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
328
|
Stocum DL, Zupanc GK. Stretching the limits: Stem cells in regeneration science. Dev Dyn 2008; 237:3648-71. [DOI: 10.1002/dvdy.21774] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
329
|
Cerletti M, Stevenson K, Neuberg D, Jurga S, Witczak CA, Hirshman MF, Shadrach JL, Goodyear LJ, Wagers AJ. Response: Skeletal Muscle Precursor Grafts in Dystrophic Mice. Cell 2008. [DOI: 10.1016/j.cell.2008.11.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
330
|
Urish KL, Vella JB, Okada M, Deasy BM, Tobita K, Keller BB, Cao B, Piganelli JD, Huard J. Antioxidant levels represent a major determinant in the regenerative capacity of muscle stem cells. Mol Biol Cell 2008; 20:509-20. [PMID: 19005220 DOI: 10.1091/mbc.e08-03-0274] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Stem cells are classically defined by their multipotent, long-term proliferation, and self-renewal capabilities. Here, we show that increased antioxidant capacity represents an additional functional characteristic of muscle-derived stem cells (MDSCs). Seeking to understand the superior regenerative capacity of MDSCs compared with myoblasts in cardiac and skeletal muscle transplantation, our group hypothesized that survival of the oxidative and inflammatory stress inherent to transplantation may play an important role. Evidence of increased enzymatic and nonenzymatic antioxidant capacity of MDSCs were observed in terms of higher levels of superoxide dismutase and glutathione, which appears to confer a differentiation and survival advantage. Further when glutathione levels of the MDSCs are lowered to that of myoblasts, the transplantation advantage of MDSCs over myoblasts is lost when transplanted into both skeletal and cardiac muscles. These findings elucidate an important cause for the superior regenerative capacity of MDSCs, and provide functional evidence for the emerging role of antioxidant capacity as a critical property for MDSC survival post-transplantation.
Collapse
Affiliation(s)
- Kenneth L Urish
- Department of Orthopaedics and Rehabilitation, and Department of Surgery, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
331
|
The origins of the identification and isolation of hematopoietic stem cells, and their capability to induce donor-specific transplantation tolerance and treat autoimmune diseases. Blood 2008; 112:3543-53. [PMID: 18948588 DOI: 10.1182/blood-2008-08-078220] [Citation(s) in RCA: 282] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Advances in the understanding of the cells of the hematopoietic system have provided a rich basis for improving clinical hematopoietic cell transplants; finding and using proteins and molecules to amplify or suppress particular blood cell types; understanding the stepwise progression of preleukemic stages leading first to chronic myeloid disorders, then the emergence of acute blastic leukemias; and treating malignant and nonmalignant diseases with cell subsets. As a result of intense scientific investigation, hematopoietic stem cells (HSCs) have been isolated and their key functional characteristics revealed-self-renewal and multilineage differentiation. These characteristics are now found to be present in all tissue/organ stem cell studies, and even in the analysis of pluripotent embryonic, nuclear transfer, and induced pluripotent stem cells. Studies on HSC have identified hematopoiesis as one of the best systems for studying developmental cell lineages and as the best for understanding molecular changes in cell fate decision-making and for finding preclinical and clinical platforms for tissue and organ replacement, regeneration, and oncogenesis. Here we review the steps, from our viewpoint, that led to HSC isolation and its importance in self-nonself immune recognition.
Collapse
|
332
|
An isogenetic myoblast expression screen identifies DUX4-mediated FSHD-associated molecular pathologies. EMBO J 2008; 27:2766-79. [PMID: 18833193 DOI: 10.1038/emboj.2008.201] [Citation(s) in RCA: 241] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2007] [Accepted: 09/10/2008] [Indexed: 12/18/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by an unusual deletion with neomorphic activity. This deletion derepresses genes in cis; however which candidate gene causes the FSHD phenotype, and through what mechanism, is unknown. We describe a novel genetic tool, inducible cassette exchange, enabling rapid generation of isogenetically modified cells with conditional and variable transgene expression. We compare the effects of expressing variable levels of each FSHD candidate gene on myoblasts. This screen identified only one gene with overt toxicity: DUX4 (double homeobox, chromosome 4), a protein with two homeodomains, each similar in sequence to Pax3 and Pax7. DUX4 expression recapitulates key features of the FSHD molecular phenotype, including repression of MyoD and its target genes, diminished myogenic differentiation, repression of glutathione redox pathway components, and sensitivity to oxidative stress. We further demonstrate competition between DUX4 and Pax3/Pax7: when either Pax3 or Pax7 is expressed at high levels, DUX4 is no longer toxic. We propose a hypothesis for FSHD in which DUX4 expression interferes with Pax7 in satellite cells, and inappropriately regulates Pax targets, including myogenic regulatory factors, during regeneration.
