751
|
Alway SE, Myers MJ, Mohamed JS. Regulation of satellite cell function in sarcopenia. Front Aging Neurosci 2014; 6:246. [PMID: 25295003 PMCID: PMC4170136 DOI: 10.3389/fnagi.2014.00246] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/01/2014] [Indexed: 01/08/2023] Open
Abstract
The mechanisms contributing to sarcopenia include reduced satellite cell (myogenic stem cell) function that is impacted by the environment (niche) of these cells. Satellite cell function is affected by oxidative stress, which is elevated in aged muscles, and this along with changes in largely unknown systemic factors, likely contribute to the manner in which satellite cells respond to stressors such as exercise, disuse, or rehabilitation in sarcopenic muscles. Nutritional intervention provides one therapeutic strategy to improve the satellite cell niche and systemic factors, with the goal of improving satellite cell function in aging muscles. Although many elderly persons consume various nutraceuticals with the hope of improving health, most of these compounds have not been thoroughly tested, and the impacts that they might have on sarcopenia and satellite cell function are not clear. This review discusses data pertaining to the satellite cell responses and function in aging skeletal muscle, and the impact that three compounds: resveratrol, green tea catechins, and β-Hydroxy-β-methylbutyrate have on regulating satellite cell function and therefore contributing to reducing sarcopenia or improving muscle mass after disuse in aging. The data suggest that these nutraceutical compounds improve satellite cell function during rehabilitative loading in animal models of aging after disuse (i.e., muscle regeneration). While these compounds have not been rigorously tested in humans, the data from animal models of aging provide a strong basis for conducting additional focused work to determine if these or other nutraceuticals can offset the muscle losses, or improve regeneration in sarcopenic muscles of older humans via improving satellite cell function.
Collapse
Affiliation(s)
- Stephen E. Alway
- Laboratory of Muscle Biology and Sarcopenia, Department of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- West Virginia Clinical and Translational Science Institute, Morgantown, WV, USA
- Center for Cardiovascular and Respiratory Sciences, Morgantown, WV, USA
| | - Matthew J. Myers
- Laboratory of Muscle Biology and Sarcopenia, Department of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Junaith S. Mohamed
- Laboratory of Muscle Biology and Sarcopenia, Department of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
752
|
Cittadella Vigodarzere G, Mantero S. Skeletal muscle tissue engineering: strategies for volumetric constructs. Front Physiol 2014; 5:362. [PMID: 25295011 PMCID: PMC4170101 DOI: 10.3389/fphys.2014.00362] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 09/03/2014] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle tissue is characterized by high metabolic requirements, defined structure and high regenerative potential. As such, it constitutes an appealing platform for tissue engineering to address volumetric defects, as proven by recent works in this field. Several issues common to all engineered constructs constrain the variety of tissues that can be realized in vitro, principal among them the lack of a vascular system and the absence of reliable cell sources; as it is, the only successful tissue engineering constructs are not characterized by active function, present limited cellular survival at implantation and possess low metabolic requirements. Recently, functionally competent constructs have been engineered, with vascular structures supporting their metabolic requirements. In addition to the use of biochemical cues, physical means, mechanical stimulation and the application of electric tension have proven effective in stimulating the differentiation of cells and the maturation of the constructs; while the use of co-cultures provided fine control of cellular developments through paracrine activity. This review will provide a brief analysis of some of the most promising improvements in the field, with particular attention to the techniques that could prove easily transferable to other branches of tissue engineering.
Collapse
Affiliation(s)
| | - Sara Mantero
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano Milano, Italy
| |
Collapse
|
753
|
Bobadilla M, Sáinz N, Rodriguez JA, Abizanda G, Orbe J, de Martino A, García Verdugo JM, Páramo JA, Prósper F, Pérez-Ruiz A. MMP-10 is required for efficient muscle regeneration in mouse models of injury and muscular dystrophy. Stem Cells 2014; 32:447-61. [PMID: 24123596 DOI: 10.1002/stem.1553] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/20/2013] [Accepted: 08/23/2013] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteinases (MMPs), a family of endopeptidases that are involved in the degradation of extracellular matrix components, have been implicated in skeletal muscle regeneration. Among the MMPs, MMP-2 and MMP-9 are upregulated in Duchenne muscular dystrophy (DMD), a fatal X-linked muscle disorder. However, inhibition or overexpression of specific MMPs in a mouse model of DMD (mdx) has yielded mixed results regarding disease progression, depending on the MMP studied. Here, we have examined the role of MMP-10 in muscle regeneration during injury and muscular dystrophy. We found that skeletal muscle increases MMP-10 protein expression in response to damage (notexin) or disease (mdx mice), suggesting its role in muscle regeneration. In addition, we found that MMP-10-deficient muscles displayed impaired recruitment of endothelial cells, reduced levels of extracellular matrix proteins, diminished collagen deposition, and decreased fiber size, which collectively contributed to delayed muscle regeneration after injury. Also, MMP-10 knockout in mdx mice led to a deteriorated dystrophic phenotype. Moreover, MMP-10 mRNA silencing in injured muscles (wild-type and mdx) reduced muscle regeneration, while addition of recombinant human MMP-10 accelerated muscle repair, suggesting that MMP-10 is required for efficient muscle regeneration. Furthermore, our data suggest that MMP-10-mediated muscle repair is associated with VEGF/Akt signaling. Thus, our findings indicate that MMP-10 is critical for skeletal muscle maintenance and regeneration during injury and disease.
Collapse
Affiliation(s)
- Míriam Bobadilla
- Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
754
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes: multitasking cells in the regeneration of injured, diseased, and aged skeletal muscle. Front Aging Neurosci 2014; 6:245. [PMID: 25278877 PMCID: PMC4166895 DOI: 10.3389/fnagi.2014.00245] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/29/2014] [Indexed: 12/16/2022] Open
Abstract
Pericytes are perivascular cells that envelop and make intimate connections with adjacent capillary endothelial cells. Recent studies show that they may have a profound impact in skeletal muscle regeneration, innervation, vessel formation, fibrosis, fat accumulation, and ectopic bone formation throughout life. In this review, we summarize and evaluate recent advances in our understanding of pericytes' influence on adult skeletal muscle pathophysiology. We also discuss how further elucidating their biology may offer new approaches to the treatment of conditions characterized by muscle wasting.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA ; Neuroscience Program, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Maria L Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine Winston-Salem, NC, USA ; Neuroscience Program, Wake Forest School of Medicine Winston-Salem, NC, USA
| |
Collapse
|
755
|
STAT3 signaling controls satellite cell expansion and skeletal muscle repair. Nat Med 2014; 20:1182-6. [PMID: 25194572 DOI: 10.1038/nm.3656] [Citation(s) in RCA: 295] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/10/2014] [Indexed: 12/24/2022]
Abstract
The progressive loss of muscle regenerative capacity with age or disease results in part from a decline in the number and function of satellite cells, the direct cellular contributors to muscle repair. However, little is known about the molecular effectors underlying satellite cell impairment and depletion. Elevated levels of inflammatory cytokines, including interleukin-6 (IL-6), are associated with both age-related and muscle-wasting conditions. The levels of STAT3, a downstream effector of IL-6, are also elevated with muscle wasting, and STAT3 has been implicated in the regulation of self-renewal and stem cell fate in several tissues. Here we show that IL-6-activated Stat3 signaling regulates satellite cell behavior, promoting myogenic lineage progression through myogenic differentiation 1 (Myod1) regulation. Conditional ablation of Stat3 in Pax7-expressing satellite cells resulted in their increased expansion during regeneration, but compromised myogenic differentiation prevented the contribution of these cells to regenerating myofibers. In contrast, transient Stat3 inhibition promoted satellite cell expansion and enhanced tissue repair in both aged and dystrophic muscle. The effects of STAT3 inhibition on cell fate and proliferation were conserved in human myoblasts. The results of this study indicate that pharmacological manipulation of STAT3 activity can be used to counteract the functional exhaustion of satellite cells in pathological conditions, thereby maintaining the endogenous regenerative response and ameliorating muscle-wasting diseases.
Collapse
|
756
|
Sawano S, Suzuki T, Do MKQ, Ohtsubo H, Mizunoya W, Ikeuchi Y, Tatsumi R. Supplementary immunocytochemistry of hepatocyte growth factor production in activated macrophages early in muscle regeneration. Anim Sci J 2014; 85:994-1000. [DOI: 10.1111/asj.12264] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 06/06/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Shoko Sawano
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Takahiro Suzuki
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Mai-Khoi Q. Do
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Hideaki Ohtsubo
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Wataru Mizunoya
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Yoshihide Ikeuchi
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| |
Collapse
|
757
|
Long C, McAnally JR, Shelton JM, Mireault AA, Bassel-Duby R, Olson EN. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA. Science 2014; 345:1184-1188. [PMID: 25123483 DOI: 10.1126/science.1254445] [Citation(s) in RCA: 519] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an inherited X-linked disease caused by mutations in the gene encoding dystrophin, a protein required for muscle fiber integrity. DMD is characterized by progressive muscle weakness and a shortened life span, and there is no effective treatment. We used clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9)-mediated genome editing to correct the dystrophin gene (Dmd) mutation in the germ line of mdx mice, a model for DMD, and then monitored muscle structure and function. Genome editing produced genetically mosaic animals containing 2 to 100% correction of the Dmd gene. The degree of muscle phenotypic rescue in mosaic mice exceeded the efficiency of gene correction, likely reflecting an advantage of the corrected cells and their contribution to regenerating muscle. With the anticipated technological advances that will facilitate genome editing of postnatal somatic cells, this strategy may one day allow correction of disease-causing mutations in the muscle tissue of patients with DMD.
