201
|
Liu Y, Yan X, Sun Z, Chen B, Han Q, Li J, Zhao RC. Flk-1+ adipose-derived mesenchymal stem cells differentiate into skeletal muscle satellite cells and ameliorate muscular dystrophy in mdx mice. Stem Cells Dev 2008; 16:695-706. [PMID: 17999592 DOI: 10.1089/scd.2006.0118] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe hereditary disease characterized by the absence of dystrophin on the sarcolemma of muscle fiber. This absence results in widespread muscle damage and satellite cell activation. After depletion of the satellite cell pool, skeletal muscle is then invariably replaced by connective tissue, leading to progressive muscle weakness. Herein, we isolated Flk-1(+) mesenchymal stem cells (MSCs) from adult adipose tissue and induced them to differentiate into skeletal muscle cells in culture. Within mdx mice, an animal model of DMD, adipose tissue-derived Flk-1(+) MSCs (AD-MSCs) homed to and differentiated into cells that repaired injured muscle tissue. This repair correlated with reconstitution of dystrophin expression on the damaged fibers. Flk-1(+) AD-MSCs also differentiated into muscle satellite cells. This differentiation may have accounted for long-term reconstitution. These cells also differentiated into endothelial cells, thereby possibly improving fiber regeneration as a result of the induced angiogenesis. Therefore, Flk-1(+) AD-MSC transplants may repair muscular dystrophy.
Collapse
Affiliation(s)
- Yanning Liu
- Institute of Basic Medical Sciences & School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | | | | | | | | | | | | |
Collapse
|
202
|
Lin CS, Hsu CW. Differentially transcribed genes in skeletal muscle of Duroc and Taoyuan pigs. J Anim Sci 2008; 83:2075-86. [PMID: 16100062 DOI: 10.2527/2005.8392075x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The objective of this study was to compare gene transcription profiles of LM between two pig breeds, Duroc and Taoyuan, which display dramatically different postnatal muscle growth. We isolated LM from neonatal pigs, and the Duroc muscle length and mass were greater (P < 0.01) than for Taoyuan pigs; however, insignificant differences in the muscle fiber area and the percentage of fiber types were found. A human high-density complementary DNA (cDNA) microarray consisting of 9,182 probes was used to compare gene transcription profiles of LM between the two breeds. The results showed that the transcription level of 73 genes and 44 genes in Duroc LM were upregulated and down-regulated by at least 1.75-fold (P < 0.05) compared with Taoyuan, respectively. The strongly upregulated genes in Duroc pigs included those encoding the complex of myofibrillar proteins (e.g., myosin light and heavy chains, and troponin), ribosomal proteins, transcription regulatory proteins (e.g., skeletal muscle LIM protein 1 [SLIM1] and high-mobility group proteins), and energy metabolic enzymes (e.g., electron-transferring flavo-protein dehydrogenase, NADH dehydrogenase, malate dehydrogenase, and ATP synthases). The highly transcribed genes that encode energy metabolic enzymes indicate a more glycolytic metabolism in Duroc LM, thereby favoring carbohydrates rather than lipids for use as energy substrates in this tissue. The over-transcribed genes that encode skeletal muscle-predominant proteins or transcription regulators that control myogenesis and/or muscle growth suggest a general mechanism for the observed higher rate of postnatal muscle growth in Duroc pigs. The transcription of one such gene, SLIM1, was more highly transcribed (P < 0.01) in Duroc LM at birth and at postnatal d 7 than in Taoyuan. The transcription of SLIM1 increased (P < 0.05) in Duroc LM from neonate through 7 d of age, whereas its transcription remained essentially constant in Taoyuan during this period. These results suggest that SLIM1 may be useful for the development of markers associated with the postnatal muscle growth of pigs.
Collapse
Affiliation(s)
- C S Lin
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan, Republic of China.
| | | |
Collapse
|
203
|
Deponti D, François S, Baesso S, Sciorati C, Innocenzi A, Broccoli V, Muscatelli F, Meneveri R, Clementi E, Cossu G, Brunelli S. Necdin mediates skeletal muscle regeneration by promoting myoblast survival and differentiation. ACTA ACUST UNITED AC 2007; 179:305-19. [PMID: 17954612 PMCID: PMC2064766 DOI: 10.1083/jcb.200701027] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Regeneration of muscle fibers that are lost during pathological muscle degeneration or after injuries is sustained by the production of new myofibers. An important cell type involved in muscle regeneration is the satellite cell. Necdin is a protein expressed in satellite cell–derived myogenic precursors during perinatal growth. However, its function in myogenesis is not known. We compare transgenic mice that overexpress necdin in skeletal muscle with both wild-type and necdin null mice. After muscle injury the necdin null mice show a considerable defect in muscle healing, whereas mice that overexpress necdin show a substantial increase in myofiber regeneration. We also find that in muscle, necdin increases myogenin expression, accelerates differentiation, and counteracts myoblast apoptosis. Collectively, these data clarify the function and mechanism of necdin in skeletal muscle and show the importance of necdin in muscle regeneration.
Collapse
Affiliation(s)
- Daniela Deponti
- Department of Histology and Medical Embryology, University of Roma-La Sapienza, 00161 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Ozernyuk ND, Balan OV. Muscle satellite cells and regulation of recovery potential of muscle. BIOL BULL+ 2007. [DOI: 10.1134/s1062359007060027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
205
|
Watanabe S, Kondo S, Hayasaka M, Hanaoka K. Functional analysis of homeodomain-containing transcription factor Lbx1 in satellite cells of mouse skeletal muscle. J Cell Sci 2007; 120:4178-87. [PMID: 18003701 DOI: 10.1242/jcs.011668] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Satellite cells are usually mitotically quiescent muscle stem cells, located between the sarcolemma and the basement membrane of muscle fibers. When muscles are damaged, satellite cells become activated, proliferate and differentiate to form multinucleate myofibers. The molecular mechanisms underlying these processes are poorly understood. In the present study, we found that, following treatment with cardiotoxin, homeodomain-containing transcription factor Lbx1 was strongly expressed in the satellite cells of regenerating adult skeletal muscle. Our immunohistochemical and northern blot analyses indicate that Lbx1 is expressed in activated but not quiescent satellite cells. In vitro, this Lbx1 expression was gradually downregulated when satellite cells differentiate into mature myofibers. Transfection and forced expression of Lbx1 in satellite-cell-derived C2C12 myoblast cells resulted in severe depression of myogenic differentiation and incomplete myotube formation, concomitantly with the activation of the paired-box transcription factor Pax7 and depression of the myogenic regulatory factor MyoD. Moreover, knockdown of Lbx1 in in-vitro-cultured satellite cells resulted in downregulation of Pax7. These results suggest that Lbx1 plays important roles in differentiation and maintenance of satellite cells of mature myofibers, probably through the regulation of Pax7.
Collapse
Affiliation(s)
- Shuichi Watanabe
- Molecular Embryology, Department of Biosciences, School of Science, Kitasato University, Kanagawa, 228-8555, Japan
| | | | | | | |
Collapse
|
206
|
Kumai Y, Ito T, Miyamaru S, Yumoto E. Modulation of MyoD- and Ki-67-Positive Satellite Cells in the Short-Term Denervated Rat Thyroarytenoid Muscle. Laryngoscope 2007; 117:2063-7. [PMID: 17828047 DOI: 10.1097/mlg.0b013e318133a13c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To evaluate the effects of short-term denervation on the kinetics of satellite cells (SCs) and myocytes in the rat thyroarytenoid (TA) muscle using immunohistochemistry for a myogenic regulatory factor, MyoD, and a cell proliferation marker, Ki-67. STUDY DESIGN Quantitative immunohistochemical assessment of MyoD and Ki-67 expression in SCs and myocytes after denervation. METHODS The left recurrent laryngeal nerve (RLN) was transected in 15 adult Wistar rats, which were sacrificed 1, 3, or 7 days after the treatment (n = 5, each group). Fluorescein immunostaining was used to visualize the localization of MyoD+ and Ki-67+ SCs in combination with M-cadherin immunostaining for detection of SCs. We examined the temporal changes of the ratios of MyoD+ and Ki-67+ SCs for all of the counted muscle fibers and M-cadherin+ cells in both the denervated and the contralateral control TA muscle. RESULTS On the denervated side, the TA muscle contained 3.8 +/- 0.4% MyoD+ SCs and 1.3 +/- 0.4% Ki-67+ SCs. Of the SCs, 22.6 +/- 2.2% were MyoD+, and 14.5 +/- 4.4% were Ki-67+, whereas SCs on the control side did not express these markers. CONCLUSIONS In the rat TA muscle, denervation induces SC activation, and some SCs enter the cell cycle, whereas others are involved in the differentiation process. The number of activated SCs is relatively small compared with all M-cadherin+ SCs. Therefore, SCs might be good targets for a therapy to prevent TA muscle atrophy after RLN paralysis.
Collapse
Affiliation(s)
- Yoshihiko Kumai
- Department of Otolaryngology Head and Neck Surgery, Kumamoto University, Graduate School of Medicine, Honjo, Kumamoto, Japan.
| | | | | | | |
Collapse
|
207
|
Tsukamoto Y, Hijiya N, Yano S, Yokoyama S, Nakada C, Uchida T, Matsuura K, Moriyama M. Arpp/Ankrd2, a member of the muscle ankyrin repeat proteins (MARPs), translocates from the I-band to the nucleus after muscle injury. Histochem Cell Biol 2007; 129:55-64. [PMID: 17926058 DOI: 10.1007/s00418-007-0348-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2007] [Indexed: 01/01/2023]
Abstract
Ankyrin-repeat protein with a PEST motif and a proline-rich region (Arpp), also designated as Ankrd2, is a member of the muscle ankyrin repeat proteins (MARPs), which have been proposed to be involved in muscle stress response pathways. Arpp/Ankrd2 is localized mainly in the I-band of striated muscle. However, it has recently been reported that Arpp/Ankrd2 can interact with nuclear proteins, such as premyelocytic leukemia protein (PML), p53 and YB-1 in vitro. In this study, to determine whether nuclear accumulation of Arpp/Ankrd2 actually occurs, we performed an immunohistochemical investigation of gastrocnemius muscles that had been injured by injection of cardiotoxin or contact with dry ice. We found that Arpp/Ankrd2 accumulated in the nuclei of myofibers located adjacent to severely damaged myofibers after muscle injury. Double-labeled immunohistochemistry revealed that Arpp/Ankrd2 accumulated in the nuclei of sarcomere-damaged myofibers. Furthermore, we found that Arpp/Ankrd2 tended to be localized in euchromatin where genes are transcriptionally activated. Based on these findings, we suggest that Arpp/Ankrd2 may translocate from the I-band to the nucleus in response to muscle damage and may participate in the regulation of gene expression.
