Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biol Res ; 56(1): 21, 2023 May 05.
Article in English | MEDLINE | ID: mdl-37147738

ABSTRACT

BACKGROUND: Satellite cells are tissue-specific stem cells primarily responsible for the regenerative capacity of skeletal muscle. Satellite cell function and maintenance are regulated by extrinsic and intrinsic mechanisms, including the ubiquitin-proteasome system, which is key for maintaining protein homeostasis. In this context, it has been shown that ubiquitin-ligase NEDD4-1 targets the transcription factor PAX7 for proteasome-dependent degradation, promoting muscle differentiation in vitro. Nonetheless, whether NEDD4-1 is required for satellite cell function in regenerating muscle remains to be determined. RESULTS: Using conditional gene ablation, we show that NEDD4-1 loss, specifically in the satellite cell population, impairs muscle regeneration resulting in a significant reduction of whole-muscle size. At the cellular level, NEDD4-1-null muscle progenitors exhibit a significant decrease in the ability to proliferate and differentiate, contributing to the formation of myofibers with reduced diameter. CONCLUSIONS: These results indicate that NEDD4-1 expression is critical for proper muscle regeneration in vivo and suggest that it may control satellite cell function at multiple levels.


Subject(s)
Muscle, Skeletal , Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Cell Proliferation/physiology , Muscle, Skeletal/metabolism , Stem Cells , Cell Differentiation , Ubiquitins/metabolism , Muscle Development/physiology , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism
2.
Biol. Res ; 56: 21-21, 2023. ilus, graf
Article in English | LILACS | ID: biblio-1513734

ABSTRACT

BACKGROUND: Satellite cells are tissue-specific stem cells primarily responsible for the regenerative capacity of skeletal muscle. Satellite cell function and maintenance are regulated by extrinsic and intrinsic mechanisms, including the ubiquitin-proteasome system, which is key for maintaining protein homeostasis. In this context, it has been shown that ubiquitin-ligase NEDD4-1 targets the transcription factor PAX7 for proteasome-dependent degradation, promoting muscle differentiation in vitro. Nonetheless, whether NEDD4-1 is required for satellite cell function in regenerating muscle remains to be determined. RESULTS: Using conditional gene ablation, we show that NEDD4-1 loss, specifically in the satellite cell population, impairs muscle regeneration resulting in a significant reduction of whole-muscle size. At the cellular level, NEDD4-1-null muscle progenitors exhibit a significant decrease in the ability to proliferate and differentiate, contributing to the formation of myofibers with reduced diameter. CONCLUSIONS: These results indicate that NEDD4-1 expression is critical for proper muscle regeneration in vivo and suggest that it may control satellite cell function at multiple levels.


Subject(s)
Muscle, Skeletal/metabolism , Proteasome Endopeptidase Complex/metabolism , Stem Cells , Ubiquitins/metabolism , Cell Differentiation , Muscle Development/physiology , Cell Proliferation/physiology , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism
3.
Front Cell Dev Biol ; 9: 821839, 2021.
Article in English | MEDLINE | ID: mdl-35127730

ABSTRACT

In recent years, the ubiquitin-proteasome system (UPS) has emerged as an important regulator of stem cell function. Here we review recent findings indicating that UPS also plays critical roles in the biology of satellite cells, the muscle stem cell responsible for its maintenance and regeneration. While we focus our attention on the control of key transcriptional regulators of satellite cell function, we briefly discuss early studies suggesting the UPS participates more broadly in the regulation of satellite cell stemness and regenerative capacity.