Collapse
Affiliation(s)
- Chengzu Long
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - John R McAnally
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - John M Shelton
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alex A Mireault
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
758
|
Specific pattern of cell cycle during limb fetal myogenesis. Dev Biol 2014; 392:308-23. [DOI: 10.1016/j.ydbio.2014.05.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/24/2014] [Accepted: 05/21/2014] [Indexed: 01/20/2023]
|
759
|
Murphy MM, Keefe AC, Lawson JA, Flygare SD, Yandell M, Kardon G. Transiently active Wnt/β-catenin signaling is not required but must be silenced for stem cell function during muscle regeneration. Stem Cell Reports 2014; 3:475-88. [PMID: 25241745 PMCID: PMC4266007 DOI: 10.1016/j.stemcr.2014.06.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 06/27/2014] [Accepted: 06/30/2014] [Indexed: 12/22/2022] Open
Abstract
Adult muscle’s exceptional capacity for regeneration is mediated by muscle stem cells, termed satellite cells. As with many stem cells, Wnt/β-catenin signaling has been proposed to be critical in satellite cells during regeneration. Using new genetic reagents, we explicitly test in vivo whether Wnt/β-catenin signaling is necessary and sufficient within satellite cells and their derivatives for regeneration. We find that signaling is transiently active in transit-amplifying myoblasts, but is not required for regeneration or satellite cell self-renewal. Instead, downregulation of transiently activated β-catenin is important to limit the regenerative response, as continuous regeneration is deleterious. Wnt/β-catenin activation in adult satellite cells may simply be a vestige of their developmental lineage, in which β-catenin signaling is critical for fetal myogenesis. In the adult, surprisingly, we show that it is not activation but rather silencing of Wnt/β-catenin signaling that is important for muscle regeneration. Wnt/β-catenin signaling is transiently active in myoblasts during muscle regeneration β-catenin is not required in myogenic cells for muscle regeneration β-catenin signaling in myoblasts must be silenced to limit the regenerative response β-catenin requirement and sensitivity differs in fetal and adult muscle stem cells
Collapse
Affiliation(s)
- Malea M Murphy
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandra C Keefe
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jennifer A Lawson
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Steven D Flygare
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Mark Yandell
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
760
|
Grounds MD. Therapies for sarcopenia and regeneration of old skeletal muscles: more a case of old tissue architecture than old stem cells. BIOARCHITECTURE 2014; 4:81-7. [PMID: 25101758 DOI: 10.4161/bioa.29668] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Age related loss of skeletal muscle mass and function (sarcopenia) reduces independence and the quality of life for individuals, and leads to falls and fractures with escalating health costs for the rapidly aging human population. Thus there is much interest in developing interventions to reduce sarcopenia. One area that has attracted recent attention is the proposed use of myogenic stem cells to improve regeneration of old muscles. This mini-review challenges the fundamental need for myogenic stem cell therapy for sarcopenia. It presents evidence that demonstrates the excellent capacity of myogenic stem cells from very old rodent and human muscles to form new muscles after experimental myofiber necrosis. The many factors required for successful muscle regeneration are considered with a strong focus on integration of components of old muscle bioarchitecture. The fundamental role of satellite cells in homeostasis of normal aging muscles and the incidence of endogenous regeneration in old muscles is questioned. These issues, combined with problems for clinical myogenic stem cell therapies for severe muscle diseases, raise fundamental concerns about the justification for myogenic stem cell therapy for sarcopenia.
Collapse
Affiliation(s)
- Miranda D Grounds
- School of Anatomy, Physiology and Human Biology; University of Western Australia; Crawley, Australia
| |
Collapse
|
761
|
Brien P, Pugazhendhi D, Woodhouse S, Oxley D, Pell JM. p38α MAPK regulates adult muscle stem cell fate by restricting progenitor proliferation during postnatal growth and repair. Stem Cells 2014; 31:1597-610. [PMID: 23592450 DOI: 10.1002/stem.1399] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 03/05/2013] [Accepted: 03/18/2013] [Indexed: 11/05/2022]
Abstract
Stem cell function is essential for the maintenance of adult tissue homeostasis. Controlling the balance between self-renewal and differentiation is crucial to maintain a receptive satellite cell pool capable of responding to growth and regeneration cues. The mitogen-activated protein kinase p38α has been implicated in the regulation of these processes but its influence in adult muscle remains unknown. Using conditional satellite cell p38α knockout mice we have demonstrated that p38α restricts excess proliferation in the postnatal growth phase while promoting timely myoblast differentiation. Differentiation was still able to occur in the p38α-null satellite cells, however, but was delayed. An absence of p38α resulted in a postnatal growth defect along with the persistence of an increased reservoir of satellite cells into adulthood. This population was still capable of responding to cardiotoxin-induced injury, resulting in complete, albeit delayed, regeneration, with further enhancement of the satellite cell population. Increased p38γ phosphorylation accompanied the absence of p38α, and inhibition of p38γ ex vivo substantially decreased the myogenic defect. We have used genome-wide transcriptome analysis to characterize the changes in expression that occur between resting and regenerating muscle, and the influence p38α has on these expression profiles. This study provides novel evidence for the fundamental role of p38α in adult muscle homeostasis in vivo.
Collapse
|
762
|
Hart ML, Neumayer KMH, Vaegler M, Daum L, Amend B, Sievert KD, Di Giovanni S, Kraushaar U, Guenther E, Stenzl A, Aicher WK. Cell-based therapy for the deficient urinary sphincter. Curr Urol Rep 2014; 14:476-87. [PMID: 23824516 DOI: 10.1007/s11934-013-0352-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
When sterile culture techniques of mammalian cells first became state of the art, there was tremendous anticipation that such cells could be eventually applied for therapeutic purposes. The discovery of adult human stem or progenitor cells further motivated scientists to pursue research in cell-based therapies. Although evidence from animal studies suggests that application of cells yields measurable benefits, in urology and many other disciplines, progenitor-cell-based therapies are not yet routinely clinically available. Stress urinary incontinence (SUI) is a condition affecting a large number of patients. The etiology of SUI includes, but is not limited to, degeneration of the urinary sphincter muscle tissue and loss of innervation, as well as anatomical and biomechanical causes. Therefore, different regimens were developed to treat SUI. However, at present, a curative functional treatment is not at hand. A progenitor-cell-based therapy that can tackle the etiology of incontinence, rather than the consequences, is a promising strategy. Therefore, several research teams have intensified their efforts to develop such a therapy for incontinence. Here, we introduce candidate stem and progenitor cells suitable for SUI treatment, show how the functional homogeneity and state of maturity of differentiated cells crucial for proper tissue integration can be assessed electrophysiologically prior to their clinical application, and discuss the trophic potential of adult mesenchymal stromal (or stem) cells in regeneration of neuronal function.
Collapse
Affiliation(s)
- Melanie L Hart
- KFO273, Department of Urology, UKT, University of Tuebingen, Paul-Ehrlich-Str. 15, 72076, Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
763
|
Jiang C, Wen Y, Kuroda K, Hannon K, Rudnicki MA, Kuang S. Notch signaling deficiency underlies age-dependent depletion of satellite cells in muscular dystrophy. Dis Model Mech 2014; 7:997-1004. [PMID: 24906372 PMCID: PMC4107328 DOI: 10.1242/dmm.015917] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating disease characterized by muscle wasting, loss of mobility and death in early adulthood. Satellite cells are muscle-resident stem cells responsible for the repair and regeneration of damaged muscles. One pathological feature of DMD is the progressive depletion of satellite cells, leading to the failure of muscle repair. Here, we attempted to explore the molecular mechanisms underlying satellite cell ablation in the dystrophin mutant mdx mouse, a well-established model for DMD. Initial muscle degeneration activates satellite cells, resulting in increased satellite cell number in young mdx mice. This is followed by rapid loss of satellite cells with age due to the reduced self-renewal ability of mdx satellite cells. In addition, satellite cell composition is altered even in young mdx mice, with significant reductions in the abundance of non-committed (Pax7+ and Myf5−) satellite cells. Using a Notch-reporter mouse, we found that the mdx satellite cells have reduced activation of Notch signaling, which has been shown to be necessary to maintain satellite cell quiescence and self-renewal. Concomitantly, the expression of Notch1, Notch3, Jag1, Hey1 and HeyL are reduced in the mdx primary myoblast. Finally, we established a mouse model to constitutively activate Notch signaling in satellite cells, and show that Notch activation is sufficient to rescue the self-renewal deficiencies of mdx satellite cells. These results demonstrate that Notch signaling is essential for maintaining the satellite cell pool and that its deficiency leads to depletion of satellite cells in DMD.
Collapse
Affiliation(s)
- Chunhui Jiang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Yefei Wen
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Kazuki Kuroda
- Molecular Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada
| | - Kevin Hannon
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Michael A Rudnicki
- Molecular Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
764
|
Lund DK, Mouly V, Cornelison DDW. MMP-14 is necessary but not sufficient for invasion of three-dimensional collagen by human muscle satellite cells. Am J Physiol Cell Physiol 2014; 307:C140-9. [PMID: 24898588 DOI: 10.1152/ajpcell.00032.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The twenty-five known matrix metalloproteases (MMPs) and their endogenous inhibitors, tissue inhibitors of metalloproteases (TIMPs), mediate cell invasion through the extracellular matrix (ECM). In a comparative three-dimensional assay, we analyzed human and mouse satellite cells' competence to invade an artificial ECM (collagen I). We identified a single MMP that 1) is expressed by human muscle satellite cells; 2) is induced at the mRNA/protein level by adhesion to collagen I; and 3) is necessary for invasion into a collagen I matrix. Interestingly, murine satellite cells neither express this MMP, nor invade the collagen matrix. However, exogenous human MMP-14 is not sufficient to induce invasion of a collagen matrix by murine cells, emphasizing species differences.
Collapse
Affiliation(s)
- Dane K Lund
- Division of Biology and Bond Life Sciences Center, University of Missouri, Columbia, Missouri; and
| | - Vincent Mouly
- Institut de Myologie, Université Pierre et Marie Curie, Paris, France
| | - D D W Cornelison
- Division of Biology and Bond Life Sciences Center, University of Missouri, Columbia, Missouri; and
| |
Collapse
|
765
|
Southard S, Low S, Li L, Rozo M, Harvey T, Fan CM, Lepper C. A series of Cre-ER(T2) drivers for manipulation of the skeletal muscle lineage. Genesis 2014; 52:759-70. [PMID: 24844572 DOI: 10.1002/dvg.22792] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 05/14/2014] [Accepted: 05/16/2014] [Indexed: 12/20/2022]
Abstract
We report the generation of five mouse strains with the tamoxifen-inducible Cre (Cre-ER(T) (2) ; CE) gene cassette knocked into the endogenous loci of Pax3, Myod1, Myog, Myf6, and Myl1, collectively as a resource for the skeletal muscle research community. We characterized these CE strains using the Cre reporter mice, R26R(L) (acZ) , during embryogenesis and show that they direct tightly controlled tamoxifen-inducible reporter expression within the expected cell lineage determined by each myogenic gene. We also examined a few selected adult skeletal muscle groups for tamoxifen-inducible reporter expression. None of these new CE alleles direct reporter expression in the cardiac muscle. All these alleles follow the same knock-in strategy by replacing the first exon of each gene with the CE cassette, rendering them null alleles of the endogenous gene. Advantages and disadvantages of this design are discussed. Although we describe potential immediate use of these strains, their utility likely extends beyond foreseeable questions in skeletal muscle biology.