Collapse
Affiliation(s)
- Yoshiyuki Tsukamoto
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Yufu-city, Oita, 879-5593, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
208
|
Marotta M, Sarria Y, Ruiz-Roig C, Munell F, Roig-Quilis M. Laser microdissection-based expression analysis of key genes involved in muscle regeneration in mdx mice. Neuromuscul Disord 2007; 17:707-18. [PMID: 17611107 DOI: 10.1016/j.nmd.2007.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Revised: 05/03/2007] [Accepted: 05/16/2007] [Indexed: 10/23/2022]
Abstract
We have used the mdx mice strain (C57BL/10ScSn-mdx) as an experimental subject for the study of reiterative skeletal muscle necrosis-regeneration with basement membrane preservation. In young mdx muscle, by means of Hematoxylin-Eosin staining, different types of degenerative-regenerative groups (DRG) can be recognized and assigned to a defined muscle regeneration phase. To evaluate the expression of known key-regulatory genes in muscle regeneration, we have applied Laser Capture Microdissection technique to obtain tissue from different DRGs encompassing the complete skeletal muscle regenerative process. The expression of MyoD, Myf-5 and Myogenin showed a rapid increase in the first two days post-necrosis, which were followed by MRF4 expression, when newly regenerating fibers started to appear (3-5days post-necrosis). MHCd mRNA levels, undetectable in mature non-injured fibers, increased progressively from the first day post-necrosis and reached its maximum level of expression in DRGs showing basophilic regenerating fibers. TGFbeta-1 mRNA expression showed a prompt and strong increase following fiber necrosis that persisted during the inflammatory phase, and progressively decreased when new regenerating fibers began to appear. In contrast, IGF-2 mRNA expression decreased during the first days post-necrosis but was followed by a progressive rise in its expression coinciding with the appearance of the newly formed myofibers, reaching the maximum expression levels in DRGs composed of medium caliber basophilic regenerating myofibers (5-7 days post-necrosis). mdx degenerative-regenerative group typing, in conjunction with laser microdissection-based gene expression analysis, opens up a new approach to the molecular study of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Mario Marotta
- Laboratori de Neurología Infantil, Institut de Recerca Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain.
| | | | | | | | | |
Collapse
|
209
|
Involvement of CAPON and Nitric Oxide Synthases in Rat Muscle Regeneration After Peripheral Nerve Injury. J Mol Neurosci 2007; 34:89-100. [DOI: 10.1007/s12031-007-9005-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Accepted: 08/23/2007] [Indexed: 11/24/2022]
|
210
|
L'Honoré A, Coulon V, Marcil A, Lebel M, Lafrance-Vanasse J, Gage P, Camper S, Drouin J. Sequential expression and redundancy of Pitx2 and Pitx3 genes during muscle development. Dev Biol 2007; 307:421-33. [PMID: 17540357 DOI: 10.1016/j.ydbio.2007.04.034] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 04/26/2007] [Accepted: 04/26/2007] [Indexed: 11/24/2022]
Abstract
The myogenic program is controlled by different groups of transcription factors acting during muscle development, including bHLH muscle regulatory factors (MRFs), the paired factors Pax3 and Pax7 and the homeobox factors Six1 and Six4. This program is critically dependent on MRFs that target downstream muscle-specific genes. We now report the expression of Pitx2 and Pitx3 transcription factors throughout muscle development. Pitx2 is first expressed in muscle progenitor cells of the dermomyotome and myotome. The onset of myoblast differentiation is concomitant with expression of Pitx3; its expression is maintained in all skeletal muscles while Pitx2 expression decreases thereafter. We have generated Pitx3 mutant mice and this deficiency does not significantly perturb muscle development but it is completely compensated by the maintenance of Pitx2 expression in all skeletal muscles. These experiments suggest that Pitx genes are important for myogenesis and that Pitx2 and Pitx3 may have partly redundant roles.
Collapse
Affiliation(s)
- Aurore L'Honoré
- Laboratoire de génétique moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), 110, avenue des Pins Ouest, Montréal, QC, Canada H2W 1R7
| | | | | | | | | | | | | | | |
Collapse
|
211
|
Parise G, O'Reilly CE, Rudnicki MA. Molecular regulation of myogenic progenitor populations. Appl Physiol Nutr Metab 2007; 31:773-81. [PMID: 17213899 DOI: 10.1139/h06-055] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Skeletal muscle regeneration and adaptation to exercise require the actions of muscle satellite cells. Muscle satellite cells are thought to play an integral role in the process of exercise adaptation, but have also been shown to possess the capacity to fully regenerate muscle tissue following destructive muscle injury. We now know that molecular regulation of satellite cells involves the coordinated actions of a series of transcriptional networks that leads to myogenic commitment, cell-cycle entry, proliferation, and terminal differentiation. Additionally, Pax7 is a paired-box transcription factor that has been identified as playing a critical role in satellite cell regulation. It remains debatable, however, whether Pax7 is required for the specification of satellite cells and (or) whether it is playing a vital role in self-renewal and maintenance of the satellite cell population. In recent years, the emergence of atypical myogenic progenitor populations has added a new dimension to muscle repair, and significant interest has been focused on identifying populations such as bone-marrow-derived stem cells that have the ability to contribute to muscle. Interestingly, elucidating the molecular regulation of myogenic progenitor populations has involved animal models of muscle regeneration, with questionable relevance for human muscle adaptation to exercise. This paper highlights the current state of knowledge on the molecular regulation of satellite cells, explores the potential contribution of atypical myogenic progenitors, and discusses the information gathered from animal regeneration models in terms of its relevance to the process of exercise adaptation.
Collapse
Affiliation(s)
- Gianni Parise
- Department of Kinesiology, McMaster University, 1280 Main St. W., Hamilton, ON L8S 4K1, Canada.
| | | | | |
Collapse
|
212
|
Fukada SI, Uezumi A, Ikemoto M, Masuda S, Segawa M, Tanimura N, Yamamoto H, Miyagoe-Suzuki Y, Takeda S. Molecular signature of quiescent satellite cells in adult skeletal muscle. Stem Cells 2007; 25:2448-59. [PMID: 17600112 DOI: 10.1634/stemcells.2007-0019] [Citation(s) in RCA: 349] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Skeletal muscle satellite cells play key roles in postnatal muscle growth and regeneration. To study molecular regulation of satellite cells, we directly prepared satellite cells from 8- to 12-week-old C57BL/6 mice and performed genome-wide gene expression analysis. Compared with activated/cycling satellite cells, 507 genes were highly upregulated in quiescent satellite cells. These included negative regulators of cell cycle and myogenic inhibitors. Gene set enrichment analysis revealed that quiescent satellite cells preferentially express the genes involved in cell-cell adhesion, regulation of cell growth, formation of extracellular matrix, copper and iron homeostasis, and lipid transportation. Furthermore, reverse transcription-polymerase chain reaction on differentially expressed genes confirmed that calcitonin receptor (CTR) was exclusively expressed in dormant satellite cells but not in activated satellite cells. In addition, CTR mRNA is hardly detected in nonmyogenic cells. Therefore, we next examined the expression of CTR in vivo. CTR was specifically expressed on quiescent satellite cells, but the expression was not found on activated/proliferating satellite cells during muscle regeneration. CTR-positive cells reappeared at the rim of regenerating myofibers in later stages of muscle regeneration. Calcitonin stimulation delayed the activation of quiescent satellite cells. Our data provide roles of CTR in quiescent satellite cells and a solid scaffold to further dissect molecular regulation of satellite cells. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- So-ichiro Fukada
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Invernici G, Emanueli C, Madeddu P, Cristini S, Gadau S, Benetti A, Ciusani E, Stassi G, Siragusa M, Nicosia R, Peschle C, Fascio U, Colombo A, Rizzuti T, Parati E, Alessandri G. Human fetal aorta contains vascular progenitor cells capable of inducing vasculogenesis, angiogenesis, and myogenesis in vitro and in a murine model of peripheral ischemia. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1879-92. [PMID: 17525256 PMCID: PMC1899439 DOI: 10.2353/ajpath.2007.060646] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/15/2007] [Indexed: 12/19/2022]
Abstract
Vasculogenesis, the formation of blood vessels in embryonic or fetal tissue mediated by immature vascular cells (ie, angioblasts), is poorly understood. We report the identification of a population of vascular progenitor cells (hVPCs) in the human fetal aorta composed of undifferentiated mesenchymal cells that coexpress endothelial and myogenic markers. Under culture conditions that promoted cell differentiation, hVPCs gave rise to a mixed population of mature endothelial and mural cells when progenitor cells were stimulated with vascular endothelial growth factor-A or platelet-derived growth factor-betabeta. hVPCs grew as nonadherent cells and, when embedded in a three-dimensional collagen gel, reorganized into cohesive cellular cords that resembled mature vascular structures. hVPC-conditioned medium contained angiogenic substances (vascular endothelial growth factor-A and angiopoietin-2) and strongly stimulated the proliferation of endothelial cells. We also demonstrate the therapeutic efficacy of a small number of hVPCs transplanted into ischemic limb muscle of immunodeficient mice. hVPCs markedly improved neovascularization and inhibited the loss of endogenous endothelial cells and myocytes, thus ameliorating the clinical outcome from ischemia. We conclude that fetal aorta represents an important source for the investigation of the phenotypic and functional features of human vascular progenitor cells.