4.
J Biomed Mater Res A ; 105(8): 2241-2251, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28426898

ABSTRACT

Polymer scaffolds are used as an alternative to support tissue regeneration when it does not occur on its own. Cell response on polymer scaffolds is determined by factors such as polymer composition, topology, and the presence of other molecules. We evaluated the cellular response of murine skeletal muscle myoblasts on aligned or unaligned fibers obtained by electrospinning poly(ε-caprolactone) (PCL), and blends with poly(lactic-co-glycolic acid) (PLGA) or decorin, a proteoglycan known to regulate myogenesis. The results showed that aligned PCL fibers with higher content of PLGA promote cell growth and improve the quality of differentiation with PLGA scaffolds having the highest confluence at over 68% of coverage per field of view for myoblasts and more than 7% of coverage for myotubes. At the same time, the addition of decorin greatly improves the quantity and quality of differentiated cells in terms of cell fusion, myotube length and thickness, being 71, 10, and 51% greater than without the protein, respectively. Interestingly, our results suggest that at certain concentrations, the effect of decorin on myoblast differentiation exceeds the topological effect of fiber alignment. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2241-2251, 2017.


Subject(s)
Myoblasts/cytology , Polyesters/chemistry , Tissue Scaffolds/chemistry , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Decorin/chemistry , Lactic Acid/chemistry , Mice , Muscle Development , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
5.
J Biomed Mater Res A ; 105(1): 118-130, 2017 01.
Article in English | MEDLINE | ID: mdl-27593555

ABSTRACT

Satellite cells are a small cell population that function as muscle-specific adult stem cells. When muscle damage occurs, these cells are able to activate, proliferate, and ultimately fuse with each other in order to form new myofibers or fuse with existing ones. For tissue engineering applications, obtaining a sufficient number of myoblasts prior transplantation that maintains their regenerative capacity is critical. This can be obtained by in vitro expansion of autologous satellite cells. However, once plated, the self-renewal and regenerative capacity of myoblasts is rapidly lost, obtaining low yields per biopsy. For this purpose, we evaluated in vitro culture of the murine myoblast cell line C2C12 and mouse primary myoblasts with chitosan and chitosan/poly-octanoic acid 2-thiophen-3-yl-ethyl ester blends (poly(OTE)). The films of chitosan/poly(OTE) blends were heterogeneous and slightly rougher than chitosan and poly(OTE) films. Poly(OTE) presence improved myoblast adhesion in both cell types and prevented complete differentiation, but maintaining their differentiation potential in vitro. We identified that the polymer blend chitosan/poly(OTE) could be a suitable substrate to culture satellite cells/myoblasts in vitro preventing differentiation prior transplantation. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 118-130, 2017.


Subject(s)
Chitosan/pharmacology , Myoblasts/physiology , Polyesters/pharmacology , Regeneration/drug effects , Tissue Scaffolds/chemistry , Animals , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Chitosan/chemistry , Mice , Myoblasts/cytology , Polyesters/chemistry
6.
PLoS One ; 11(5): e0154919, 2016.
Article in English | MEDLINE | ID: mdl-27144531

ABSTRACT

Skeletal muscle regeneration and long term maintenance is directly link to the balance between self-renewal and differentiation of resident adult stem cells known as satellite cells. In turn, satellite cell fate is influenced by a functional interaction between the transcription factor Pax7 and members of the MyoD family of muscle regulatory factors. Thus, changes in the Pax7-to-MyoD protein ratio may act as a molecular rheostat fine-tuning acquisition of lineage identity while preventing precocious terminal differentiation. Pax7 is expressed in quiescent and proliferating satellite cells, while its levels decrease sharply in differentiating progenitors Pax7 is maintained in cells (re)acquiring quiescence. While the mechanisms regulating Pax7 levels based on differentiation status are not well understood, we have recently described that Pax7 levels are directly regulated by the ubiquitin-ligase Nedd4, thus promoting proteasome-dependent Pax7 degradation in differentiating satellite cells. Here we show that Pax7 levels are maintained in proliferating muscle progenitors by a mechanism involving casein kinase 2-dependent Pax7 phosphorylation at S201. Point mutations preventing S201 phosphorylation or casein kinase 2 inhibition result in decreased Pax7 protein in proliferating muscle progenitors. Accordingly, this correlates directly with increased Pax7 ubiquitination. Finally, Pax7 down regulation induced by casein kinase 2 inhibition results in precocious myogenic induction, indicating early commitment to terminal differentiation. These observations highlight the critical role of post translational regulation of Pax7 as a molecular switch controlling muscle progenitor fate.