Collapse
Affiliation(s)
- Sheryl Southard
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland
| | | | | | | | | | | | | |
Collapse
|
766
|
Narasimhan M, Hong J, Atieno N, Muthusamy VR, Davidson CJ, Abu-Rmaileh N, Richardson RS, Gomes AV, Hoidal JR, Rajasekaran NS. Nrf2 deficiency promotes apoptosis and impairs PAX7/MyoD expression in aging skeletal muscle cells. Free Radic Biol Med 2014; 71:402-414. [PMID: 24613379 PMCID: PMC4493911 DOI: 10.1016/j.freeradbiomed.2014.02.023] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 02/17/2014] [Accepted: 02/25/2014] [Indexed: 12/20/2022]
Abstract
Skeletal muscle redox homeostasis is transcriptionally regulated by nuclear erythroid-2-p45-related factor-2 (Nrf2). We recently demonstrated that age-associated stress impairs Nrf2-ARE (antioxidant-response element) transcriptional signaling. Here, we hypothesize that age-dependent decline or genetic ablation of Nrf2 leads to accelerated apoptosis and skeletal muscle degeneration. Under basal-physiological conditions, disruption of Nrf2 significantly downregulates antioxidants and causes oxidative stress. Surprisingly, Nrf2-null mice had enhanced antioxidant capacity identical to wild-type (WT) upon acute endurance exercise stress (AEES), suggesting activation of Nrf2-independent mechanisms (i.e., PGC1α) against oxidative stress. Analysis of prosurvival pathways in the basal state reveals decreased AKT levels, whereas p-p53, a repressor of AKT, was increased in Nrf2-null vs WT mice. Upon AEES, AKT and p-AKT levels were significantly (p < 0.001) increased (>10-fold) along with profound downregulation of p-p53 (p < 0.01) in Nrf2-null vs WT skeletal muscle, indicating the onset of prosurvival mechanisms to compensate for the loss of Nrf2 signaling. However, we found a decreased stem cell population (PAX7) and MyoD expression (differentiation) along with profound activation of ubiquitin and apoptotic pathways in Nrf2-null vs WT mice upon AEES, suggesting that compensatory prosurvival mechanisms failed to overcome the programmed cell death and degeneration in skeletal muscle. Further, the impaired regeneration was sustained in Nrf2-null vs WT mice after 1 week of post-AEES recovery. In an age-associated oxidative stress condition, ablation of Nrf2 results in induction of apoptosis and impaired muscle regeneration.
Collapse
Affiliation(s)
- Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Jennifer Hong
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Nancy Atieno
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Vasanthi R Muthusamy
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Christopher J Davidson
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Naser Abu-Rmaileh
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Russell S Richardson
- Division of Geriatrics, and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; Department of Exercise & Sports Sciences, College of Health, University of Utah, Salt Lake City, UT 84112, USA; Geriatric Research, Education, and Clinical Center, Salt Lake City Veteran's Medical Center
| | | | - John R Hoidal
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Namakkal S Rajasekaran
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; Department of Exercise & Sports Sciences, College of Health, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
767
|
Chakkalakal JV, Christensen J, Xiang W, Tierney MT, Boscolo FS, Sacco A, Brack AS. Early forming label-retaining muscle stem cells require p27kip1 for maintenance of the primitive state. Development 2014; 141:1649-59. [PMID: 24715455 PMCID: PMC3978835 DOI: 10.1242/dev.100842] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Across different niches, subsets of highly functional stem cells are maintained in a relatively dormant rather than proliferative state. Our understanding of proliferative dynamics in tissue-specific stem cells during conditions of increased tissue turnover remains limited. Using a TetO-H2B-GFP reporter of proliferative history, we identify skeletal muscle stem cell, or satellite cells, that retain (LRC) or lose (nonLRC) the H2B-GFP label. We show in mice that LRCs and nonLRCs are formed at birth and persist during postnatal growth and adult muscle repair. Functionally, LRCs and nonLRCs are born equivalent and transition during postnatal maturation into distinct and hierarchically organized subsets. Adult LRCs give rise to LRCs and nonLRCs; the former are able to self-renew, whereas the latter are restricted to differentiation. Expression analysis revealed the CIP/KIP family members p21(cip1) (Cdkn1a) and p27(kip1) (Cdkn1b) to be expressed at higher levels in LRCs. In accordance with a crucial role in LRC fate, loss of p27(kip1) promoted proliferation and differentiation of LRCs in vitro and impaired satellite cell self-renewal after muscle injury. By contrast, loss of p21(cip1) only affected nonLRCs, in which myogenic commitment was inhibited. Our results provide evidence that restriction of self-renewal potential to LRCs is established early in life and is maintained during increased tissue turnover through the cell cycle inhibitor p27(kip1). They also reveal the differential role of CIP/KIP family members at discrete steps within the stem cell hierarchy.
Collapse
Affiliation(s)
- Joe V Chakkalakal
- Massachusetts General Hospital, Center of Regenerative Medicine, Harvard University, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
768
|
Pichavant C, Pavlath GK. Incidence and severity of myofiber branching with regeneration and aging. Skelet Muscle 2014; 4:9. [PMID: 24855558 PMCID: PMC4030050 DOI: 10.1186/2044-5040-4-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 05/01/2014] [Indexed: 11/30/2022] Open
Abstract
Background Myofibers with an abnormal branching cytoarchitecture are commonly found in muscular dystrophy and in regenerated or aged nondystrophic muscles. Such branched myofibers from dystrophic mice are more susceptible to damage than unbranched myofibers in vitro, suggesting that muscles containing a high percentage of these myofibers are more prone to injury. Little is known about the regulation of myofiber branching. Methods To gain insights into the formation and fate of branched myofibers, we performed in-depth analyses of single myofibers isolated from dystrophic and nondystrophic (myotoxin-injured or aged) mouse muscles. The proportion of branched myofibers, the number of branches per myofiber and the morphology of the branches were assessed. Results Aged dystrophic mice exhibited the most severe myofiber branching as defined by the incidence of branched myofibers and the number of branches per myofiber, followed by myotoxin-injured, wild-type muscles and then aged wild-type muscles. In addition, the morphology of the branched myofibers differed among the various models. In response to either induced or ongoing muscle degeneration, branching was restricted to regenerated myofibers containing central nuclei. In myotoxin-injured muscles, the amount of branched myofibers remained stable over time. Conclusion We suggest that myofiber branching is a consequence of myofiber remodeling during muscle regeneration. Our present study lays valuable groundwork for identifying the molecular pathways leading to myofiber branching in dystrophy, trauma and aging. Decreasing myofiber branching in dystrophic patients may improve muscle resistance to mechanical stress.
Collapse
Affiliation(s)
- Christophe Pichavant
- Department of Pharmacology, Rollins Research Center, Emory University, 1510 Clifton Road, Atlanta, GA 30322, USA
| | - Grace K Pavlath
- Department of Pharmacology, Rollins Research Center, Emory University, 1510 Clifton Road, Atlanta, GA 30322, USA
| |
Collapse
|
769
|
Kharraz Y, Guerra J, Pessina P, Serrano AL, Muñoz-Cánoves P. Understanding the process of fibrosis in Duchenne muscular dystrophy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:965631. [PMID: 24877152 PMCID: PMC4024417 DOI: 10.1155/2014/965631] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/08/2014] [Indexed: 02/06/2023]
Abstract
Fibrosis is the aberrant deposition of extracellular matrix (ECM) components during tissue healing leading to loss of its architecture and function. Fibrotic diseases are often associated with chronic pathologies and occur in a large variety of vital organs and tissues, including skeletal muscle. In human muscle, fibrosis is most readily associated with the severe muscle wasting disorder Duchenne muscular dystrophy (DMD), caused by loss of dystrophin gene function. In DMD, skeletal muscle degenerates and is infiltrated by inflammatory cells and the functions of the muscle stem cells (satellite cells) become impeded and fibrogenic cells hyperproliferate and are overactivated, leading to the substitution of skeletal muscle with nonfunctional fibrotic tissue. Here, we review new developments in our understanding of the mechanisms leading to fibrosis in DMD and several recent advances towards reverting it, as potential treatments to attenuate disease progression.
Collapse
Affiliation(s)
- Yacine Kharraz
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Joana Guerra
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Patrizia Pessina
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Antonio L. Serrano
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| |
Collapse
|
770
|
Uezumi A, Fukada S, Yamamoto N, Ikemoto-Uezumi M, Nakatani M, Morita M, Yamaguchi A, Yamada H, Nishino I, Hamada Y, Tsuchida K. Identification and characterization of PDGFRα+ mesenchymal progenitors in human skeletal muscle. Cell Death Dis 2014; 5:e1186. [PMID: 24743741 PMCID: PMC4001314 DOI: 10.1038/cddis.2014.161] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 02/07/2023]
Abstract
Fatty and fibrous connective tissue formation is a hallmark of diseased skeletal muscle and deteriorates muscle function. We previously identified non-myogenic mesenchymal progenitors that contribute to adipogenesis and fibrogenesis in mouse skeletal muscle. In this study, we report the identification and characterization of a human counterpart to these progenitors. By using PDGFRα as a specific marker, mesenchymal progenitors can be identified in the interstitium and isolated from human skeletal muscle. PDGFRα+ cells represent a cell population distinct from CD56+ myogenic cells, and adipogenic and fibrogenic potentials were highly enriched in the PDGFRα+ population. Activation of PDGFRα stimulates proliferation of PDGFRα+ cells through PI3K-Akt and MEK2-MAPK signaling pathways, and aberrant accumulation of PDGFRα+ cells was conspicuous in muscles of patients with both genetic and non-genetic muscle diseases. Our results revealed the pathological relevance of PDGFRα+ mesenchymal progenitors to human muscle diseases and provide a basis for developing therapeutic strategy to treat muscle diseases.