Collapse
Affiliation(s)
- Gloria Invernici
- Neurobiology and Neuroregenerative Therapies Unit, Carlo Besta Neurological Institute, Milan 20133, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Qin RF, Mao TQ, Gu XM, Hu KJ, Liu YP, Chen JW, Nie X. Regulation of skeletal muscle differentiation in fibroblasts by exogenous MyoD gene in vitro and in vivo. Mol Cell Biochem 2007; 302:233-9. [PMID: 17415623 DOI: 10.1007/s11010-007-9446-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 03/02/2007] [Indexed: 01/20/2023]
Abstract
MyoD of the myogenic regulatory factors (MRFs) family regulates the skeletal muscle differentiation program. In this study, stably transfected NIH3T3-derived cell lines were established, in which exogenous MyoD was expressed at high levels. Transcriptional activation of endogenous muscle regulatory gene and induction towards the skeletal muscle lineages were observed with phase-contrast microscopy when continuously cultured in vitro. Moreover, to determine their ability of myogenic formation in vivo, the transfected cells were implanted in nude mice subcutaneously for up to 10 weeks. The morphological characterization of inductive cells was observed using transmission electron microscope and histological staining. Myogenesis of fibroblasts incubated in the medium was activated by overexpression of MyoD, and the cells were accumulated and fused into multinucleated myotubes. Correlatively, RT-PCR and immunohistochemistry confirmed the increased expression of characteristic downstream molecule myogenin and mysion heavy chains during myogenic differentiation. Ecoptic myogenesis was found and remained stable phenotype when the transfected cells were seeded in vivo. Our results suggest that MyoD can be considered to be a determining factor of myogenic lineages, and it may play an important role in the cell therapy and cell-mediated gene therapy of the skeletal muscle.
Collapse
Affiliation(s)
- Rui-Feng Qin
- Department of Oral Maxillofacial Surgery, Qin Du Stomatological College, Fourth Military Medical University, Xian, P.R. China
| | | | | | | | | | | | | |
Collapse
|
215
|
Ko JA, Kimura Y, Matsuura K, Yamamoto H, Gondo T, Inui M. PDZRN3 (LNX3, SEMCAP3) is required for the differentiation of C2C12 myoblasts into myotubes. J Cell Sci 2006; 119:5106-13. [PMID: 17118964 DOI: 10.1242/jcs.03290] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PDZRN3 contains a RING-finger motif in its N-terminal region, two PDZ domains in its central region and a consensus-binding motif for PDZ domains at its C-terminus. It was identified in silico as a homolog of the protein known as LNX1 or SEMCAP1, which possesses ubiquitin ligase activity and binds the membrane protein Semaphorin 4C. However, PDZRN3 itself has not previously been characterized. We have now evaluated the properties and functions of PDZRN3. The PDZRN3 gene was shown to be expressed in various human tissues including the heart, skeletal muscle and liver and its expression in mouse skeletal muscle was developmentally regulated. Both the differentiation of C2C12 mouse skeletal myoblasts into myotubes and injury-induced muscle regeneration in vivo were found to be accompanied by up-regulation of PDZRN3. The differentiation-associated increase in the expression of PDZRN3 in C2C12 cells follows that of myogenin and precedes that of myosin heavy chain. Depletion of PDZRN3 by RNA interference inhibited the formation of myotubes as well as the associated up-regulation of myosin heavy chain in C2C12 cells. Our data suggest that PDZRN3 plays an essential role in the differentiation of myoblasts into myotubes by acting either downstream or independently of myogenin.
Collapse
Affiliation(s)
- Ji-Ae Ko
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | | | |
Collapse
|
216
|
Brzóska E, Grabowska I, Hoser G, Stremińska W, Wasilewska D, Machaj EK, Pojda Z, Moraczewski J, Kawiak J. Participation of stem cells from human cord blood in skeletal muscle regeneration of SCID mice. Exp Hematol 2006; 34:1262-70. [PMID: 16939819 DOI: 10.1016/j.exphem.2006.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2005] [Revised: 04/28/2006] [Accepted: 05/10/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE In this report, we demonstrate the participation of human cord blood (HUCB) stem cells in the skeletal muscle regeneration of SCID (severe combined immunodeficient) mice. MATERIALS AND METHODS The HUCB cells were labeled with the PKH26 fluorescent marker or recognized by an anti-HLA-ABC or anti-beta-2-microglobulin antibody. The HUCB cells were implanted directly into the damaged mouse muscle. The regeneration process and the implanted HUCB cells were traced each day after the damage, throughout a period of 7 days, and additionally at day 30 with the use of flow cytometry and confocal microscopy. RESULTS The PKH26-labeled cells isolated from the regenerating muscle were positive for the anti-HLA-ABC antibody. The percentage of the PKH26(+) and HLA-ABC(+) cells decreased from day 1 to day 5. In the regenerating muscle, the percentage of the HLA-ABC(+) cells increased, as measured on days 7 and 30. Moreover, myofibers containing fragments of the PKH26-labeled sarcolemma were noticed. Labeling with the anti-human beta(2)-microglobulin antibody showed the presence of positive cells and myofibers at day 7 of the regeneration, suggesting fusion of human and mouse cells. CONCLUSIONS We suggest that the HUCB cells implanted into the damaged muscle are present there for at least 30 days and that they participate in the muscle regeneration. Moreover, our study shows that the implanted HUCB cells form human muscle precursor cells residing in the repaired mouse muscle. We suggest that the HUCB cell circulation after transplantation depends on SDF-1 (stromal-derived factor-1) expression in regenerating muscle.
Collapse
Affiliation(s)
- Edyta Brzóska
- Department of Clinical Cytology, Medical Center of Postgraduate Education, Warsaw, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Contreras-Shannon V, Ochoa O, Reyes-Reyna SM, Sun D, Michalek JE, Kuziel WA, McManus LM, Shireman PK. Fat accumulation with altered inflammation and regeneration in skeletal muscle of CCR2-/- mice following ischemic injury. Am J Physiol Cell Physiol 2006; 292:C953-67. [PMID: 17020936 DOI: 10.1152/ajpcell.00154.2006] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chemokines recruit inflammatory cells to sites of injury, but the role of the CC chemokine receptor 2 (CCR2) during regenerative processes following ischemia is poorly understood. We studied injury, inflammation, perfusion, capillary formation, monocyte chemotactic protein-1 (MCP-1) levels, muscle regeneration, fat accumulation, and transcription factor activation in hindlimb muscles of CCR2-/- and wild-type (WT) mice following femoral artery excision (FAE). In both groups, muscle injury and restoration of vascular perfusion were similar. Nevertheless, edema and neutrophil accumulation were significantly elevated in CCR2-/- compared with WT mice at day 1 post-FAE and fewer macrophages were present at day 3. MCP-1 levels in post-ischemic calf muscle of CCR2-/- animals were significantly elevated over baseline through 14 days post-FAE and were higher than WT mice at days 1, 7, and 14. In addition, CCR2-/- mice exhibited impaired muscle regeneration, decreased muscle fiber size, and increased intermuscular adipocytes with similar capillaries/mm(2) postinjury. Finally, the transcription factors, MyoD and signal transducers of and activators of transcription-3 (STAT3), were significantly increased above baseline but did not differ significantly between groups at any time point post-FAE. These findings suggest that increases in MCP-1, and possibly, MyoD and STAT3, may modulate molecular signaling in CCR2-/- mice during inflammatory and regenerative events. Furthermore, alterations in neutrophil and macrophage recruitment in CCR2-/- mice may critically alter the normal progression of downstream regenerative events in injured skeletal muscle and may direct myogenic precursor cells in the regenerating milieu toward an adipogenic phenotype.
Collapse
|
218
|
Périé S, Mamchaoui K, Mouly V, Blot S, Bouazza B, Thornell LE, St Guily JL, Butler-Browne G. Premature proliferative arrest of cricopharyngeal myoblasts in oculo-pharyngeal muscular dystrophy: Therapeutic perspectives of autologous myoblast transplantation. Neuromuscul Disord 2006; 16:770-81. [PMID: 17005403 DOI: 10.1016/j.nmd.2006.07.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Revised: 05/19/2006] [Accepted: 07/05/2006] [Indexed: 11/29/2022]
Abstract
Cultures of myoblasts isolated from cricopharyngeal muscles from patients with oculopharyngeal muscular dystrophy (OPMD) have been performed to study the effect of the expanded (GCG)8-13 repeat, located on the poly(A) binding protein nuclear-1 (PABPN1), on satellite cell phenotype. Cell cultures exhibited a reduced myogenicity, as well as a rapid decrease in proliferative lifespan, as compared to controls. The incorporation of BrdU decreased during the proliferative lifespan, due to a progressive accumulation of non-dividing cells. A lower fusion index was also observed, but myoblasts were able to form large myotubes when OPMD cultures were purified, although a rapid loss of myogenicity during successive passages was also observed. Myoblasts isolated from unaffected muscles did not show the defects observed in cricopharyngeal muscle cultures. The PABPN1 was predominantly located in nuclei of myoblasts and in both the nuclei and cytoplasm of myotubes in OPMD cultures. In vivo analysis of OPMD muscles showed that the number of satellite cells was slightly higher than that observed in age matched controls. Mutation of the PABPN1 in OPMD provokes premature senescence in dividing myoblasts, that may be due to intranuclear toxic aggregates. These results suggest that myoblast autografts, isolated from unaffected muscles, and injected into the dystrophic pharyngeal muscles, may be a useful therapeutic strategy to restore muscular function. Its tolerance and feasibility has been preclinically demonstrated in the dog.
Collapse
Affiliation(s)
- Sophie Périé
- Inserm U787 Groupe Myologie and Institut de Myologie, Faculté de Médecine Pitié Salpêtrière, Université Paris VI Pierre et Marie Curie, 105, Boulevard de l'Hôpital, 75013 Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
219
|
Ohtake Y, Tojo H, Seiki M. Multifunctional roles of MT1-MMP in myofiber formation and morphostatic maintenance of skeletal muscle. J Cell Sci 2006; 119:3822-32. [PMID: 16926191 DOI: 10.1242/jcs.03158] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sequential activation of muscle-specific transcription factors is the critical basis for myogenic differentiation. However, the complexity of this process does not exclude the possibility that other molecules and systems are regulatory as well. We observed that myogenic differentiation proceeded through three distinct stages of proliferation, elongation and fusion, which are distinguishable by their cellular morphologies and gene expression patterns of proliferation- and differentiation-specific markers. Treatment of the differentiating myoblasts with inhibitors of matrix metalloproteinases (MMPs) revealed that MMP activity at the elongation stage is a critical prerequisite to complete the successive myoblast cell fusion. The MMP regulated the myogenic differentiation independently from the genetic program that governs expression of the myogenic genes. Membrane-type 1 matrix metalloproteinase (MT1-MMP) was identified as a major contributor to this checkpoint for morphological differentiation and degraded fibronectin, a possible inhibitory factor for myogenic cell fusion. A MT1-MMP deficiency caused similar myogenic impediments forming smaller myofibers in situ. Additionally, the mutant mice demonstrated some central nucleation of the myofibers typically found in muscular dystrophy and MT1-MMP was found to cleave laminin-2/4 in the basement membrane. Thus, MT1-MMP is a new multilateral regulator for muscle differentiation and maintenance through processing of stage-specific distinct ECM substrates.