Subject(s)
Casein Kinase II/metabolism , Cell Proliferation/physiology , Muscle Development/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , PAX7 Transcription Factor/metabolism , Phosphorylation/physiology , Animals , Cell Differentiation/physiology , Cell Line , Down-Regulation/physiology , Mice , MyoD Protein/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/physiology , Ubiquitination/physiology
7.
Cell Tissue Res ; 364(3): 647-660, 2016 06.
Article in English | MEDLINE | ID: mdl-26742767

ABSTRACT

Fibrosis occurs in skeletal muscle under various pathophysiological conditions such as Duchenne muscular dystrophy (DMD), a devastating disease characterized by fiber degeneration that results in progressive loss of muscle mass, weakness and increased extracellular matrix (ECM) accumulation. Fibrosis is also observed after skeletal muscle denervation and repeated cycles of damage followed by regeneration. The ECM is synthesized largely by fibroblasts in the muscle connective tissue under normal conditions. Myofibroblasts, cells that express α-smooth muscle actin (α-SMA), play a role in many tissues affected by fibrosis. In skeletal muscle, fibro/adipogenic progenitors (FAPs) that express cell-surface platelet-derived growth factor receptor-α (PDGFR-α) and the transcription factor Tcf4 seem to be responsible for connective tissue synthesis and are good candidates for the origin of myofibroblasts. We show that cells positive for Tcf4 and PDGFR-α are expressed in skeletal muscle under normal conditions and are increased in various skeletal muscles of mdx mice, a murine model for DMD, wild type muscle after sciatic denervation and muscle subjected to chronic damage. These cells co-label with the myofibroblast marker α-SMA in dystrophic muscle but not in normal tissue. The Tcf4-positive cells lie near macrophages mainly concentrated in dystrophic necrotic-regenerating foci. The close proximity of Tcf4-positive cells to inflammatory cells and their previously described role in muscle regeneration might reflect an active interaction between these cell types and growth factors, possibly resulting in a muscular regenerative or fibrotic condition.


Subject(s)
Adipogenesis , Biomarkers/metabolism , Cell Differentiation , Connective Tissue/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myofibroblasts/pathology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Count , Chronic Disease , Denervation , Fibrosis , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Muscle, Skeletal/innervation , Muscular Dystrophy, Animal/pathology , Necrosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Transcription Factor 4 , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Stem Cells ; 33(10): 3138-51, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26304770

ABSTRACT

The transcription factor Pax7 regulates skeletal muscle stem cell (satellite cells) specification and maintenance through various mechanisms, including repressing the activity of the muscle regulatory factor MyoD. Hence, Pax7-to-MyoD protein ratios can determine maintenance of the committed-undifferentiated state or activation of the differentiation program. Pax7 expression decreases sharply in differentiating myoblasts but is maintained in cells (re)acquiring quiescence, yet the mechanisms regulating Pax7 levels based on differentiation status are not well understood. Here we show that Pax7 levels are directly regulated by the ubiquitin-ligase Nedd4. Our results indicate that Nedd4 is expressed in quiescent and activated satellite cells, that Nedd4 and Pax7 physically interact during early muscle differentiation-correlating with Pax7 ubiquitination and decline-and that Nedd4 loss of function prevented this effect. Furthermore, even transient nuclear accumulation of Nedd4 induced a drop in Pax7 levels and precocious muscle differentiation. Consequently, we propose that Nedd4 functions as a novel Pax7 regulator, which activity is temporally and spatially controlled to modulate the Pax7 protein levels and therefore satellite cell fate.