Collapse
Affiliation(s)
- A Uezumi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi 470-1192, Japan
| | - S Fukada
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - N Yamamoto
- Laboratory of Molecular Biology and Histochemistry, Fujita Health University, Aichi, Japan
| | - M Ikemoto-Uezumi
- Department of Regenerative Medicine, National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, 35 Gengo, Morioka, Obu, Aichi 474-8511, Japan
| | - M Nakatani
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi 470-1192, Japan
| | - M Morita
- Department of Orthopaedic Surgery, Fujita Health University, Aichi, Japan
| | - A Yamaguchi
- Department of Orthopaedic Surgery, Fujita Health University, Aichi, Japan
| | - H Yamada
- Department of Orthopaedic Surgery, Fujita Health University, Aichi, Japan
| | - I Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan
| | - Y Hamada
- Department of Orthopedics, Tokushima Prefectural Central Hospital, 1-10-3 Kuramoto, Tokushima 770-8539, Japan
| | - K Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
771
|
Fry CS, Noehren B, Mula J, Ubele MF, Westgate PM, Kern PA, Peterson CA. Fibre type-specific satellite cell response to aerobic training in sedentary adults. J Physiol 2014; 592:2625-35. [PMID: 24687582 DOI: 10.1113/jphysiol.2014.271288] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In the present study, we sought to determine the effect of a traditional, 12 week aerobic training protocol on skeletal muscle fibre type distribution and satellite cell content in sedentary subjects. Muscle biopsies were obtained from the vastus lateralis [n = 23 subjects (six male and 17 female); body mass index 30.7 ± 1.2 kg m(-2)] before and after 12 weeks of aerobic training performed on a cycle ergometer. Immunohistochemical analyses were used to quantify myosin heavy chain (MyHC) isoform expression, cross-sectional area and satellite cell and myonuclear content. Following training, a decrease in MyHC hybrid type IIa/IIx fibre frequency occurred, with a concomitant increase in pure MyHC type IIa fibres. Pretraining fibre type correlated with body mass index, and the change in fibre type following training was associated with improvements in maximal oxygen consumption. Twelve weeks of aerobic training also induced increases in mean cross-sectional area in both MyHC type I and type IIa fibres. Satellite cell content was also increased following training, specifically in MyHC type I fibres, with no change in the number of satellite cells associated with MyHC type II fibres. With the increased satellite cell content following training, an increase in myonuclear number per fibre also occurred in MyHC type I fibres. Hypertrophy of MyHC type II fibres occurred without detectable myonuclear addition, suggesting that the mechanisms underlying growth in fast and slow fibres differ. These data provide intriguing evidence for a fibre type-specific role of satellite cells in muscle adaptation following aerobic training.
Collapse
Affiliation(s)
- Christopher S Fry
- Department of Rehabilitation Sciences, College of Health Sciences and Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Brian Noehren
- Department of Rehabilitation Sciences, College of Health Sciences and Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Jyothi Mula
- Department of Rehabilitation Sciences, College of Health Sciences and Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Margo F Ubele
- Department of Rehabilitation Sciences, College of Health Sciences and Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Philip M Westgate
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Philip A Kern
- Department of Internal Medicine/Division of Endocrinology, College of Medicine and the Barnstable Brown Diabetes and Obesity Center, University of Kentucky, Lexington, KY, USA
| | - Charlotte A Peterson
- Department of Rehabilitation Sciences, College of Health Sciences and Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
772
|
Rossi G, Messina G. Comparative myogenesis in teleosts and mammals. Cell Mol Life Sci 2014; 71:3081-99. [PMID: 24664432 PMCID: PMC4111864 DOI: 10.1007/s00018-014-1604-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/17/2014] [Accepted: 03/06/2014] [Indexed: 01/02/2023]
Abstract
Skeletal myogenesis has been and is currently under extensive study in both mammals and teleosts, with the latter providing a good model for skeletal myogenesis because of their flexible and conserved genome. Parallel investigations of muscle studies using both these models have strongly accelerated the advances in the field. However, when transferring the knowledge from one model to the other, it is important to take into account both their similarities and differences. The main difficulties in comparing mammals and teleosts arise from their different temporal development. Conserved aspects can be seen for muscle developmental origin and segmentation, and for the presence of multiple myogenic waves. Among the divergences, many fish have an indeterminate growth capacity throughout their entire life span, which is absent in mammals, thus implying different post-natal growth mechanisms. This review covers the current state of the art on myogenesis, with a focus on the most conserved and divergent aspects between mammals and teleosts.
Collapse
Affiliation(s)
- Giuliana Rossi
- Department of Biosciences, University of Milan, 20133, Milan, Italy
| | | |
Collapse
|
773
|
Cellular players in skeletal muscle regeneration. BIOMED RESEARCH INTERNATIONAL 2014; 2014:957014. [PMID: 24779022 PMCID: PMC3980925 DOI: 10.1155/2014/957014] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 01/12/2014] [Accepted: 01/28/2014] [Indexed: 12/12/2022]
Abstract
Skeletal muscle, a tissue endowed with remarkable endogenous regeneration potential, is still under focused experimental investigation mainly due to treatment potential for muscle trauma and muscular dystrophies. Resident satellite cells with stem cell features were enthusiastically described quite a long time ago, but activation of these cells is not yet controlled by any medical interventions. However, after thorough reports of their existence, survival, activation, and differentiation there are still many questions to be answered regarding the intimate mechanism of tissue regeneration. This review delivers an up-to-date inventory of the main known key players in skeletal muscle repair, revealed by various models of tissue injuries in mechanical trauma, toxic lesions, and muscular dystrophy. A better understanding of the spatial and temporal relationships between various cell populations, with different physical or paracrine interactions and phenotype changes induced by local or systemic signalling, might lead to a more efficient approach for future therapies.
Collapse
|
774
|
Otis JS, Niccoli S, Hawdon N, Sarvas JL, Frye MA, Chicco AJ, Lees SJ. Pro-inflammatory mediation of myoblast proliferation. PLoS One 2014; 9:e92363. [PMID: 24647690 PMCID: PMC3960233 DOI: 10.1371/journal.pone.0092363] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 02/20/2014] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle satellite cell function is largely dictated by the surrounding environment following injury. Immune cell infiltration dominates the extracellular space in the injured area, resulting in increased cytokine concentrations. While increased pro-inflammatory cytokine expression has been previously established in the first 3 days following injury, less is known about the time course of cytokine expression and the specific mechanisms of cytokine induced myoblast function. Therefore, the expression of IL-1β and IL-6 at several time points following injury, and their effects on myoblast proliferation, were examined. In order to do this, skeletal muscle was injured using barium chloride in mice and tissue was collected 1, 5, 10, and 28 days following injury. Mechanisms of cytokine induced proliferation were determined in cell culture using both primary and C2C12 myoblasts. It was found that there is a ∼20-fold increase in IL-1β (p≤0.05) and IL-6 (p = 0.06) expression 5 days following injury. IL-1β increased proliferation of both primary and C2C12 cells ∼25%. IL-1β stimulation also resulted in increased NF-κB activity, likely contributing to the increased proliferation. These data demonstrate for the first time that IL-1β alone can increase the mitogenic activity of primary skeletal muscle satellite cells and offer insight into the mechanisms dictating satellite cell function following injury.
Collapse
Affiliation(s)
- Jeffrey S. Otis
- Department of Kinesiology and Health, Georgia State University, Atlanta, Georgia, United States of America
| | - Sarah Niccoli
- Medical Sciences Division, Northern Ontario School of Medicine, Thunder Bay, Ontario, Canada
| | - Nicole Hawdon
- Medical Sciences Division, Northern Ontario School of Medicine, Thunder Bay, Ontario, Canada
| | - Jessica L. Sarvas
- Medical Sciences Division, Northern Ontario School of Medicine, Thunder Bay, Ontario, Canada
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada
| | - Melinda A. Frye
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Adam J. Chicco
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States of America
| | - Simon J. Lees
- Medical Sciences Division, Northern Ontario School of Medicine, Thunder Bay, Ontario, Canada
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada
- Department of Health and Exercise Science, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
775
|
Bernet JD, Doles JD, Hall JK, Kelly-Tanaka K, Carter TA, Olwin BB. p38 MAPK signaling underlies a cell-autonomous loss of stem cell self-renewal in skeletal muscle of aged mice. Nat Med 2014; 20:265-71. [PMID: 24531379 PMCID: PMC4070883 DOI: 10.1038/nm.3465] [Citation(s) in RCA: 423] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/10/2014] [Indexed: 12/13/2022]
Abstract
Skeletal muscle aging results in a gradual loss of skeletal muscle mass, skeletal muscle function and regenerative capacity, which can lead to sarcopenia and increased mortality. Although the mechanisms underlying sarcopenia remain unclear, the skeletal muscle stem cell, or satellite cell, is required for muscle regeneration. Therefore, identification of signaling pathways affecting satellite cell function during aging may provide insights into therapeutic targets for combating sarcopenia. Here, we show that a cell-autonomous loss in self-renewal occurs via alterations in fibroblast growth factor receptor-1, p38α and p38β mitogen-activated protein kinase signaling in satellite cells from aged mice. We further demonstrate that pharmacological manipulation of these pathways can ameliorate age-associated self-renewal defects. Thus, our data highlight an age-associated deregulation of a satellite cell homeostatic network and reveal potential therapeutic opportunities for the treatment of progressive muscle wasting.