Collapse
MESH Headings
- Animals
- Cell Differentiation/physiology
- Cell Fusion
- Cells, Cultured
- Fibronectins/metabolism
- Gene Expression Regulation
- Laminin/metabolism
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinases, Membrane-Associated/deficiency
- Matrix Metalloproteinases, Membrane-Associated/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Biological
- Muscle Development/physiology
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Myoblasts, Skeletal/cytology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Regeneration
- Tissue Inhibitor of Metalloproteinase-2/genetics
Collapse
Affiliation(s)
- Yohei Ohtake
- Division of Cancer Cell Research, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | | | | |
Collapse
|
220
|
Anderson JE. The satellite cell as a companion in skeletal muscle plasticity: currency, conveyance, clue, connector and colander. ACTA ACUST UNITED AC 2006; 209:2276-92. [PMID: 16731804 DOI: 10.1242/jeb.02088] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Satellite cells are companions to voluntary muscle fibres, and are named for their intimate positional or ;satellite' relationship, as if revolving around fibres, like a satellite moon around the earth. Studies on the nature of at least some satellite cells, including their capabilities for self-renewal and for giving rise to multiple lineages in a stem cell-like function, are exploring the molecular basis of phenotypes described by markers of specialized function and gene expression in normal development, neuromuscular disease and aging. In adult skeletal muscle, the self-renewing capacity of satellite cells contributes to muscle growth, adaptation and regeneration. Muscle remodeling, such as demonstrated by changes in myofibre cross-sectional area and length, nerve and tendon junctions, and fibre-type distribution, occur in the absence of injury and provide broad functional and structural diversity among skeletal muscles. Those contributions to plasticity involve the satellite cell in at least five distinct roles, here described using metaphors for behaviour or the investigator's perspective. Satellite cells are the 'currency' of muscle; have a 'conveyance' role in adaptation by domains of cytoplasm along a myofibre; serve researchers, through a marker role, as 'clues' to various activities of muscle; are 'connectors' that physically, and through signalling and cell-fibre communications, bridge myofibres to the intra- and extra-muscular environment; and are equipped as metabolic and genetic filters or 'colanders' that can rectify or modulate particular signals. While all these roles are still under exploration, each contributes to the plasticity of skeletal muscle and thence to the overall biology and function of an organism. The use of metaphor for describing these roles helps to clarify and scrutinize the definitions that form the basis of our understanding of satellite cell biology: the metaphors provide the construct for various approaches to detect or test the nature of satellite cell functions in skeletal muscle plasticity.
Collapse
Affiliation(s)
- Judy E Anderson
- Department of Human Anatomy and Cell Science, Faculty of Medicine, University of Manitoba, Winnipeg, MB, R3E 0W3, Canada.
| |
Collapse
|
221
|
Zhou Z, Cornelius CP, Eichner M, Bornemann A. Reinnervation-induced alterations in rat skeletal muscle. Neurobiol Dis 2006; 23:595-602. [PMID: 16877003 DOI: 10.1016/j.nbd.2006.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Revised: 05/09/2006] [Accepted: 05/18/2006] [Indexed: 11/19/2022] Open
Abstract
Denervation-induced myofiber atrophy can be reversed by reinnervation. Growing reinnervated myofibers upregulate numerous molecules, many of which determine the muscle fiber type. In the present study we aimed at identifying factors that might contribute specifically to myofiber growth after reinnervation. The common peroneal nerve of 15 male Wistar rats was cut and resutured without delay (9 animals) or with a delay of 4 weeks (6 animals). We studied the transcriptional repertoire of intact reinnervated tibialis anterior muscle by microarray gene analysis. We assessed SC activation by immunolabeling using anti-MyoD and -myogenin antibodies. The percentage of SC expressing MyoD reached up to 50% of M-cadherin+ cells whereas the percentage of SC expressing myogenin was normal (<10%) in all muscles examined. The values of ipsi- and contralateral muscles did not differ significantly from one another between right and left leg (p<0.05). Thirteen known genes were differentially regulated after reinnervation compared with contralateral muscles. Five of them determine the slow-twitch fiber type (four and a half LIM domains 3, cardiac beta-myosin heavy chain, calsequestrin 2, troponin C (slow), and heart myosin light chain), and three of them are neurally regulated (thrombospondin 4, transferrin receptor, cardiac ankyrin repeat protein). The results strengthen the notion that reinnervaton affects the molecular repertoire of the myofibers directly, leading to fiber type transformation and partial reversal of the denervation phenotype. By contrast, SC do not appear to be affected by reinnervation directly. They can be activated both in reinnervated and contralateral muscles, and they do not fully differentiate. This makes them unlikely to contribute to myofiber growth.
Collapse
Affiliation(s)
- Z Zhou
- Institute of Brain Research, University of Tübingen, Calwerstr. 3, D-72076 Tübingen, Germany
| | | | | | | |
Collapse
|
222
|
Abstract
Quiescent satellite cells are responsible for the repair of post-natal skeletal muscle. These cells are easily identified by their unique morphology within skeletal muscle as well as by several recently elucidated molecular markers. Careful examination of the function of these markers has provided insight into the early events surrounding satellite cell specification and activation. However, the origin of these cells, as well as the mechanisms by which this population is maintained within the adult remain elusive. Furthermore, the ability of non-muscle derived stem cells and the potential multipotency of satellite cells have altered the traditional views of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Chet E Holterman
- Ottawa Health Research Institute, Molecular Medicine Program, Ottawa, Ontario, Canada K1H 8L6
| | | |
Collapse
|
223
|
Chen Y, Lin G, Slack JMW. Control of muscle regeneration in the Xenopus tadpole tail by Pax7. Development 2006; 133:2303-13. [PMID: 16687446 DOI: 10.1242/dev.02397] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The tail of the Xenopus tadpole will regenerate completely after transection. Much of the mass of the regenerate is composed of skeletal muscle, but there has been some uncertainty about the source of the new myofibres. Here, we show that the growing tail contains many muscle satellite cells. They are active in DNA replication, whereas the myonuclei are not. As in mammals, the satellite cells express pax7. We show that a domain-swapped construct, pax7EnR, can antagonize pax7function. Transgenic tadpoles were prepared containing pax7EnR driven by a heat-inducible promoter. When induced, this reduces the proportion of satellite cells formed in the regenerate. A second amputation of the resulting tails yielded second regenerates containing notochord and spinal cord but little or no muscle. This shows that inhibition of pax7 action does not prevent differentiation of satellite cells to myofibres, but it does prevent their maintenance as a stem cell population.
Collapse
Affiliation(s)
- Ying Chen
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | | | | |
Collapse
|
224
|
Wozniak AC, Anderson JE. Single-fiber isolation and maintenance of satellite cell quiescence. Biochem Cell Biol 2006; 83:674-6. [PMID: 16234857 DOI: 10.1139/o05-046] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The activity of satellite cells during myogenesis, development, or skeletal muscle regeneration is strongly modelled using cultures of single muscle fibers. However, there are variations in reported features of gene or protein expression as examined with single-fiber cultures. Here, we examined the potential differences in activation of satellite cells on normal mouse muscle fibers produced during a standard isolation protocol, with or without agitation during collagenase digestion. Activation was detected in satellite cells on fibers after 24 and 48 h of culture in basal growth medium using immunodetection of the incorporation of bromodeoxyuridine (BrdU) into DNA and quantification of the number of BrdU-positive cells per fiber. After 24 and 48 h in culture under nonactivating conditions, the number of activated (BrdU+) satellite cells was greater on fibers that had received gentle agitation during collagenase digestion than on those that were subject to digestion without agitation during isolation. The findings are interpreted to mean that at least some of the variation among published reports may derive from the application of various methods of fiber isolation. The information should be useful for maintaining satellite cell quiescence during studies of the regulatory steps that lead to satellite cell activation.
Collapse
Affiliation(s)
- Ashley C Wozniak
- Department of HumanAnatomy and Cell Science, University of Manitoba,730 William Ave., Winnipeg, MB R3E 0W3, Canada
| | | |
Collapse
|
225
|
de Luna N, Gallardo E, Soriano M, Dominguez-Perles R, de la Torre C, Rojas-García R, García-Verdugo JM, Illa I. Absence of dysferlin alters myogenin expression and delays human muscle differentiation "in vitro". J Biol Chem 2006; 281:17092-17098. [PMID: 16608842 DOI: 10.1074/jbc.m601885200] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutations in dysferlin cause a type of muscular dystrophy known as dysferlinopathy. Dysferlin may be involved in muscle repair and differentiation. We compared normal human skeletal muscle cultures expressing dysferlin with muscle cultures from dysferlinopathy patients. We quantified the fusion index of myoblasts as a measure of muscle development and conducted optic and electronic microscopy, immunofluorescence, Western blot, flow cytometry, and real-time PCR at different developmental stages. Short interference RNA was used to corroborate the results obtained in dysferlin-deficient cultures. A luciferase reporter assay was performed to study myogenin activity in dysferlin-deficient cultures. Myoblasts fusion was consistently delayed as compared with controls whereas the proliferation rate did not change. Electron microscopy showed that control cultured cells at 10 days were fusiform, whereas dysferlin-deficient cells were star-shaped and large. After 15 days the normal multinucleated appearance and structured myofibrils were not present in dysferlin-deficient cells. Strikingly, myogenin was not detected in myotubes from dysferlin-deficient cultures using Western blot, and mRNA analysis showed low levels (p < 0.05) compared with controls. Flow cytometry and immunofluorescence also showed reduced levels of myogenin in dysferlin-deficient cultures. When the dysferlin gene was knocked down ( approximately 80%), myogenin mRNA leveled down to approximately 70%. MyoD and desmin mRNA levels in controls and dysferlin-deficient cultures were similar. The reporter luciferase assay demonstrated a low myogenin activity in dysferlin-deficient cultures. These results point to a functional link between dysferlin and myogenin, and both proteins may share a new signaling pathway involved in differentiation of skeletal muscle in vitro.