Subject(s)
Cell Differentiation/genetics , Endosomal Sorting Complexes Required for Transport/biosynthesis , Muscle Development , PAX7 Transcription Factor/biosynthesis , Satellite Cells, Skeletal Muscle/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Animals , Cell Proliferation/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Gene Expression Regulation, Developmental , Humans , Mice , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , MyoD Protein/biosynthesis , Nedd4 Ubiquitin Protein Ligases , PAX7 Transcription Factor/genetics , Proteasome Endopeptidase Complex/genetics , Satellite Cells, Skeletal Muscle/cytology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
J Cell Mol Med ; 16(5): 1013-25, 2012 May.
Article in English | MEDLINE | ID: mdl-21615681

ABSTRACT

Post-natal growth and regeneration of skeletal muscle is highly dependent on a population of resident myogenic precursors known as satellite cells. Transcription factors from the Pax gene family, Pax3 and Pax7, are critical for satellite cell biogenesis, survival and potentially self-renewal; however, the underlying molecular mechanisms remain unsolved. This is particularly true in the case of Pax7, which appears to regulate myogenesis at multiple levels. Accordingly, recent data have highlighted the importance of a functional relationship between Pax7 and the MyoD family of muscle regulatory transcription factors during normal muscle formation and disease. Here we will critically review key findings suggesting that Pax7 may play a dual role by promoting resident muscle progenitors to commit to the skeletal muscle lineage while preventing terminal differentiation, thus keeping muscle progenitors poised to differentiate upon environmental cues. In addition, potential regulatory mechanisms for the control of Pax7 activity will be proposed.


Subject(s)
Muscle Development/physiology , PAX7 Transcription Factor/physiology , Satellite Cells, Skeletal Muscle/physiology , Animals , Gene Expression Regulation, Developmental/physiology , Humans , Mice , Muscle, Skeletal/growth & development , Muscle, Skeletal/physiology , Myoblasts/physiology , Protein Processing, Post-Translational/physiology , Rats , Receptors, Notch/physiology , Syndecans/physiology , TGF-beta Superfamily Proteins/physiology , Tumor Necrosis Factor-alpha/physiology , Wnt Signaling Pathway/physiology
10.
J Cell Biochem ; 112(5): 1410-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21321994

ABSTRACT

Alveolar rhabdomyosarcoma (ARMS) are characterized by the expression of chimeric transcription factors Pax3-FKHR and Pax7-FKHR, due to chromosomal translocations fusing PAX3 or PAX7 with the FKHR gene. Although ARMS exhibits a muscle lineage phenotype, the cells evade terminal differentiation despite expressing the potent myogenic transcriptional regulator MyoD. Here we show that while Pax7-FKHR inhibits MyoD-dependent transcription, MyoD enhances Pax7-FKHR activity in myogenic cell cultures. Importantly, this effect is not recapitulated by close related transcription factor myogenin and involves specific MyoD functional domains, distinct from those required for Pax7 to regulate MyoD during muscle formation. Together, these results suggest that although repressed as a myogenic regulatory factor, MyoD can play an active role in ARMS by augmenting Pax7-FKHR function.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , MyoD Protein/metabolism , Neoplasms, Muscle Tissue/genetics , Oncogene Proteins, Fusion/metabolism , PAX7 Transcription Factor/metabolism , Rhabdomyosarcoma, Alveolar/genetics , Forkhead Box Protein O1 , Humans , Muscle Development/genetics , MyoD Protein/genetics , Myogenin/genetics , Myogenin/metabolism , Phenotype , Transcription, Genetic
11.
Biol Res ; 44(4): 323-7, 2011.
Article in English | MEDLINE | ID: mdl-22446594

ABSTRACT

The transcription factor Pax7 negatively regulates the activity of the muscle regulatory transcription factor MyoD, preventing muscle precursor cells from undergoing terminal differentiation. In this context, the ratio between Pax7 and MyoD protein levels is thought to be critical in allowing myogenesis to proceed or to maintain the undifferentiated muscle precursor state. We have previously shown that Pax7 is subject to rapid down regulation in differentiating myoblasts, via a proteasome-dependent pathway. Here we present evidence indicating that Pax7 is also subject to caspase-3-dependent regulation. Furthermore, simultaneous inhibition of caspase-3 and proteasome activity induced further accumulation of Pax7 protein in differentiating myoblasts. These results suggest that at early stages of muscle differentiation, Pax7 levels are regulated by at least two independent mechanisms involving caspase-3 and proteasome activity.