Collapse
Affiliation(s)
- Jennifer D. Bernet
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309
| | - Jason D. Doles
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309
| | - John K. Hall
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309
| | - Kathleen Kelly-Tanaka
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309
| | - Thomas A. Carter
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309
| | - Bradley B. Olwin
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309
| |
Collapse
|
776
|
Piccioni A, Gaetani E, Palladino M, Gatto I, Smith RC, Neri V, Marcantoni M, Giarretta I, Silver M, Straino S, Capogrossi M, Landolfi R, Pola R. Sonic hedgehog gene therapy increases the ability of the dystrophic skeletal muscle to regenerate after injury. Gene Ther 2014; 21:413-21. [PMID: 24572787 DOI: 10.1038/gt.2014.13] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 01/04/2014] [Accepted: 01/15/2014] [Indexed: 11/09/2022]
Abstract
The Hedgehog (Hh) pathway is a crucial regulator of muscle development during embryogenesis. We have previously demonstrated that Sonic hedgehog (Shh) regulates postnatal myogenesis in the adult skeletal muscle both directly, by acting on muscle satellite cells, and indirectly, by promoting the production of growth factors from interstitial fibroblasts. Here, we show that in mdx mice, the murine equivalent of Duchenne muscular dystrophy in humans, progression of the dystrophic pathology corresponds to progressive inhibition of the Hh signaling pathway in the skeletal muscle. We also show that the upregulation of the Hh pathway in response to injury and during regeneration is significantly impaired in mdx muscle. Shh treatment increases the proliferative potential of satellite cells isolated from the muscles of mdx mice. This treatment also increases the production of proregenerative factors, such as insulin-like growth factor-1 and vascular endothelial growth factor, from fibroblasts isolated from the muscle of mdx mice. In vivo, overexpression of the Hh pathway using a plasmid encoding the human Shh gene promotes successful regeneration after injury in terms of increased number of proliferating myogenic cells and newly formed myofibers, as well as enhanced vascularization and decreased fibrosis.
Collapse
Affiliation(s)
- A Piccioni
- 1] Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy [2] Division of Cardiovascular Research, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - E Gaetani
- Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy
| | - M Palladino
- 1] Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy [2] Division of Cardiovascular Research, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - I Gatto
- Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy
| | - R C Smith
- Division of Cardiovascular Research, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - V Neri
- 1] Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy [2] Division of Cardiovascular Research, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - M Marcantoni
- Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy
| | - I Giarretta
- Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy
| | - M Silver
- Division of Cardiovascular Research, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - S Straino
- 1] Laboratory of Vascular Pathology, IDI IRCCS Research Institute, Rome, Italy [2] Explora Biotech, srl, Rome, Italy
| | - M Capogrossi
- Laboratory of Vascular Pathology, IDI IRCCS Research Institute, Rome, Italy
| | - R Landolfi
- Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy
| | - R Pola
- 1] Department of Medicine, A. Gemelli University Hospital, Catholic University School of Medicine, Rome, Italy [2] Division of Cardiovascular Research, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
777
|
Uezumi A, Ikemoto-Uezumi M, Tsuchida K. Roles of nonmyogenic mesenchymal progenitors in pathogenesis and regeneration of skeletal muscle. Front Physiol 2014; 5:68. [PMID: 24605102 PMCID: PMC3932482 DOI: 10.3389/fphys.2014.00068] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 02/04/2014] [Indexed: 12/25/2022] Open
Abstract
Adult skeletal muscle possesses a remarkable regenerative ability that is dependent on satellite cells. However, skeletal muscle is replaced by fatty and fibrous connective tissue in several pathological conditions. Fatty and fibrous connective tissue becomes a major cause of muscle weakness and leads to further impairment of muscle function. Because the occurrence of fatty and fibrous connective tissue is usually associated with severe destruction of muscle, the idea that dysregulation of the fate switch in satellite cells may underlie this pathological change has emerged. However, recent studies identified nonmyogenic mesenchymal progenitors in skeletal muscle and revealed that fatty and fibrous connective tissue originates from these progenitors. Later, these progenitors were also demonstrated to be the major contributor to heterotopic ossification in skeletal muscle. Because nonmyogenic mesenchymal progenitors represent a distinct cell population from satellite cells, targeting these progenitors could be an ideal therapeutic strategy that specifically prevents pathological changes of skeletal muscle, while preserving satellite cell-dependent regeneration. In addition to their roles in pathogenesis of skeletal muscle, nonmyogenic mesenchymal progenitors may play a vital role in muscle regeneration by regulating satellite cell behavior. Conversely, muscle cells appear to regulate behavior of nonmyogenic mesenchymal progenitors. Thus, these cells regulate each other reciprocally and a proper balance between them is a key determinant of muscle integrity. Furthermore, nonmyogenic mesenchymal progenitors have been shown to maintain muscle mass in a steady homeostatic condition. Understanding the nature of nonmyogenic mesenchymal progenitors will provide valuable insight into the pathophysiology of skeletal muscle. In this review, we focus on nonmyogenic mesenchymal progenitors and discuss their roles in muscle pathogenesis, regeneration, and homeostasis.
Collapse
Affiliation(s)
- Akiyoshi Uezumi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University Aichi, Japan
| | - Madoka Ikemoto-Uezumi
- Department of Regenerative Medicine, National Center for Geriatrics and Gerontology, National Institute for Longevity Sciences Aichi, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University Aichi, Japan
| |
Collapse
|
778
|
Motohashi N, Asakura A. Muscle satellite cell heterogeneity and self-renewal. Front Cell Dev Biol 2014; 2:1. [PMID: 25364710 PMCID: PMC4206996 DOI: 10.3389/fcell.2014.00001] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/14/2014] [Indexed: 01/17/2023] Open
Abstract
Adult skeletal muscle possesses extraordinary regeneration capacities. After muscle injury or exercise, large numbers of newly formed muscle fibers are generated within a week as a result of expansion and differentiation of a self-renewing pool of muscle stem cells termed muscle satellite cells. Normally, satellite cells are mitotically quiescent and reside beneath the basal lamina of muscle fibers. Upon regeneration, satellite cells are activated, and give rise to daughter myogenic precursor cells. After several rounds of proliferation, these myogenic precursor cells contribute to the formation of new muscle fibers. During cell division, a minor population of myogenic precursor cells returns to quiescent satellite cells as a self-renewal process. Currently, accumulating evidence has revealed the essential roles of satellite cells in muscle regeneration and the regulatory mechanisms, while it still remains to be elucidated how satellite cell self-renewal is molecularly regulated and how satellite cells are important in aging and diseased muscle. The number of satellite cells is decreased due to the changing niche during ageing, resulting in attenuation of muscle regeneration capacity. Additionally, in Duchenne muscular dystrophy (DMD) patients, the loss of satellite cell regenerative capacity and decreased satellite cell number due to continuous needs for satellite cells lead to progressive muscle weakness with chronic degeneration. Thus, it is necessary to replenish muscle satellite cells continuously. This review outlines recent findings regarding satellite cell heterogeneity, asymmetric division and molecular mechanisms in satellite cell self-renewal which is crucial for maintenance of satellite cells as a muscle stem cell pool throughout life. In addition, we discuss roles in the stem cell niche for satellite cell maintenance, as well as related cell therapies for approaching treatment of DMD.
Collapse
Affiliation(s)
- Norio Motohashi
- Department of Neurology, Paul and Sheila Wellstone Muscular Dystrophy Center, Stem Cell Institute, University of Minnesota Medical School Minneapolis, MN, USA
| | - Atsushi Asakura
- Department of Neurology, Paul and Sheila Wellstone Muscular Dystrophy Center, Stem Cell Institute, University of Minnesota Medical School Minneapolis, MN, USA
| |
Collapse
|
779
|
Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dynamic microenvironment for stem cell niche. Biochim Biophys Acta Gen Subj 2014; 1840:2506-19. [PMID: 24418517 PMCID: PMC4081568 DOI: 10.1016/j.bbagen.2014.01.010] [Citation(s) in RCA: 901] [Impact Index Per Article: 81.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/05/2014] [Accepted: 01/06/2014] [Indexed: 02/08/2023]
Abstract
Background Extracellular matrix (ECM) is a dynamic and complex environment characterized by biophysical, mechanical and biochemical properties specific for each tissue and able to regulate cell behavior. Stem cells have a key role in the maintenance and regeneration of tissues and they are located in a specific microenvironment, defined as niche. Scope of review We overview the progresses that have been made in elucidating stem cell niches and discuss the mechanisms by which ECM affects stem cell behavior. We also summarize the current tools and experimental models for studying ECM–stem cell interactions. Major conclusions ECM represents an essential player in stem cell niche, since it can directly or indirectly modulate the maintenance, proliferation, self-renewal and differentiation of stem cells. Several ECM molecules play regulatory functions for different types of stem cells, and based on its molecular composition the ECM can be deposited and finely tuned for providing the most appropriate niche for stem cells in the various tissues. Engineered biomaterials able to mimic the in vivo characteristics of stem cell niche provide suitable in vitro tools for dissecting the different roles exerted by the ECM and its molecular components on stem cell behavior. General significance ECM is a key component of stem cell niches and is involved in various aspects of stem cell behavior, thus having a major impact on tissue homeostasis and regeneration under physiological and pathological conditions. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties. Stem cells have a key role in the maintenance and regeneration of tissues. The extracellular matrix is a critical regulator of stem cell function. Stem cells reside in a dynamic and specialized microenvironment denoted as niche. The extracellular matrix represents an essential component of stem cell niches. Bioengineered niches can be used for investigating stem cell–matrix interactions.
Collapse
Affiliation(s)
- Francesca Gattazzo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Anna Urciuolo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy.
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
780
|
Moyle LA, Zammit PS. Isolation, culture and immunostaining of skeletal muscle fibres to study myogenic progression in satellite cells. Methods Mol Biol 2014; 1210:63-78. [PMID: 25173161 DOI: 10.1007/978-1-4939-1435-7_6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Satellite cells are the resident stem cells of skeletal muscle, located on the surface of a myofibre, beneath the surrounding basal lamina. Satellite cells are responsible for the homeostasis, hypertrophy and repair of skeletal muscle fibres, being activated to enter proliferation and generate myoblasts that either fuse to existing myofibres, or fuse together for de novo myofibre formation. Isolating muscle fibres allows the associated satellite cells to be obtained while remaining in their anatomical niche beneath the basal lamina, free of interstitial and vascular tissue. Myofibres can then be immunostained to examine gene expression in quiescent satellite cells, or cultured to activate satellite cells before immunostaining to investigate gene expression dynamics during myogenic progression and self-renewal. Here, we describe methods for the isolation, culture and immunostaining of muscle fibres for examining satellite cell biology.