Collapse
Affiliation(s)
- Noemí de Luna
- Servei de Neurologia i Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| | - Eduard Gallardo
- Servei de Neurologia i Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| | - Mario Soriano
- Centro de Investigaciones Príncipe Felipe and Instituto Cavanilles, Universidad de Valencia, 46980 Valencia, Spain
| | - Raúl Dominguez-Perles
- Servei de Neurologia i Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| | - Carolina de la Torre
- Servei de Neurologia i Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| | - Ricardo Rojas-García
- Servei de Neurologia i Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| | - Jose M García-Verdugo
- Centro de Investigaciones Príncipe Felipe and Instituto Cavanilles, Universidad de Valencia, 46980 Valencia, Spain
| | - Isabel Illa
- Servei de Neurologia i Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain.
| |
Collapse
|
226
|
Maier A, Zhou Z, Bornemann A. The expression profile of myogenic transcription factors in satellite cells from denervated rat muscle. Brain Pathol 2006; 12:170-7. [PMID: 11958370 PMCID: PMC8095746 DOI: 10.1111/j.1750-3639.2002.tb00431.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The muscle-specific transcription factors of the MyoD family are altered after denervation. In order to determine whether this shift takes place in satellite cells (SC), we investigated the expression pattern of MyoD, myf5, myogenin, and MRF4 in SC. Hindlimb muscles of rats were denervated for 2 days to 4 weeks. SC were isolated, pooled and the transcription of all 4 factors was assessed by RT-PCR. Protein expression was assessed in histological sections of soleus and anterior tibial (TA) muscles; SC were identified by M-cadherin. Pooled SC from innervated muscles expressed myf5 mRNA, and very weakly MyoD and myogenin mRNA. MyoD and myogenin protein was found in only few SC. After denervation, pooled SC expressed myf5 mRNA, and very weakly myogenin and MRF4 mRNA. Myogenin protein was found in less than about 10% of the cells, whereas MRF4 protein was absent from SC. We conclude that the presence of myf5 and the absence of MyoD and MRF4 protein in SC after denervation indicated the quiescent state of the cell cycle. A subset of SC has additionally acquired myogenin. SC after denervation might be less easily recruited into the mitotic cycle than SC from normal muscle, rendering regeneration of denervated muscle less efficient than normal muscle.
Collapse
Affiliation(s)
- Annette Maier
- Institute of Brain Research, University of Tübingen, Germany
| | - Zhe Zhou
- Institute of Brain Research, University of Tübingen, Germany
| | - Antje Bornemann
- Institute of Brain Research, University of Tübingen, Germany
| |
Collapse
|
227
|
Wehrman TS, von Degenfeld G, Krutzik PO, Nolan GP, Blau HM. Luminescent imaging of β-galactosidase activity in living subjects using sequential reporter-enzyme luminescence. Nat Methods 2006; 3:295-301. [PMID: 16554835 DOI: 10.1038/nmeth868] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Accepted: 02/17/2006] [Indexed: 02/06/2023]
Abstract
We generated a sequential reporter-enzyme luminescence (SRL) technology for in vivo detection of beta-galactosidase (beta-gal) activity. The substrate, a caged D-luciferin-galactoside conjugate, must first be cleaved by beta-gal before it can be catalyzed by firefly luciferase (FLuc) to generate light. As a result, luminescence is dependent on beta-gal activity. Using this technology, constitutive beta-gal activity in engineered cells and inducible tissue-specific beta-gal expression in transgenic mice can now be visualized noninvasively over time. A substantial advantage of beta-gal as a bioluminescent probe is that the enzyme retains full activity outside of cells, unlike FLuc, which requires intracellular cofactors. As a result, antibodies conjugated to the recombinant beta-gal enzyme can be used to detect endogenous cells and extracellular antigens in vivo. Thus, coupling the properties of FLuc to the advantages of beta-gal permits bioluminescent imaging applications that previously were not possible.
Collapse
Affiliation(s)
- Thomas S Wehrman
- Baxter Laboratory in Genetic Pharmacology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | |
Collapse
|
228
|
Tsujimura T, Kinoshita M, Abe M. Response of rabbit skeletal muscle to tibial lengthening. J Orthop Sci 2006; 11:185-90. [PMID: 16568392 DOI: 10.1007/s00776-005-0991-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2005] [Accepted: 11/22/2005] [Indexed: 10/24/2022]
Abstract
BACKGROUND Experimental and clinical studies have provided knowledge regarding osteogenesis during limb lengthening. However, response of skeletal muscle to limb lengthening is not fully understood, especially as concerns histogenesis. We studied the morphological response of rabbit skeletal muscle to limb lengthening. In this study, we investigated proliferation of satellite cells, responsible for generation of new myonuclei, during limb lengthening. METHODS Tibialis anterior muscles of young and adult rabbits were subjected to lengthening at a rate of 0.5 mm twice per day for 20 days. After lengthening, muscle wet weight was measured to assess skeletal muscle growth, then proliferating cell nuclear antigen was measured. Immunostaining was performed to analyze proliferating cells in the proximal, middle, and distal portions of the muscle belly and the musculotendinous junction. RESULTS Muscle wet weight increased significantly after lengthening both in adult (0.4 g) and young (0.1 g) rabbits. Satellite cells showed proliferation in response to lengthening. In adult rabbits, satellite cell proliferation increased along the entire lengthened muscle to a similar degree (from 7.1% in the middle portion to 8.6% in the musculotendinous junction). In young rabbits, proliferation was greater in the musculotendinous junction (4.8%) than that in other muscle portions (2.3% in the middle and distal portions, and 2.4% in the proximal portion). In adult rabbits, the rate of increase in satellite cell proliferation was 1780% in the middle portion to 2860% in the musculotendinous junction, whereas the rate was between 210% in the middle portion and 290% in the distal portion in young rabbit. The rate of increase in cell proliferation by lengthening was higher in adult muscle than that in young muscles as well as satellite cell proliferation. CONCLUSION These findings indicate that limb lengthening promotes muscle growth in both young and adult rabbits.
Collapse
Affiliation(s)
- Tomoyuki Tsujimura
- Department of Orthopedic Surgery, Osaka Medical College, Takatsuki, Japan
| | | | | |
Collapse
|
229
|
Negroni E, Butler-Browne GS, Mouly V. Myogenic stem cells: regeneration and cell therapy in human skeletal muscle. ACTA ACUST UNITED AC 2006; 54:100-8. [PMID: 16246502 DOI: 10.1016/j.patbio.2005.09.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Accepted: 09/12/2005] [Indexed: 01/24/2023]
Abstract
Human skeletal muscle has been considered as an ideal target for cell-mediated therapy. However, the positive results obtained in dystrophic animal models using the resident precursor satellite cell population have been followed by discouraging evidences obtained in the clinical trials involving Duchenne muscular dystrophy patients. This text reviews the recent advances that many groups have achieved to identify from the stem cell compartment putative candidates for cell therapy. We focused our attention on stem cells with myogenic potential which might be able to improve transplantation efficiency and therefore could be used as a therapeutic tool for neuromuscular diseases.
Collapse
Affiliation(s)
- E Negroni
- Cytosquelette et Développement, FRE 2853, Faculté de Médecine Pitié-Salpétrière, Université Pierre et Marie Curie, 105, boulevard de l'Hôpital, 75013 Paris, France
| | | | | |
Collapse
|
230
|
Blazevich AJ, Sharp NCC. Understanding Muscle Architectural Adaptation: Macro- and Micro-Level Research. Cells Tissues Organs 2006; 181:1-10. [PMID: 16439814 DOI: 10.1159/000089964] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2005] [Indexed: 12/28/2022] Open
Abstract
Recent research using muscle-imaging techniques has revealed a remarkable plasticity of human muscle architecture where significant changes in fascicle lengths and angles have resulted from the chronic performance, or cessation, of strong muscle contractions. However, there is a paucity of data describing architectural adaptations to chronic stretching, disuse and immobilization, illness, and aging, and those data that are available are equivocal. Understanding their impact is important in order that effective interventions for illness/injury management and rehabilitation, and programs to improve the physical capacity of workers, the aged and athletes can be determined. Nonetheless, recent advances in myocellular research could provide a framework allowing the prediction of architectural changes in these understudied areas. Examination of the site-specific response to mechanical stress of calpain-dependent ubiquitin-proteasome proteolysis, or of the cellular response to stress after the knockout (or incapacitation) of sarcomeric and cytoskeletal proteins involved in cellular signal transduction, provides an exciting paradigm by which myocellular adaptation can be described. Such research might contribute to the understanding of macro-level changes in muscle architecture.
Collapse
Affiliation(s)
- Anthony J Blazevich
- Centre for Sports Medicine and Human Performance, Brunel University, Uxbridge UB8 3PH, UK
| | | |
Collapse
|
231
|
Brack AS, Bildsoe H, Hughes SM. Evidence that satellite cell decrement contributes to preferential decline in nuclear number from large fibres during murine age-related muscle atrophy. J Cell Sci 2006; 118:4813-21. [PMID: 16219688 DOI: 10.1242/jcs.02602] [Citation(s) in RCA: 207] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Skeletal muscle fibres are multinucleate syncitial cells that change size during adult life depending on functional demand. The relative contribution of change in nuclear number and/or cell growth to fibre size change is unclear. We report that nuclei/unit length decreases in larger fibres during skeletal muscle ageing. This leads to an increased size of nuclear domain (quantity of cytoplasm/number of nuclei within that cytoplasm). Initially, larger fibres have more satellite cells than small fibres, but this advantage is lost as satellite cells decline with age. These changes are accompanied by an overall decline in fibre size, returning domain size to the normal range. Exacerbated loss of fibre nuclei per unit length during ageing of myoD-null mice provides the first experimental support for the hypothesis that a satellite cell defect causes inadequate nuclear replacement. We propose a model in which a decline in satellite cell function and/or number during ageing leads to a loss of nuclei from large fibres and an associated domain size increase that triggers cytoplasmic atrophy through the normal cell-size-regulating machinery.