Subject(s)
Caspase 3/physiology , Cell Differentiation/physiology , Muscle Development/physiology , MyoD Protein/metabolism , Myoblasts, Skeletal/physiology , PAX7 Transcription Factor/metabolism , Proteasome Endopeptidase Complex/physiology , Animals , Down-Regulation , Horses , Myoblasts, Skeletal/enzymology
12.
Biol. Res ; 44(4): 323-327, 2011. ilus
Article in English | LILACS | ID: lil-626730

ABSTRACT

The transcription factor Pax7 negatively regulates the activity of the muscle regulatory transcription factor MyoD, preventing muscle precursor cells from undergoing terminal differentiation. In this context, the ratio between Pax7 and MyoD protein levels is thought to be critical in allowing myogenesis to proceed or to maintain the undifferentiated muscle precursor state. We have previously shown that Pax7 is subject to rapid down regulation in differentiating myoblasts, via a proteasome-dependent pathway. Here we present evidence indicating that Pax7 is also subject to caspase-3-dependent regulation. Furthermore, simultaneous inhibition of caspase-3 and proteasome activity induced further accumulation of Pax7 protein in differentiating myoblasts. These results suggest that at early stages of muscle differentiation, Pax7 levels are regulated by at least two independent mechanisms involving caspase-3 and proteasome activity.


Subject(s)
Animals , /physiology , Cell Differentiation/physiology , Muscle Development/physiology , MyoD Protein/metabolism , Myoblasts, Skeletal/physiology , /metabolism , Proteasome Endopeptidase Complex/physiology , Down-Regulation , Horses , Myoblasts, Skeletal/enzymology
13.
J Cell Biol ; 190(3): 427-41, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20696709

ABSTRACT

Skeletal muscle postnatal growth and repair depend on satellite cells and are regulated by molecular signals within the satellite cell niche. We investigated the molecular and cellular events that lead to altered myogenesis upon genetic ablation of Syndecan-3, a component of the satellite cell niche. In the absence of Syndecan-3, satellite cells stall in S phase, leading to reduced proliferation, increased cell death, delayed onset of differentiation, and markedly reduced numbers of Pax7(+) satellite cells accompanied by myofiber hypertrophy and an increased number of centrally nucleated myofibers. We show that the aberrant cell cycle and impaired self-renewal of explanted Syndecan-3-null satellite cells are rescued by ectopic expression of the constitutively active Notch intracellular domain. Furthermore, we show that Syndecan-3 interacts with Notch and is required for Notch processing by ADAM17/tumor necrosis factor-alpha-converting enzyme (TACE) and signal transduction. Together, our data support the conclusion that Syndecan-3 and Notch cooperate in regulating homeostasis of the satellite cell population and myofiber size.


Subject(s)
Muscle Development , Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Syndecan-3/metabolism , Animals , Cell Cycle , Cell Differentiation , Cell Membrane/enzymology , Cell Membrane/metabolism , Cell Proliferation , Cells, Cultured , Mice , Mice, Inbred Strains , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction , Syndecan-3/deficiency , Syndecan-3/genetics
14.
J Cell Biol ; 177(5): 769-79, 2007 Jun 04.
Article in English | MEDLINE | ID: mdl-17548510

ABSTRACT

Postnatal growth and regeneration of skeletal muscle requires a population of resident myogenic precursors named satellite cells. The transcription factor Pax7 is critical for satellite cell biogenesis and survival and has been also implicated in satellite cell self-renewal; however, the underlying molecular mechanisms remain unclear. Previously, we showed that Pax7 overexpression in adult primary myoblasts down-regulates MyoD and prevents myogenin induction, inhibiting myogenesis. We show that Pax7 prevents muscle differentiation independently of its transcriptional activity, affecting MyoD function. Conversely, myogenin directly affects Pax7 expression and may be critical for Pax7 down-regulation in differentiating cells. Our results provide evidence for a cross-inhibitory interaction between Pax7 and members of the muscle regulatory factor family. This could represent an additional mechanism for the control of satellite cell fate decisions resulting in proliferation, differentiation, and self-renewal, necessary for skeletal muscle maintenance and repair.