Collapse
Affiliation(s)
- Louise A Moyle
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, King's College London, Guy's Campus, London, SE1 1UL, UK
| | | |
Collapse
|
781
|
|
782
|
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
|
783
|
Fry CS, Lee JD, Jackson JR, Kirby TJ, Stasko SA, Liu H, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. Regulation of the muscle fiber microenvironment by activated satellite cells during hypertrophy. FASEB J 2013; 28:1654-65. [PMID: 24376025 DOI: 10.1096/fj.13-239426] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Our aim in the current study was to determine the necessity of satellite cells for long-term muscle growth and maintenance. We utilized a transgenic Pax7-DTA mouse model, allowing for the conditional depletion of > 90% of satellite cells with tamoxifen treatment. Synergist ablation surgery, where removal of synergist muscles places functional overload on the plantaris, was used to stimulate robust hypertrophy. Following 8 wk of overload, satellite cell-depleted muscle demonstrated an accumulation of extracellular matrix (ECM) and fibroblast expansion that resulted in reduced specific force of the plantaris. Although the early growth response was normal, an attenuation of hypertrophy measured by both muscle wet weight and fiber cross-sectional area occurred in satellite cell-depleted muscle. Isolated primary myogenic progenitor cells (MPCs) negatively regulated fibroblast ECM mRNA expression in vitro, suggesting a novel role for activated satellite cells/MPCs in muscle adaptation. These results provide evidence that satellite cells regulate the muscle environment during growth.
Collapse
Affiliation(s)
- Christopher S Fry
- 1900 S. Limestone, CTW 105, University of Kentucky, Lexington, KY 40536, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
784
|
Hung C, Linn G, Chow YH, Kobayashi A, Mittelsteadt K, Altemeier WA, Gharib SA, Schnapp LM, Duffield JS. Role of lung pericytes and resident fibroblasts in the pathogenesis of pulmonary fibrosis. Am J Respir Crit Care Med 2013; 188:820-30. [PMID: 23924232 DOI: 10.1164/rccm.201212-2297oc] [Citation(s) in RCA: 278] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
RATIONALE The origin of cells that make pathologic fibrillar collagen matrix in lung disease has been controversial. Recent studies suggest mesenchymal cells may contribute directly to fibrosis. OBJECTIVES To characterize discrete populations of mesenchymal cells in the normal mouse lung and to map their fate after bleomycin-induced lung injury. METHODS We mapped the fate of Foxd1-expressing embryonic progenitors and their progeny during lung development, adult homeostasis, and after fibrosing injury in Foxd1-Cre; Rs26-tdTomato-R mice. We studied collagen-I(α)1-producing cells in normal and diseased lungs using Coll-GFP(Tg) mice. MEASUREMENTS AND MAIN RESULTS Foxd1-expressing embryonic progenitors enter lung buds before 13.5 days post-conception, expand, and form an extensive lineage of mesenchymal cells that have characteristics of pericytes. A collagen-I(α)1-expressing mesenchymal population of distinct lineage is also found in adult lung, with features of a resident fibroblast. In contrast to resident fibroblasts, Foxd1 progenitor-derived pericytes are enriched in transcripts for innate immunity, vascular development, WNT signaling pathway, and cell migration. Foxd1 progenitor-derived pericytes expand after bleomycin lung injury, and activate expression of collagen-I(α)1 and the myofibroblast marker αSMA in fibrotic foci. In addition, our studies suggest a distinct lineage of collagen-I(α)1-expressing resident fibroblasts that also expands after lung injury is a second major source of myofibroblasts. CONCLUSIONS We conclude that the lung contains an extensive population of Foxd1 progenitor-derived pericytes that are an important lung myofibroblast precursor population.
Collapse
Affiliation(s)
- Chi Hung
- 1 Center for Lung Biology, Pulmonary and Critical Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
785
|
c-MET regulates myoblast motility and myocyte fusion during adult skeletal muscle regeneration. PLoS One 2013; 8:e81757. [PMID: 24260586 PMCID: PMC3834319 DOI: 10.1371/journal.pone.0081757] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 10/16/2013] [Indexed: 11/19/2022] Open
Abstract
Adult muscle stem cells, satellite cells (SCs), endow skeletal muscle with tremendous regenerative capacity. Upon injury, SCs activate, proliferate, and migrate as myoblasts to the injury site where they become myocytes that fuse to form new muscle. How migration is regulated, though, remains largely unknown. Additionally, how migration and fusion, which both require dynamic rearrangement of the cytoskeleton, might be related is not well understood. c-MET, a receptor tyrosine kinase, is required for myogenic precursor cell migration into the limb for muscle development during embryogenesis. Using a genetic system to eliminate c-MET function specifically in adult mouse SCs, we found that c-MET was required for muscle regeneration in response to acute muscle injury. c-MET mutant myoblasts were defective in lamellipodia formation, had shorter ranges of migration, and migrated slower compared to control myoblasts. Surprisingly, c-MET was also required for efficient myocyte fusion, implicating c-MET in dual functions of regulating myoblast migration and myocyte fusion.
Collapse
|
786
|
Cellular dynamics in the muscle satellite cell niche. EMBO Rep 2013; 14:1062-72. [PMID: 24232182 DOI: 10.1038/embor.2013.182] [Citation(s) in RCA: 269] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 10/21/2013] [Indexed: 12/14/2022] Open
Abstract
Satellite cells, the quintessential skeletal muscle stem cells, reside in a specialized local environment whose anatomy changes dynamically during tissue regeneration. The plasticity of this niche is attributable to regulation by the stem cells themselves and to a multitude of functionally diverse cell types. In particular, immune cells, fibrogenic cells, vessel-associated cells and committed and differentiated cells of the myogenic lineage have emerged as important constituents of the satellite cell niche. Here, we discuss the cellular dynamics during muscle regeneration and how disease can lead to perturbation of these mechanisms. To define the role of cellular components in the muscle stem cell niche is imperative for the development of cell-based therapies, as well as to better understand the pathobiology of degenerative conditions of the skeletal musculature.
Collapse
|
787
|
Boppart MD, De Lisio M, Zou K, Huntsman HD. Defining a role for non-satellite stem cells in the regulation of muscle repair following exercise. Front Physiol 2013; 4:310. [PMID: 24204344 PMCID: PMC3817631 DOI: 10.3389/fphys.2013.00310] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/10/2013] [Indexed: 11/16/2022] Open
Abstract
Skeletal muscle repair is essential for effective remodeling, tissue maintenance, and initiation of beneficial adaptations post-eccentric exercise. A series of well characterized events, such as recruitment of immune cells and activation of satellite cells, constitute the basis for muscle regeneration. However, details regarding the fine-tuned regulation of this process in response to different types of injury are open for investigation. Muscle-resident non-myogenic, non-satellite stem cells expressing conventional mesenchymal stem cell (MSC) markers, have the potential to significantly contribute to regeneration given the role for bone marrow-derived MSCs in whole body tissue repair in response to injury and disease. The purpose of this mini-review is to highlight a regulatory role for Pnon-satellite stem cells in the process of skeletal muscle healing post-eccentric exercise. The non-myogenic, non-satellite stem cell fraction will be defined, its role in tissue repair will be briefly reviewed, and recent studies demonstrating a contribution to eccentric exercise-induced regeneration will be presented.
Collapse
Affiliation(s)
- Marni D Boppart
- Department of Kinesiology and Community Health, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana, IL, USA
| | | | | | | |
Collapse
|
788
|
Dayton WR, White ME. MEAT SCIENCE AND MUSCLE BIOLOGY SYMPOSIUM--role of satellite cells in anabolic steroid-induced muscle growth in feedlot steers. J Anim Sci 2013; 92:30-8. [PMID: 24166993 DOI: 10.2527/jas.2013-7077] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Both androgenic and estrogenic steroids are widely used as growth promoters in feedlot steers because they significantly enhance feed efficiency, rate of gain, and muscle growth. However, despite their widespread use relatively little is known about the biological mechanism by which androgenic and estrogenic steroids enhance rate and efficiency of muscle growth in cattle. Treatment of feedlot steers with a combined estradiol (E2) and trenbolone acetate (TBA) implant results in an increased number of muscle satellite cells, increased expression of IGF-1 mRNA in muscle tissue, and increased levels of circulating IGF-1. Similarly, treatment of bovine satellite cell (BSC) cultures with either TBA or E2 results in increased expression of IGF-1 mRNA, increased rates of proliferation and protein synthesis, and decreased rates of protein degradation. Effects of E2 on BSC are mediated at least in part through the classical E2 receptor, estrogen receptor-α (ESR1), the IGF-1 receptor (IGFR1), and the G protein-coupled estrogen receptor-1 (GPER-1), formerly known as G protein-coupled receptor-30 (GPR30). The effects of TBA appear to be primarily mediated through the androgen receptor. Based on current research results, it is becoming clear that anabolic steroid-enhanced bovine muscle growth involves a complex interaction of numerous pathways and receptors. Consequently, additional in vivo and in vitro studies are necessary to understand the mechanisms involved in this complex process. The fundamental information generated by this research will help in developing future, safe, and effective strategies to increase rate and efficiency of muscle growth in beef cattle.
Collapse
Affiliation(s)
- W R Dayton
- Department of Animal Science, University of Minnesota, St. Paul 55108
| | | |
Collapse
|
789
|
Agley CC, Rowlerson AM, Velloso CP, Lazarus NR, Harridge SDR. Human skeletal muscle fibroblasts, but not myogenic cells, readily undergo adipogenic differentiation. J Cell Sci 2013; 126:5610-25. [PMID: 24101731 DOI: 10.1242/jcs.132563] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We characterised the adherent cell types isolated from human skeletal muscle by enzymatic digestion, and demonstrated that even at 72 hours after isolation these cultures consisted predominantly of myogenic cells (CD56(+), desmin(+)) and fibroblasts (TE-7(+), collagen VI(+), PDGFRα(+), vimentin(+), fibronectin(+)). To evaluate the behaviour of the cell types obtained, we optimised a double immuno-magnetic cell-sorting method for the separation of myogenic cells from fibroblasts. This procedure gave purities of >96% for myogenic (CD56(+), desmin(+)) cells. The CD56(-) fraction obtained from the first sort was highly enriched in TE-7(+) fibroblasts. Using quantitative analysis of immunofluorescent staining for lipid content, lineage markers and transcription factors, we tested if the purified cell populations could differentiate into adipocytes in response to treatment with either fatty acids or adipocyte-inducing medium. Both treatments caused the fibroblasts to differentiate into adipocytes, as shown by loss of intracellular TE-7, upregulation of the adipogenic transcription factors PPARγ and C/EBPα, and adoption of a lipid-laden adipocyte morphology. By contrast, myogenic cells did not undergo adipogenesis and showed differential regulation of PPARγ and C/EBPα in response to these adipogenic treatments. Our results show that human skeletal muscle fibroblasts are at least bipotent progenitors that can remain as extracellular-matrix-producing cells or differentiate into adipocytes.