Collapse
Affiliation(s)
- Andrew S Brack
- MRC Centre for Developmental Neurobiology and Randall Division for Cell and Molecular Biophysics, New Hunt's House, King's College London, London, SE1 1UL, UK
| | | | | |
Collapse
|
232
|
Relaix F, Montarras D, Zaffran S, Gayraud-Morel B, Rocancourt D, Tajbakhsh S, Mansouri A, Cumano A, Buckingham M. Pax3 and Pax7 have distinct and overlapping functions in adult muscle progenitor cells. ACTA ACUST UNITED AC 2005; 172:91-102. [PMID: 16380438 PMCID: PMC2063537 DOI: 10.1083/jcb.200508044] [Citation(s) in RCA: 509] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The growth and repair of skeletal muscle after birth depends on satellite cells that are characterized by the expression of Pax7. We show that Pax3, the paralogue of Pax7, is also present in both quiescent and activated satellite cells in many skeletal muscles. Dominant-negative forms of both Pax3 and -7 repress MyoD, but do not interfere with the expression of the other myogenic determination factor, Myf5, which, together with Pax3/7, regulates the myogenic differentiation of these cells. In Pax7 mutants, satellite cells are progressively lost in both Pax3-expressing and -nonexpressing muscles. We show that this is caused by satellite cell death, with effects on the cell cycle. Manipulation of the dominant-negative forms of these factors in satellite cell cultures demonstrates that Pax3 cannot replace the antiapoptotic function of Pax7. These findings underline the importance of cell survival in controlling the stem cell populations of adult tissues and demonstrate a role for upstream factors in this context.
Collapse
Affiliation(s)
- Frédéric Relaix
- Unité de Génétique Moléculaire du Développement, Centre National de la Recherche Scientifique URA 2578, Département de Biologie du Développement
| | | | | | | | | | | | | | | | | |
Collapse
|
233
|
McCroskery S, Thomas M, Platt L, Hennebry A, Nishimura T, McLeay L, Sharma M, Kambadur R. Improved muscle healing through enhanced regeneration and reduced fibrosis in myostatin-null mice. J Cell Sci 2005; 118:3531-41. [PMID: 16079293 DOI: 10.1242/jcs.02482] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Numerous stimulatory growth factors that can influence muscle regeneration are known. Recently, it has been demonstrated that neutralization of muscle growth inhibitory factors, such as myostatin (Mstn; also known as growth differentiation factor 8, Gdf8), also leads to increased muscle regeneration in mdx mice that are known to have cycles of degeneration. However, the precise mechanism by which Mstn regulates muscle regeneration has not yet been fully determined. To investigate the role of Mstn in adult skeletal muscle regeneration, wild-type and myostatin-null (Mstn-/-) mice were injured with notexin. Forty-eight hours after injury, accelerated migration and enhanced accretion of myogenic cells (MyoD1+) and macrophages (Mac-1+) was observed at the site of regeneration in Mstn-/- muscle as compared with wild-type muscle. Inflammatory cell numbers decreased more rapidly in the Mstn-/- muscle, indicating that the whole process of inflammatory cell response is accelerated in Mstn-/- mice. Consistent with this result, the addition of recombinant Mstn reduced the activation of satellite cells (SCs) and chemotactic movements of both myoblasts and macrophages ex vivo. Examination of regenerated muscle (28 days after injury) also revealed that Mstn-/- mice showed increased expression of decorin mRNA, reduced fibrosis and improved healing as compared with wild-type mice. On the basis of these results, we propose that Mstn negatively regulates muscle regeneration not only by controlling SC activation but also by regulating the migration of myoblasts and macrophages to the site of injury. Thus, antagonists of Mstn could potentially be useful as pharmacological agents for the treatment of disorders of overt degeneration and regeneration.
Collapse
Affiliation(s)
- Seumas McCroskery
- Animal Genomics, AgResearch, Private Bag 3123, East Street, Hamilton, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
234
|
Barash IA, Mathew L, Lahey M, Greaser ML, Lieber RL. Muscle LIM protein plays both structural and functional roles in skeletal muscle. Am J Physiol Cell Physiol 2005; 289:C1312-20. [PMID: 16093282 DOI: 10.1152/ajpcell.00117.2005] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Muscle LIM protein (MLP) has been suggested to be an important mediator of mechanical stress in cardiac tissue, but the role that it plays in skeletal muscle remains unclear. Previous studies have shown that it is dramatically upregulated in fast-to-slow fiber-type transformation and also after eccentric contraction (EC)-induced muscle injury. The functional consequences of this upregulation, if any, are unclear. In the present study, we have examined the skeletal muscle phenotype of MLP-knockout (MLPKO) mice in terms of their response to EC-induced muscle injuries. The data suggest that while the MLPKO mice recover completely after EC-induced injury, their torque production lags behind that of heterozygous littermates in the early stages of the recovery process. This lag is accompanied by decreased expression of the muscle regulatory factor MyoD, suggesting that MLP may influence gene expression. In addition, there is evidence of type I fiber atrophy and a shorter resting sarcomere length in the MLPKO mice, but no significant differences in fiber type distribution. In summary, MLP appears to play a subtle role in the maintenance of normal muscle characteristics and in the early events of the recovery process of skeletal muscle to injury, serving both structural and gene-regulatory roles.
Collapse
Affiliation(s)
- Ilona A Barash
- Deptartment of Orthopaedics, Veterans Affairs Medical Center and Univ. of California, San Diego, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| | | | | | | | | |
Collapse
|
235
|
Goding GS, Al-Sharif KI, McLoon LK. Myonuclear addition to uninjured laryngeal myofibers in adult rabbits. Ann Otol Rhinol Laryngol 2005; 114:552-7. [PMID: 16134353 DOI: 10.1177/000348940511400711] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVES In normal mature limb skeletal muscle, satellite cells are quiescent and myonuclei do not divide after formation of their associated myofibers in the absence of injury. The possibility of myonuclear addition in uninjured laryngeal myofibers of adult rabbits was investigated in an immunohistochemical pilot study. METHODS Bromodeoxyuridine (brdU), a marker for cell division, was administered by intraperitoneal injection over a 12-hour period in rabbits. The number of brdU-positive myonuclei per myofiber was determined on cross sections through the thyroarytenoid (TA) and posterior cricoarytenoid (PCA) muscles. RESULTS In the TA muscle, 0.13% +/- 0.03% (mean +/- SEM) of the myofibers counted had a brdU-positive nucleus. In the PCA muscle, 0.13% +/- 0.01% of the myofibers counted had a brdU-positive nucleus. Approximately 0.2% and 0.3% of the myofibers of the TA and PCA muscles, respectively, had brdU-positive satellite cells associated with them. Tibialis anterior and pectoralis major muscle controls were negative for brdU-positive myonuclei. CONCLUSIONS These data support the possibility of continuous remodeling in uninjured adult laryngeal myofibers and accentuate the distinct nature of laryngeal muscle relative to limb skeletal muscle in the rabbit model.
Collapse
Affiliation(s)
- George S Goding
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
236
|
Ko JA, Gondo T, Inagaki S, Inui M. Requirement of the transmembrane semaphorin Sema4C for myogenic differentiation. FEBS Lett 2005; 579:2236-42. [PMID: 15811348 DOI: 10.1016/j.febslet.2005.03.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Revised: 03/03/2005] [Accepted: 03/03/2005] [Indexed: 12/28/2022]
Abstract
Semaphorins constitute a large family of signaling proteins that contribute to axonal guidance. Here we demonstrate that the transmembrane semaphorin Sema4C is up-regulated both in the early stage of differentiation of C2C12 mouse skeletal myoblasts into myotubes and during injury-induced muscle regeneration in vivo. Depletion of Sema4C in C2C12 cells resulted in marked attenuation of myotube formation. A fusion protein containing the extracellular Sema domain and a peptide corresponding to the intracellular COOH-terminal region of Sema4C each inhibited the differentiation of C2C12 cells. These findings indicate that Sema4C-mediated interaction among myoblasts plays an important role in terminal myogenic differentiation.
Collapse
Affiliation(s)
- Ji-Ae Ko
- Department of Pharmacology, Yamaguchi University School of Medicine, Yamaguchi University Hospital, Ube, Yamaguchi 755-8505, Japan
| | | | | | | |
Collapse
|
237
|
Sun L, Trausch-Azar JS, Ciechanover A, Schwartz AL. Ubiquitin-proteasome-mediated degradation, intracellular localization, and protein synthesis of MyoD and Id1 during muscle differentiation. J Biol Chem 2005; 280:26448-56. [PMID: 15888449 DOI: 10.1074/jbc.m500373200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mammalian skeletal myogenesis results in the differentiation of myoblasts to mature syncytial myotubes, a process regulated by an intricate genetic network of at least three protein families: muscle regulatory factors, E proteins, and Id proteins. MyoD, a key muscle regulatory factor, and its negative regulator Id1 have both been shown to be degraded by the ubiquitin-proteasome system. Using C2C12 cells and confocal fluorescence microscopy, we showed that MyoD and Id1 co-localize within the nucleus in proliferating myoblasts. In mature myotubes, in contrast, they reside in distinctive subcellular compartments, with MyoD within the nucleus and Id1 exclusively in the cytoplasm. Cellular abundance of Id1 was markedly diminished from the very onset of muscle differentiation, whereas MyoD abundance was reduced to a much lesser extent and only at the later stages of differentiation. These reductions in MyoD and Id1 protein levels seem to result from a change in the rate of protein synthesis rather than the rate of degradation. In vivo protein stability studies revealed that the rates of ubiquitin-proteasome-mediated MyoD and Id1 degradation are independent of myogenic differentiation state. Id1 and MyoD were both rapidly degraded, each with a t 1/2 approximately = 1 h in myoblasts and in myotubes. Furthermore, relative protein synthesis rates for MyoD and Id1 were significantly diminished during myoblast to myotube differentiation. These results provide insight as to the interaction between MyoD and Id1 in the process of muscle differentiation and have implications for the involvement of the ubiquitin-proteasome-mediated protein degradation and protein synthesis in muscle differentiation and metabolism under abnormal and pathological conditions.