Collapse
|
333
|
Affiliation(s)
- Helen M Blau
- Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
334
|
Bosnakovski D, Xu Z, Li W, Thet S, Cleaver O, Perlingeiro RCR, Kyba M. Prospective isolation of skeletal muscle stem cells with a Pax7 reporter. Stem Cells 2008; 26:3194-204. [PMID: 18802040 DOI: 10.1634/stemcells.2007-1017] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Muscle regeneration occurs through activation of quiescent satellite cells whose progeny proliferate, differentiate, and fuse to make new myofibers. We used a transgenic Pax7-ZsGreen reporter mouse to prospectively isolate stem cells of skeletal muscle by flow cytometry. We show that Pax7-expressing cells (satellite cells) in the limb, head, and diaphragm muscles are homogeneous in size and granularity and uniformly labeled by certain cell surface markers, including CD34 and CD29. The frequency of the satellite cells varies between muscle types and with age. Clonal analysis demonstrated that all colonies arising from single cells within the Pax7-sorted fraction have myogenic potential. In response to injury, Pax7(+) cells reduce CD34, CD29, and CXCR4 expression, increase in size, and acquire Sca-1. When directly isolated and cultured in vitro, Pax7(+) cells display the hallmarks of activation and proliferate, initially as suspension aggregates and later distributed between suspension and adherence. During in vitro expansion, Pax7 (ZsGreen) and CD34 expression decline, whereas expression of PSA-NCAM is acquired. The nonmyogenic, Pax7(neg) cells expand as Sca1(+) PDGRalpha(+) PSA-NCAM(neg) cells. Satellite cells expanded exclusively in suspension can engraft and produce dystrophin(+) fibers in mdx(-/-) mice. These results establish a novel animal model for the study of muscle stem cell physiology and a culture system for expansion of engraftable muscle progenitors.
Collapse
Affiliation(s)
- Darko Bosnakovski
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | | | | | | |
Collapse
|
335
|
Sacco A, Doyonnas R, Kraft P, Vitorovic S, Blau HM. Self-renewal and expansion of single transplanted muscle stem cells. Nature 2008; 456:502-6. [PMID: 18806774 DOI: 10.1038/nature07384] [Citation(s) in RCA: 635] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Accepted: 09/01/2008] [Indexed: 01/25/2023]
Abstract
Adult muscle satellite cells have a principal role in postnatal skeletal muscle growth and regeneration. Satellite cells reside as quiescent cells underneath the basal lamina that surrounds muscle fibres and respond to damage by giving rise to transient amplifying cells (progenitors) and myoblasts that fuse with myofibres. Recent experiments showed that, in contrast to cultured myoblasts, satellite cells freshly isolated or satellite cells derived from the transplantation of one intact myofibre contribute robustly to muscle repair. However, because satellite cells are known to be heterogeneous, clonal analysis is required to demonstrate stem cell function. Here we show that when a single luciferase-expressing muscle stem cell is transplanted into the muscle of mice it is capable of extensive proliferation, contributes to muscle fibres, and Pax7(+)luciferase(+) mononucleated cells can be readily re-isolated, providing evidence of muscle stem cell self-renewal. In addition, we show using in vivo bioluminescence imaging that the dynamics of muscle stem cell behaviour during muscle repair can be followed in a manner not possible using traditional retrospective histological analyses. By imaging luciferase activity, real-time quantitative and kinetic analyses show that donor-derived muscle stem cells proliferate and engraft rapidly after injection until homeostasis is reached. On injury, donor-derived mononucleated cells generate massive waves of cell proliferation. Together, these results show that the progeny of a single luciferase-expressing muscle stem cell can both self-renew and differentiate after transplantation in mice, providing new evidence at the clonal level that self-renewal is an autonomous property of a single adult muscle stem cell.
Collapse
Affiliation(s)
- Alessandra Sacco
- Baxter Laboratory in Genetic Pharmacology, Department of Microbiology and Immunology, Stem Cell Institute, Stanford University School of Medicine, Stanford, California 94305-5175, USA
| | | | | | | | | |
Collapse
|
336
|
Mozzetta C, Minetti G, Puri PL. Regenerative pharmacology in the treatment of genetic diseases: the paradigm of muscular dystrophy. Int J Biochem Cell Biol 2008; 41:701-10. [PMID: 18804548 DOI: 10.1016/j.biocel.2008.08.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 08/18/2008] [Accepted: 08/28/2008] [Indexed: 01/21/2023]
Abstract
Current evidence supports the therapeutic potential of pharmacological interventions that counter the progression of genetic disorders by promoting regeneration of the affected organs or tissues. The rationale behind this concept lies on the evidence that targeting key events downstream of the genetic defect can compensate, at least partially, the pathological consequence of the related disease. In this regard, the beneficial effect exerted on animal models of muscular dystrophy by pharmacological strategies that enhance muscle regeneration provides an interesting paradigm. In this review, we describe and discuss the potential targets of pharmacological strategies that promote regeneration of dystrophic muscles and alleviate the consequence of the primary genetic defect. Regenerative pharmacology provides an immediate and suitable therapeutic opportunity to slow down the decline of muscles in the present generation of dystrophic patients, with the perspective to hold them in conditions such that they could benefit of future, more definitive, therapies.
Collapse
Affiliation(s)
- Chiara Mozzetta
- Dulbecco Telethon Institute at Fondazione Santa Lucia/EBRI, Via di Fosso Fiorano, 64-00143 Roma, Italy
| | | | | |
Collapse
|
337
|
|
338
|
Research Highlights. Nat Biotechnol 2008. [DOI: 10.1038/nbt0808-888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
339
|
Research highlights. Nat Genet 2008. [DOI: 10.1038/ng0808-937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|