Collapse
Affiliation(s)
- Chibeza C Agley
- Centre of Human and Aerospace Physiological Sciences, School of Biomedical Sciences, King's College London, Shepherd's House, Guy's Campus, London SE1 1UL, UK
| | | | | | | | | |
Collapse
|
790
|
Corona BT, Garg K, Ward CL, McDaniel JS, Walters TJ, Rathbone CR. Autologous minced muscle grafts: a tissue engineering therapy for the volumetric loss of skeletal muscle. Am J Physiol Cell Physiol 2013; 305:C761-75. [DOI: 10.1152/ajpcell.00189.2013] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Volumetric muscle loss (VML) results in a large void deficient in the requisite materials for regeneration for which there is no definitive clinical standard of care. Autologous minced muscle grafts (MG), which contain the essential components for muscle regeneration, may embody an ideal tissue engineering therapy for VML. The purpose of this study was to determine if orthotopic transplantation of MG acutely after VML in the tibialis anterior muscle of male Lewis rats promotes functional tissue regeneration. Herein we report that over the first 16 wk postinjury, MG transplantation 1) promotes remarkable regeneration of innervated muscle fibers within the defect area (i.e., de novo muscle fiber regeneration); 2) reduced evidence of chronic injury in the remaining muscle mass compared with nonrepaired muscles following VML (i.e., transplantation attenuated chronically upregulated transforming growth factor-β1 gene expression and the presence of centrally located nuclei in 30% of fibers observed in nonrepaired muscles); and 3) significantly improves net torque production (i.e., ∼55% of the functional deficit in nonrepaired muscles was restored). Additionally, voluntary wheel running was shown to reduce the heightened accumulation of extracellular matrix deposition observed within the regenerated tissue of MG-repaired sedentary rats 8 wk postinjury (collagen 1% area: sedentary vs. runner, ∼41 vs. 30%), which may have been the result of an augmented inflammatory response [i.e., M1 (CCR7) and M2 (CD163) macrophage expression was significantly greater in runner than sedentary MG-repaired muscles 2 wk postinjury]. These findings support further exploration of autologous minced MGs for the treatment of VML.
Collapse
Affiliation(s)
- B. T. Corona
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - K. Garg
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - C. L. Ward
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - J. S. McDaniel
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - T. J. Walters
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - C. R. Rathbone
- Extremity Trauma and Regenerative Medicine Research Program, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| |
Collapse
|
791
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Type-1 pericytes participate in fibrous tissue deposition in aged skeletal muscle. Am J Physiol Cell Physiol 2013; 305:C1098-113. [PMID: 24067916 DOI: 10.1152/ajpcell.00171.2013] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In older adults, changes in skeletal muscle composition are associated with increased fibrosis, loss of mass, and decreased force, which can lead to dependency, morbidity, and mortality. Understanding the biological mechanisms responsible is essential to sustaining and improving their quality of life. Compared with young mice, aged mice take longer to recover from muscle injury; their tissue fibrosis is more extensive, and regenerated myofibers are smaller. Strong evidence indicates that cells called pericytes, embedded in the basement membrane of capillaries, contribute to the satellite-cell pool and muscle growth. In addition to their role in skeletal muscle repair, after tissue damage, they detach from capillaries and migrate to the interstitial space to participate in fibrosis formation. Here we distinguish two bona fide pericyte subtypes in the skeletal muscle interstitium, type-1 (Nestin-GFP(-)/NG2-DsRed(+)) and type-2 (Nestin-GFP(+)/NG2-DsRed(+)), and characterize their heretofore unknown specific roles in the aging environment. Our in vitro results show that type-1 and type-2 pericytes are either fibrogenic or myogenic, respectively. Transplantation studies in young animals indicate that type-2 pericytes are myogenic, while type-1 pericytes remain in the interstitial space. In older mice, however, the muscular regenerative capacity of type-2 pericytes is limited, and type-1 pericytes produce collagen, contributing to fibrous tissue deposition. We conclude that in injured muscles from aging mice, the pericytes involved in skeletal muscle repair differ from those associated with scar formation.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | | | | | |
Collapse
|
792
|
Wood WM, Etemad S, Yamamoto M, Goldhamer DJ. MyoD-expressing progenitors are essential for skeletal myogenesis and satellite cell development. Dev Biol 2013; 384:114-27. [PMID: 24055173 DOI: 10.1016/j.ydbio.2013.09.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 09/06/2013] [Accepted: 09/07/2013] [Indexed: 01/22/2023]
Abstract
Skeletal myogenesis in the embryo is regulated by the coordinated expression of the MyoD family of muscle regulatory factors (MRFs). MyoD and Myf-5, which are the primary muscle lineage-determining factors, function in a partially redundant manner to establish muscle progenitor cell identity. Previous diphtheria toxin (DTA)-mediated ablation studies showed that MyoD+ progenitors rescue myogenesis in embryos in which Myf-5-expressing cells were targeted for ablation, raising the possibility that the regulative behavior of distinct, MRF-expressing populations explains the functional compensatory activities of these MRFs. Using MyoD(iCre) mice, we show that DTA-mediated ablation of MyoD-expressing cells results in the cessation of myogenesis by embryonic day 12.5 (E12.5), as assayed by myosin heavy chain (MyHC) and Myogenin staining. Importantly, MyoD(iCre/+);R26(DTA/+) embryos exhibited a concomitant loss of Myf-5+ progenitors, indicating that the vast majority of Myf-5+ progenitors express MyoD, a conclusion consistent with immunofluorescence analysis of Myf-5 protein expression in MyoD(iCre) lineage-labeled embryos. Surprisingly, staining for the paired box transcription factor, Pax7, which functions genetically upstream of MyoD in the trunk and is a marker for fetal myoblasts and satellite cell progenitors, was also lost by E12.5. Specific ablation of differentiating skeletal muscle in ACTA1Cre;R26(DTA/+) embryos resulted in comparatively minor effects on MyoD+, Myf-5+ and Pax7+ progenitors, indicating that cell non-autonomous effects are unlikely to explain the rapid loss of myogenic progenitors in MyoD(iCre/+);R26(DTA/+) embryos. We conclude that the vast majority of myogenic cells transit through a MyoD+ state, and that MyoD+ progenitors are essential for myogenesis and stem cell development.
Collapse
Affiliation(s)
- William M Wood
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA
| | | | | | | |
Collapse
|
793
|
Washington TA, Brown L, Smith DA, Davis G, Baum J, Bottje W. Monocarboxylate transporter expression at the onset of skeletal muscle regeneration. Physiol Rep 2013; 1:e00075. [PMID: 24303150 PMCID: PMC3831894 DOI: 10.1002/phy2.75] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/24/2013] [Accepted: 07/26/2013] [Indexed: 12/24/2022] Open
Abstract
The onset of skeletal muscle regeneration is characterized by proliferating myoblasts. Proliferating myoblasts have an increased energy demand and lactate exchange across the sarcolemma can be used to address this increased demand. Monocarboxylate transporters (MCTs) are involved in lactate transport across the sarcolemma and are known to be affected by various physiological stimuli. However, MCT expression at the onset of skeletal muscle regeneration has not been determined. The purpose of this study was to determine if skeletal muscle regeneration altered MCT expression in regenerating tibialis anterior (TA) muscle. Male C57/BL6 mice were randomly assigned to either a control (uninjured) or bupivacaine (injured) group. Three days post injection, the TA was extracted for determination of protein and gene expression. A 21% decrease in muscle mass to tibia length (2.4 ± 0.1 mg/mm vs. 1.9 ± 0.2 mg/mm, P < 0.02) was observed. IGF-1 and MyoD gene expression increased 5.0-fold (P < 0.05) and 3.5-fold (P < 0.05), respectively, 3 days post bupivacaine injection. MCT-1 protein was decreased 32% (P < 0.03); however, MCT-1 gene expression was not altered. There was no difference in MCT4 protein or gene expression. Lactate dehydrogenase (LDH)-A protein expression increased 71% (P < 0.0004). Protein levels of LDH-B and mitochondrial enzyme cytochrome C oxidase subunit decreased 3 days post bupivacaine injection. CD147 and PKC-θ protein increased 64% (P < 0.03) and 79% (P < 0.02), respectively. MCT1 but not MCT4 expression is altered at the onset of skeletal muscle regeneration possibly in an attempt to regulate lactate uptake and use by skeletal muscle cells.
Collapse
Affiliation(s)
- Tyrone A Washington
- Exercise Muscle Biology Laboratory, University of Arkansas Fayetteville, Arkansas, 72701 ; Human Performance Laboratory, Department of Health, Human Performance and Recreation, University of Arkansas Fayetteville, Arkansas, 72701
| | | | | | | | | | | |
Collapse
|
794
|
Van Ry PM, Minogue P, Hodges BL, Burkin DJ. Laminin-111 improves muscle repair in a mouse model of merosin-deficient congenital muscular dystrophy. Hum Mol Genet 2013; 23:383-96. [PMID: 24009313 DOI: 10.1093/hmg/ddt428] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Merosin-deficient congenital muscular dystrophy type 1A (MDC1A) is a severe and fatal muscle-wasting disease with no cure. MDC1A patients and the dy(W-/-) mouse model exhibit severe muscle weakness, demyelinating neuropathy, failed muscle regeneration and premature death. We have recently shown that laminin-111, a form of laminin found in embryonic skeletal muscle, can substitute for the loss of laminin-211/221 and prevent muscle disease progression in the dy(W-/-) mouse model. What is unclear from these studies is whether laminin-111 can restore failed regeneration to laminin-α2-deficient muscle. To investigate the potential of laminin-111 protein therapy to improve muscle regeneration, laminin-111 or phosphate-buffered saline-treated laminin-α2-deficient muscle was damaged with cardiotoxin and muscle regeneration quantified. Our results show laminin-111 treatment promoted an increase in myofiber size and number, and an increased expression of α7β1 integrin, Pax7, myogenin and embryonic myosin heavy chain, indicating a restoration of the muscle regenerative program. Together, our results show laminin-111 restores muscle regeneration to laminin-α2-deficient muscle and further supports laminin-111 protein as a therapy for the treatment of MDC1A.