Collapse
Affiliation(s)
- Liping Sun
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
238
|
Halevy O, Piestun Y, Allouh MZ, Rosser BWC, Rinkevich Y, Reshef R, Rozenboim I, Wleklinski-Lee M, Yablonka-Reuveni Z. Pattern of Pax7 expression during myogenesis in the posthatch chicken establishes a model for satellite cell differentiation and renewal. Dev Dyn 2005; 231:489-502. [PMID: 15390217 DOI: 10.1002/dvdy.20151] [Citation(s) in RCA: 239] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The paired-box transcription factor Pax7 plays a critical role in the specification of satellite cells in mouse skeletal muscle. In the present study, the position and number of Pax7-expressing cells found in muscles of growing and adult chickens confirm the presence of this protein in avian satellite cells. The expression pattern of Pax7 protein, along with the muscle regulatory proteins MyoD and myogenin, was additionally elucidated in myogenic cultures and in whole muscle from posthatch chickens. In cultures progressing from proliferation to differentiation, the expression of Pax7 in MyoD+ cells declined as the cells began expressing myogenin, suggesting Pax7 as an early marker for proliferating myoblasts. At all time points, some Pax7+ cells were negative for MyoD, resembling the reserve cell phenotype. Clonal analysis of muscle cell preparations demonstrated that single progenitors can give rise to both differentiating and reserve cells. In muscle tissues, Pax7 protein expression was the strongest by 1 day posthatch, declining on days 3 and 6 to a similar level. In contrast, myogenin expression peaked on day 3 and then dramatically declined. This finding was accompanied by a robust growth in fiber diameter between day 3 and 6. The distinctions in Pax7 and myogenin expression patterns, both in culture and in vivo, indicate that while some of the myoblasts differentiate and fuse into myofibers during early stages of posthatch growth, others retain their reserve cell capacity.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibodies, Monoclonal/immunology
- Biomarkers
- Cell Differentiation
- Cell Division
- Cell Lineage
- Cells, Cultured
- Chickens
- Clone Cells
- Fluorescent Antibody Technique, Direct
- Gene Expression Regulation, Developmental
- Homeodomain Proteins/immunology
- Homeodomain Proteins/metabolism
- Immunohistochemistry
- Models, Biological
- Muscle Development
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/immunology
- Muscle Fibers, Skeletal/physiology
- MyoD Protein/immunology
- MyoD Protein/metabolism
- Myoblasts/metabolism
- Myogenin/immunology
- Myogenin/metabolism
- PAX7 Transcription Factor
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/metabolism
- Time Factors
Collapse
Affiliation(s)
- Orna Halevy
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Wozniak AC, Kong J, Bock E, Pilipowicz O, Anderson JE. Signaling satellite-cell activation in skeletal muscle: markers, models, stretch, and potential alternate pathways. Muscle Nerve 2005; 31:283-300. [PMID: 15627266 DOI: 10.1002/mus.20263] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Activation of skeletal muscle satellite cells, defined as entry to the cell cycle from a quiescent state, is essential for normal growth and for regeneration of tissue damaged by injury or disease. This review focuses on early events of activation by signaling through nitric oxide and hepatocyte growth factor, and by mechanical stimuli. The impact of various model systems used to study activation and the regulation of satellite-cell quiescence are placed in the context of activation events in other tissues, concluding with a speculative model of alternate pathways signaling satellite-cell activation.
Collapse
Affiliation(s)
- Ashley C Wozniak
- Department of Human Anatomy and Cell Science, University of Manitoba, 730 William Avenue, Winnipeg, Manitoba R3E 0W2, Canada
| | | | | | | | | |
Collapse
|
240
|
Volonte D, Liu Y, Galbiati F. The modulation of caveolin-1 expression controls satellite cell activation during muscle repair. FASEB J 2005; 19:237-239. [PMID: 15545301 DOI: 10.1096/fj.04-2215fje] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We have previously shown that caveolin-1, the principal structural protein component of caveolar membrane domains, inhibits cellular proliferation and induces cell cycle arrest. We demonstrate here for the first time that caveolin-1 is expressed in satellite cells but not in mature muscle fibers. Satellite cells are quiescent myogenic precursors that, after muscle injury, become mitotically active, proliferate, and fuse together or, to existing myofibers, to form new muscle fibers. We show that down-regulation of caveolin-1 expression occurs in satellite cells/myogenic precursor cells (MPCs) during muscle regeneration and that hepatocyte growth factor, which is produced after muscle injury, down-regulates caveolin-1. We also demonstrate that down-regulation of endogenous caveolin-1 expression activates ERK and that activation of the p42/44 MAP kinase pathway is necessary to promote muscle regeneration. Finally, we show that overexpression of caveolin-1 inhibits muscle repair mechanisms both in vitro and in vivo. Taken together, these results propose caveolin-1 as a novel regulator of satellite cell functions and suggest that the following signaling pathway modulates satellite cell activation during muscle repair: injured fibers release HGF --> HGF down-regulates caveolin-1 protein expression --> down-regulation of caveolin-1 activates ERK --> activation of ERK promotes muscle repair by stimulating the proliferation and migration of MPCs toward the wounded area.
Collapse
MESH Headings
- Animals
- Caveolin 1
- Caveolins/biosynthesis
- Caveolins/physiology
- Cell Cycle/physiology
- Cell Differentiation/genetics
- Cell Line, Transformed
- Down-Regulation/genetics
- Down-Regulation/physiology
- Enzyme Activation/genetics
- Enzyme Activation/physiology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Hepatocyte Growth Factor/metabolism
- Mice
- Mice, Inbred C57BL
- Muscle Fibers, Skeletal/chemistry
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/chemistry
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myoblasts/chemistry
- Myoblasts/cytology
- Myoblasts/metabolism
- Regeneration/genetics
- Regeneration/physiology
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/physiology
- Wound Healing/genetics
Collapse
Affiliation(s)
- Daniela Volonte
- Department of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
241
|
Araya R, Eckardt D, Maxeiner S, Krüger O, Theis M, Willecke K, Sáez JC. Expression of connexins during differentiation and regeneration of skeletal muscle: functional relevance of connexin43. J Cell Sci 2004; 118:27-37. [PMID: 15601660 DOI: 10.1242/jcs.01553] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The molecular mechanisms regulating skeletal muscle regeneration and differentiation are not well understood. We analyzed the expression of connexins (Cxs) 40, 43 and 45 in normal and regenerating tibialis anterior muscle and in primary cultures of differentiating myoblasts in adult and newborn mice, respectively. Cxs 45 and 43, but not 40, were strongly expressed in normal muscle and their expression was upregulated during regeneration. Furthermore, the functional role of Cx43 during differentiation and regeneration was examined after induced deletion of Cx43 in transgenic mice. In vivo, the inducible deletion of Cx43 delayed the formation of myofibers and prolonged the expression of myogenin during regeneration. In primary cultures of satellite cell-derived myoblasts, induced deletion of Cx43 led to decreased expression of myogenin and MyoD, dye coupling, creatine kinase activity and myoblast fusion. Thus, the expression of Cx45 and Cx43 is upregulated during skeletal muscle regeneration and Cx43 is required for normal myogenesis in vitro and adult muscle regeneration in vivo.
Collapse
Affiliation(s)
- Roberto Araya
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | | | | | | | | | | |
Collapse
|
242
|
Santa María L, Rojas CV, Minguell JJ. Signals from damaged but not undamaged skeletal muscle induce myogenic differentiation of rat bone-marrow-derived mesenchymal stem cells. Exp Cell Res 2004; 300:418-26. [PMID: 15475006 DOI: 10.1016/j.yexcr.2004.07.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2004] [Revised: 07/01/2004] [Indexed: 10/26/2022]
Abstract
The regenerative capacity of skeletal muscle has been usually attributed to resident satellite cells, which, upon activation by local or distant stimuli, initiate a myogenic differentiation program. Although recent studies have revealed that bone-marrow-derived progenitor cells may also participate in regenerative myogenesis, the signals and mechanisms involved in this process have not been elucidated. This study was designed to investigate whether signals from injured rat skeletal muscle were competent to induce a program of myogenic differentiation in expanded cultures of rat bone-marrow-derived mesenchymal stem cells (MSC). We observed that the incubation of MSC with a conditioned medium prepared from chemically damaged but not undamaged muscle resulted in a time-dependent change from fibroblast-like into elongated multinucleated cells, a transient increase in the number of MyoD positive cells, and the subsequent onset of myogenin, alpha-actinin, and myosin heavy chain expression. These results show that damaged rat skeletal muscle is endowed with the capacity to induce myogenic differentiation of bone-marrow-derived mesenchymal progenitors.
Collapse
Affiliation(s)
- Lorena Santa María
- Programa de Terapias Celulares, INTA, Universidad de Chile, Santiago, Chile.
| | | | | |
Collapse
|
243
|
Seale P, Ishibashi J, Holterman C, Rudnicki MA. Muscle satellite cell-specific genes identified by genetic profiling of MyoD-deficient myogenic cell. Dev Biol 2004; 275:287-300. [PMID: 15501219 DOI: 10.1016/j.ydbio.2004.07.034] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Revised: 07/09/2004] [Accepted: 07/13/2004] [Indexed: 12/13/2022]
Abstract
Satellite cells are committed myogenic progenitors that give rise to proliferating myoblasts during postnatal growth and repair of skeletal muscle. To identify genes expressed at different developmental stages in the satellite cell myogenic program, representational difference analysis of cDNAs was employed to identify more than 50 unique mRNAs expressed in wild-type myoblasts and MyoD-/- myogenic cells. Novel expression patterns for several genes, such as Pax7, Asb5, IgSF4, and Hoxc10, were identified that were expressed in both quiescent and activated satellite cells. Several previously uncharacterized genes that represent putative MyoD target genes were also identified, including Pw1, Dapk2, Sytl2, and NLRR1. Importantly, many genes such as IgSF4, Neuritin, and Klra18 that were expressed exclusively in MyoD-/- myoblasts were also expressed by satellite cells in undamaged muscle in vivo but were not expressed by primary myoblasts. These data are consistent with a biological role for activated satellite cells that induce Myf5 but not MyoD. Lastly, additional endothelial and hematopoietic markers were identified supporting a nonsomitic developmental origin of the satellite cell myogenic lineage.