Subject(s)
Cell Differentiation/physiology , MyoD Protein/physiology , PAX7 Transcription Factor/physiology , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Line , Cell Proliferation , Gene Expression Regulation , Mice , Muscle Development/physiology , MyoD Protein/antagonists & inhibitors , Myogenin/physiology , PAX7 Transcription Factor/antagonists & inhibitors , PAX7 Transcription Factor/chemistry , Protein Interaction Mapping , Protein Structure, Tertiary
15.
Dev Biol ; 275(2): 375-88, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15501225

ABSTRACT

Satellite cells are myogenic precursors responsible for skeletal muscle regeneration. Satellite cells are absent in the Pax-7-/- mouse, suggesting that this transcription factor is crucial for satellite cell specification [Seale, P., Sabourin, L.A., Girgis-Gabardo, A., Mansouri, A., Gruss, P., Rudnicki, M.A., 2000. Pax7 is required for the specification of myogenic satellite cells. Cell 102, 777-786]. Analysis of Pax-7 expression in activated satellite cells unexpectedly revealed substantial heterogeneity within individual clones. Further analyses show that Pax-7 and myogenin expression are mutually exclusive during differentiation, where Pax-7 appears to be up-regulated in cells that escape differentiation and exit the cell cycle, suggesting a regulatory relationship between these two transcription factors. Indeed, overexpression of Pax-7 down-regulates MyoD, prevents myogenin induction, and blocks MyoD-induced myogenic conversion of 10T1/2 cells. Overexpression of Pax-7 also promotes cell cycle exit even in proliferation conditions. Together, these results suggest that Pax-7 may play a crucial role in allowing activated satellite cells to reacquire a quiescent, undifferentiated state. These data support the concept that satellite cell self-renewal may be a primary mechanism for replenishment of the satellite cell compartment during skeletal muscle regeneration.


Subject(s)
Cell Cycle/physiology , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Models, Biological , Muscle Development/physiology , Satellite Cells, Skeletal Muscle/physiology , Animals , Blotting, Western , Cell Line , Fluorescent Antibody Technique , Homeodomain Proteins/physiology , Mice , Mice, Mutant Strains , Myoblasts , PAX7 Transcription Factor , Transfection
16.
Dev Biol ; 259(2): 209-24, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12871697

ABSTRACT

During limb skeletal muscle formation, committed muscle cells proliferate and differentiate in the presence of extracellular signals that stimulate or repress each process. Proteoglycans are extracellular matrix organizers and modulators of growth factor activities, regulating muscle differentiation in vitro. Previously, we characterized proteoglycan expression during early limb muscle formation and showed a spatiotemporal relation between the onset of myogenesis and the expression of decorin, an important muscle extracellular matrix component and potent regulator of TGF-beta activity. To evaluate decorin's role during in vivo differentiation in committed muscle cells, we grafted wild type and decorin-null myoblasts onto chick limb buds. The absence of decorin enhanced the migration and distribution of myoblasts in the limb, correlating with the inhibition of skeletal muscle differentiation. Both phenotypes were reverted by de novo decorin expression. In vitro, we determined that both decorin core protein and its glycosaminoglycan chain were required to reverse the migration phenotype. Results presented here suggest that the enhanced migration observed in decorin-null myoblasts may not be dependent on chemotactic growth factor signaling nor the differentiation status of the cells. Decorin may be involved in the establishment and/or coordination of a critical myoblast density, through inhibition of migration, that permits normal muscle differentiation during embryonic myogenesis.


Subject(s)
Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Proteoglycans/physiology , Animals , Cell Differentiation , Cell Line , Cell Movement , Cell Transplantation , Chick Embryo , Coculture Techniques , Decorin , Extracellular Matrix Proteins , Gene Expression Regulation, Developmental , Genetic Vectors , Limb Buds/cytology , Limb Buds/growth & development , Mice , Muscle, Skeletal/cytology , Myoblasts, Skeletal/cytology , Myogenin/metabolism , Retroviridae/genetics , Transforming Growth Factor beta/metabolism , Transplantation, Heterologous , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...