Collapse
Affiliation(s)
- Pam M Van Ry
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA and
| | | | | | | |
Collapse
|
795
|
Kirby TJ, McCarthy JJ. MicroRNAs in skeletal muscle biology and exercise adaptation. Free Radic Biol Med 2013; 64:95-105. [PMID: 23872025 PMCID: PMC4867469 DOI: 10.1016/j.freeradbiomed.2013.07.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 07/01/2013] [Accepted: 07/02/2013] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRNAs) have emerged as important players in the regulation of gene expression, being involved in most biological processes examined to date. The proposal that miRNAs are primarily involved in the stress response of the cell makes miRNAs ideally suited to mediate the response of skeletal muscle to changes in contractile activity. Although the field is still in its infancy, the studies presented in this review highlight the promise that miRNAs will have an important role in mediating the response and adaptation of skeletal muscle to various modes of exercise. The roles of miRNAs in satellite cell biology, muscle regeneration, and various myopathies are also discussed.
Collapse
Affiliation(s)
- Tyler J. Kirby
- Department of Physiology, University of Kentucky Lexington, KY 40516-0298
| | - John J. McCarthy
- Department of Physiology, University of Kentucky Lexington, KY 40516-0298
- Center for Muscle Biology, University of Kentucky Lexington, KY 40516-0298
| |
Collapse
|
796
|
Briggs D, Morgan JE. Recent progress in satellite cell/myoblast engraftment -- relevance for therapy. FEBS J 2013; 280:4281-93. [PMID: 23560812 PMCID: PMC3795440 DOI: 10.1111/febs.12273] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/27/2013] [Accepted: 03/28/2013] [Indexed: 12/18/2022]
Abstract
There is currently no cure for muscular dystrophies, although several promising strategies are in basic and clinical research. One such strategy is cell transplantation with satellite cells (or their myoblast progeny) to repair damaged muscle and provide dystrophin protein with the aim of preventing subsequent myofibre degeneration and repopulating the stem cell niche for future use. The present review aims to cover recent advances in satellite cell/myoblast therapy and to discuss the challenges that remain for it to become a realistic therapy.
Collapse
Affiliation(s)
- Deborah Briggs
- The Dubowitz Neuromuscular Centre, UCL Institute of Child HealthLondon, UK
| | - Jennifer E Morgan
- The Dubowitz Neuromuscular Centre, UCL Institute of Child HealthLondon, UK
| |
Collapse
|
797
|
Shono JI, Sakaguchi S, Suzuki T, Do MKQ, Mizunoya W, Nakamura M, Sato Y, Furuse M, Yamada K, Ikeuchi Y, Tatsumi R. Preliminary time-course study of antiinflammatory macrophage infiltration in crush-injured skeletal muscle. Anim Sci J 2013; 84:744-50. [PMID: 23980916 DOI: 10.1111/asj.12105] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 06/13/2013] [Indexed: 12/14/2022]
Abstract
Muscle damage induces massive macrophage infiltration of the injury site, in which activated pro-inflammatory and anti-inflammatory phenotypes (currently classified as M1 and M2, respectively) have been documented as distinct functional populations predominant at different times after the conventional acute injury by intramuscular injection of snake venoms (cardiotoxin, notexin) or chemicals (bupivacaine hydrochloride, barium chloride). The present study employed a muscle-crush injury model that may better reflect the physiologic damage and repair processes initiated by contusing a gastrocnemius muscle in the lower hind-limb of adult mice with hemostat forceps, and examined the time-course invasion of M1 and M2 macrophages during muscle regeneration by immunocytochemistry of CD197 and CD206 marker proteins. CD197-positive M1 macrophages were observed exclusively at 1-4 days after crush followed by the alternative prevalence of CD206-positive M2 at 7 days of myogenic differentiation, characterized by increasing levels of myogenin messenger RNA expression. Preliminary PCR analysis showed that M2 may produce hepatocyte growth factor (HGF) in culture, providing additional benefit to understanding that M2 populations actively promote regenerative myogenesis (muscle fiber repair) and moto-neuritogenesis (re-attachment of motoneuron terminals onto damaged fibers) through their time-specific infiltration and release of growth factor at the injury site early in muscle regeneration.
Collapse
Affiliation(s)
- Jun-ichi Shono
- Department of Animal and Marine Bioresource Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
798
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Enikolopov GN, Mintz A, Delbono O. Role of pericytes in skeletal muscle regeneration and fat accumulation. Stem Cells Dev 2013; 22:2298-314. [PMID: 23517218 PMCID: PMC3730538 DOI: 10.1089/scd.2012.0647] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 03/20/2013] [Indexed: 02/06/2023] Open
Abstract
Stem cells ensure tissue regeneration, while overgrowth of adipogenic cells may compromise organ recovery and impair function. In myopathies and muscle atrophy associated with aging, fat accumulation increases dysfunction, and after chronic injury, the process of fatty degeneration, in which muscle is replaced by white adipocytes, further compromises tissue function and environment. Some studies suggest that pericytes may contribute to muscle regeneration as well as fat formation. This work reports the presence of two pericyte subpopulations in the skeletal muscle and characterizes their specific roles. Skeletal muscle from Nestin-GFP/NG2-DsRed mice show two types of pericytes, Nestin-GFP-/NG2-DsRed+ (type-1) and Nestin-GFP+/NG2-DsRed+ (type-2), in close proximity to endothelial cells. We also found that both Nestin-GFP-/NG2-DsRed+ and Nestin-GFP+/NG2-DsRed+ cells colocalize with staining of two pericyte markers, PDGFRβ and CD146, but only type-1 pericyte express the adipogenic progenitor marker PDGFRα. Type-2 pericytes participate in muscle regeneration, while type-1 contribute to fat accumulation. Transplantation studies indicate that type-1 pericytes do not form muscle in vivo, but contribute to fat deposition in the skeletal muscle, while type-2 pericytes contribute only to the new muscle formation after injury, but not to the fat accumulation. Our results suggest that type-1 and type-2 pericytes contribute to successful muscle regeneration which results from a balance of myogenic and nonmyogenic cells activation.
Collapse
MESH Headings
- Adipogenesis/genetics
- Animals
- Antigens/genetics
- Antigens/metabolism
- CD146 Antigen/genetics
- CD146 Antigen/metabolism
- Cell Lineage/genetics
- Endothelial Cells/cytology
- Female
- Gene Expression
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Male
- Mice
- Mice, Nude
- Mice, Transgenic
- Muscle, Skeletal/cytology
- Muscle, Skeletal/injuries
- Muscle, Skeletal/metabolism
- Nestin/genetics
- Nestin/metabolism
- Pericytes/cytology
- Pericytes/metabolism
- Pericytes/transplantation
- Proteoglycans/genetics
- Proteoglycans/metabolism
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Regeneration/genetics
- Red Fluorescent Protein
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Grigori N. Enikolopov
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- NBIC, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
799
|
Mitsuhashi S, Mitsuhashi H, Alexander MS, Sugimoto H, Kang PB. Cysteine mutations cause defective tyrosine phosphorylation in MEGF10 myopathy. FEBS Lett 2013; 587:2952-7. [PMID: 23954233 DOI: 10.1016/j.febslet.2013.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/27/2013] [Accepted: 08/01/2013] [Indexed: 01/06/2023]
Abstract
Recessive mutations in MEGF10 are known to cause a congenital myopathy in humans. Two mutations in the extracellular EGF-like domains of MEGF10, C326R and C774R, were associated with decreased tyrosine phosphorylation of MEGF10 in vitro. Y1030 was identified to be the major tyrosine phosphorylation site in MEGF10 and is phosphorylated at least in part by c-Src. Overexpression of wild-type MEGF10 enhanced C2C12 myoblast proliferation, while overexpression of Y1030F mutated MEGF10 did not. We conclude that MEGF10-mediated signaling via tyrosine phosphorylation helps to regulate myoblast proliferation. Defects in this signaling pathway may contribute to the disease mechanism of MEGF10 myopathy.
Collapse
Affiliation(s)
- Satomi Mitsuhashi
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
800
|
Myf5-positive satellite cells contribute to Pax7-dependent long-term maintenance of adult muscle stem cells. Cell Stem Cell 2013; 13:590-601. [PMID: 23933088 DOI: 10.1016/j.stem.2013.07.016] [Citation(s) in RCA: 206] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 07/12/2013] [Accepted: 07/19/2013] [Indexed: 11/21/2022]
Abstract
Skeletal muscle contains Pax7-expressing muscle stem or satellite cells, enabling muscle regeneration throughout most of adult life. Here, we demonstrate that induced inactivation of Pax7 in Pax7-expressing cells of adult mice leads to loss of muscle stem cells and reduced heterochromatin condensation in rare surviving satellite cells. Inactivation of Pax7 in Myf5-expressing cells revealed that the majority of adult muscle stem cells originate from myogenic lineages, which express the myogenic regulators Myf5 or MyoD. Likewise, the majority of muscle stem cells are replenished from Myf5-expressing myogenic cells during adult life, and inactivation of Pax7 in Myf5-expressing cells after muscle damage leads to a complete arrest of muscle regeneration. Finally, we demonstrate that a relatively small number of muscle stem cells are sufficient for efficient repair of skeletal muscles. We conclude that Pax7 acts at different levels in a nonhierarchical regulatory network controlling muscle-satellite-cell-mediated muscle regeneration.
Collapse
|