Collapse
Affiliation(s)
- Patrick Seale
- Department of Biology, McMaster University, Hamilton, Ontario, Canada L8S 4K1
| | | | | | | |
Collapse
|
244
|
Shefer G, Wleklinski-Lee M, Yablonka-Reuveni Z. Skeletal muscle satellite cells can spontaneously enter an alternative mesenchymal pathway. J Cell Sci 2004; 117:5393-404. [PMID: 15466890 DOI: 10.1242/jcs.01419] [Citation(s) in RCA: 235] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We show that muscle satellite cells, traditionally considered as committed myogenic precursors, are comprised of Pax7-expressing progenitors that preserve a mesenchymal repertoire extending beyond a mere myogenic potential. Mouse satellite cells from freshly isolated single myofibers, cultured individually in serum-rich growth medium, produced myogenic and non-myogenic clones. Only the myogenic clones expressed muscle-specific transcription factors and formed myotubes. Pax7 was initially expressed in all clones, but subsequently was associated only with the myogenic clones. Some cells in the non-myogenic clones expressed alpha-smooth muscle actin and nestin whereas others differentiated into mature adipocytes. This type of cell composition mirrors characteristics of mesenchymal stem cell progeny. Overall, individual myofibers persistently gave rise to both clonal phenotypes, but the ratio of myogenic to non-myogenic clones randomly varied among fibers. This randomness indicates that clonal dichotomy reflects satellite cell suppleness rather than pre-fated cell heterogeneity. We conclude that satellite cells possess mesenchymal plasticity, being able to commit either to myogenesis or to a mesenchymal alternative differentiation (MAD) program.
Collapse
Affiliation(s)
- Gabi Shefer
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|
245
|
Jacquemin V, Furling D, Bigot A, Butler-Browne GS, Mouly V. IGF-1 induces human myotube hypertrophy by increasing cell recruitment. Exp Cell Res 2004; 299:148-58. [PMID: 15302582 DOI: 10.1016/j.yexcr.2004.05.023] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2004] [Revised: 05/13/2004] [Indexed: 10/26/2022]
Abstract
Insulin-like growth factor-1 (IGF-1) has been shown in rodents (i) in vivo to induce muscle fiber hypertrophy and to prevent muscle mass decline with age and (ii) in vitro to enhance the proliferative life span of myoblasts and to induce myotube hypertrophy. In this study, performed on human primary cultures, we have shown that IGF-1 has very little effect on the proliferative life span of human myoblasts but does delay replicative senescence. IGF-1 also induces hypertrophy of human myotubes in vitro, as characterized by an increase in the mean number of nuclei per myotube, an increase in the fusion index, and an increase in myosin heavy chain (MyHC) content. In addition, muscle hypertrophy can be triggered in the absence of proliferation by recruiting more mononucleated cells. We propose that IGF-1-induced hypertrophy can involve the recruitment of reserve cells in human skeletal muscle.
Collapse
Affiliation(s)
- V Jacquemin
- CNRS UMR 7000 Cytosquelette et Développement, Paris, France
| | | | | | | | | |
Collapse
|
246
|
Zammit PS, Carvajal JJ, Golding JP, Morgan JE, Summerbell D, Zolnerciks J, Partridge TA, Rigby PWJ, Beauchamp JR. Myf5 expression in satellite cells and spindles in adult muscle is controlled by separate genetic elements. Dev Biol 2004; 273:454-65. [PMID: 15328025 DOI: 10.1016/j.ydbio.2004.05.038] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2003] [Revised: 05/05/2004] [Accepted: 05/21/2004] [Indexed: 02/01/2023]
Abstract
The myogenic regulatory factor Myf5 is integral to the initiation and control of skeletal muscle formation. In adult muscle, Myf5 is expressed in satellite cells, stem cells of mature muscle, but not in the myonuclei that sustain the myofibre. Using the Myf5(nlacZ/+) mouse, we now show that Myf5 is also constitutively expressed in muscle spindles-stretch-sensitive mechanoreceptors, while muscle denervation induces extensive reactivation of the Myf5 gene in myonuclei. To identify the elements involved in the regulation of Myf5 in adult muscle, we analysed reporter gene expression in a transgenic bacterial artificial chromosome (BAC) deletion series of the Mrf4/Myf5 locus. A BAC carrying 140 kb upstream of the Myf5 transcription start site was sufficient to drive all aspects of Myf5 expression in adult muscle. In contrast, BACs carrying 88 and 59 kb upstream were unable to drive consistent expression in satellite cells, although expression in muscle spindles and reactivation of the locus in myonuclei were retained. Therefore, as during development, multiple enhancers are required to generate the full expression pattern of Myf5 in the adult. Together, these observations show that elements controlling adult Myf5 expression are genetically separable and possibly distinct from those that control Myf5 during development. These studies are a first step towards identifying cognate transcription factors involved in muscle stem cell regulation.
Collapse
Affiliation(s)
- Peter S Zammit
- Muscle Cell Biology Group, Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, W12 0NN, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Raynaud F, Carnac G, Marcilhac A, Benyamin Y. m-Calpain implication in cell cycle during muscle precursor cell activation. Exp Cell Res 2004; 298:48-57. [PMID: 15242761 DOI: 10.1016/j.yexcr.2004.03.053] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2003] [Revised: 03/24/2004] [Indexed: 11/28/2022]
Abstract
Milli-calpain, a member of the ubiquitous cysteine protease family, is known to control late events of cell-cell fusion in skeletal muscle tissue through its involvement in cell membrane and cytoskeleton component reorganization. In this report, we describe the characterization of m-calpain compartmentalization and activation during the initial steps of muscle precursor cell recruitment and differentiation. By immunofluorescence analysis, we show that m-calpain is present throughout the cell cycle in the nucleus of proliferating myoblast C2 cells. However, when myoblasts enter a quiescent/G0 stage, m-calpain staining is detected only in the cytoplasm. Moreover, comparison of healthy and injured muscle shows distinct m-calpain localization in satellite stem cells. Indeed, m-calpain is not found in quiescent satellite cells, but following muscle injury, when satellite cells start to proliferate, m-calpain appears in the nucleus. To determine the implication of m-calpain during the cell cycle progression, quiescent myoblasts were forced to re-enter the cell cycle in the presence or not of the specific calpain inhibitor MDL 28170. We demonstrate that this calpain inhibitor blocks the cell cycle, prevents accumulation of MyoD in the G1 phase and enhances Myf5 expression. These data support an important new role for m-calpain in the control of muscle precursor cell activation and thus suggest its possible implication during the initial events of muscle regeneration.
Collapse
Affiliation(s)
- F Raynaud
- UMR 5539-CNRS, Laboratoire de Motilité Cellulaire, EPHE, cc107, University of Montpellier 2 place Eugène Bataillon, 34090 France.
| | | | | | | |
Collapse
|
248
|
Sartorelli V, Fulco M. Molecular and cellular determinants of skeletal muscle atrophy and hypertrophy. Sci Signal 2004; 2004:re11. [PMID: 15292521 DOI: 10.1126/stke.2442004re11] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The maintenance of adult skeletal muscle mass is ensured by physical exercise. Accordingly, physiological and pathological situations characterized by either impaired motor neuron activity, reduced gravity (microgravity during space flights), or reduced physical activity result in loss of muscle mass. Furthermore, a plethora of clinical conditions, including cancer, sepsis, diabetes, and AIDS, are associated with varying degrees of muscle atrophy. The cellular and molecular pathways responsible for maintaining the skeletal muscle mass are not well defined. Nonetheless, studies aimed at the understanding of the mechanisms underlying either muscular atrophy or hypertrophy have begun to identify the physiological determinants and clarify the molecular pathways responsible for the maintenance of muscle mass.
Collapse
Affiliation(s)
- Vittorio Sartorelli
- Muscle Gene Expression Group Laboratory of Muscle Biology, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
249
|
Zammit PS, Golding JP, Nagata Y, Hudon V, Partridge TA, Beauchamp JR. Muscle satellite cells adopt divergent fates: a mechanism for self-renewal? ACTA ACUST UNITED AC 2004; 166:347-57. [PMID: 15277541 PMCID: PMC2172269 DOI: 10.1083/jcb.200312007] [Citation(s) in RCA: 671] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Growth, repair, and regeneration of adult skeletal muscle depends on the persistence of satellite cells: muscle stem cells resident beneath the basal lamina that surrounds each myofiber. However, how the satellite cell compartment is maintained is unclear. Here, we use cultured myofibers to model muscle regeneration and show that satellite cells adopt divergent fates. Quiescent satellite cells are synchronously activated to coexpress the transcription factors Pax7 and MyoD. Most then proliferate, down-regulate Pax7, and differentiate. In contrast, other proliferating cells maintain Pax7 but lose MyoD and withdraw from immediate differentiation. These cells are typically located in clusters, together with Pax7−ve progeny destined for differentiation. Some of the Pax7+ve/MyoD−ve cells then leave the cell cycle, thus regaining the quiescent satellite cell phenotype. Significantly, noncycling cells contained within a cluster can be stimulated to proliferate again. These observations suggest that satellite cells either differentiate or switch from terminal myogenesis to maintain the satellite cell pool.
Collapse
Affiliation(s)
- Peter S Zammit
- Muscle Cell Biology Group, Medical Research Council Clinical Sciences Centre, Faculty of Medicine, Imperial College, Hammersmith Hospital Campus, Du Cane Rd., London W12 0NN, UK.
| | | | | | | | | | | |
Collapse
|
250
|
Abstract
The tail of the Xenopus tadpole will regenerate following amputation, and all three of the main axial structures – the spinal cord, the notochord and the segmented myotomes – are found in the regenerated tail. We have investigated the cellular origin of each of these three tissue types during regeneration.We produced Xenopus laevis embryos transgenic for the CMV (Simian Cytomegalovirus) promoter driving GFP (Green Fluorescent Protein) ubiquitously throughout the embryo. Single tissues were then specifically labelled by making grafts at the neurula stage from transgenic donors to unlabelled hosts. When the hosts have developed to tadpoles, they carry a region of the appropriate tissue labelled with GFP. These tails were amputated through the labelled region and the distribution of labelled cells in the regenerate was followed. We also labelled myofibres using the Cre-lox method.The results show that the spinal cord and the notochord regenerate from the same tissue type in the stump, with no labelling of other tissues. In the case of the muscle, we show that the myofibres of the regenerate arise from satellite cells and not from the pre-existing myofibres. This shows that metaplasia between differentiated cell types does not occur, and that the process of Xenopus tail regeneration is more akin to tissue renewal in mammals than to urodele tail regeneration.
Collapse
Affiliation(s)
- Cesare Gargioli
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK
| | | |
Collapse
|