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










Publication year range
1.
Development ; 145(23)2018 11 26.
Article in English | MEDLINE | ID: mdl-30478226

ABSTRACT

Cell fate decisions occur through the action of multiple factors, including signalling molecules and transcription factors. Recently, the regulation of translation has emerged as an important step for modulating cellular function and fate, as exemplified by ribosomes that play distinct roles in regulating cell behaviour. Notchless (Nle) is a conserved nuclear protein that is involved in a crucial step in ribosome biogenesis, and is required for the maintenance of adult haematopoietic and intestinal stem/progenitor cells. Here, we show that activated skeletal muscle satellite cells in conditional Nle mutant mice are arrested in proliferation; however, deletion of Nle in myofibres does not impair myogenesis. Furthermore, conditional deletion of Nle in satellite cells during homeostasis did not impact on their fate for up to 3 months. In contrast, loss of Nle function in primary myogenic cells blocked proliferation because of major defects in ribosome formation. Taken together, we show that muscle stem cells undergo a stage-specific regulation of ribosome biogenesis, thereby underscoring the importance of differential modulation of mRNA translation for controlling cell fate decisions.


Subject(s)
Cell Lineage , Membrane Proteins/metabolism , Muscle Development , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Organelle Biogenesis , Ribosomes/metabolism , Animals , Cell Cycle , Cell Differentiation , Cells, Cultured , Cyclin E/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Mice, Knockout , Mutation/genetics , Myoblasts/cytology , Myoblasts/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism
2.
Front Physiol ; 9: 515, 2018.
Article in English | MEDLINE | ID: mdl-29881353

ABSTRACT

Degenerative myopathies typically display a decline in satellite cells coupled with a replacement of muscle fibers by fat and fibrosis. During this pathological remodeling, satellite cells are present at lower numbers and do not display a proper regenerative function. Whether a decline in satellite cells directly contributes to disease progression or is a secondary result is unknown. In order to dissect these processes, we used a genetic model to reduce the satellite cell population by ~70-80% which leads to a nearly complete loss of regenerative potential. We observe that while no overt tissue damage is observed following satellite cell depletion, muscle fibers atrophy accompanied by changes in the stem cell niche cellular composition. Treatment of these mice with an Activin receptor type-2B (AcvR2B) pathway blocker reverses muscle fiber atrophy as expected, but also restores regenerative potential of the remaining satellite cells. These findings demonstrate that in addition to controlling fiber size, the AcvR2B pathway acts to regulate the muscle stem cell niche providing a more favorable environment for muscle regeneration.

3.
Development ; 145(6)2018 03 19.
Article in English | MEDLINE | ID: mdl-29555813

ABSTRACT

Body skeletal muscles derive from the paraxial mesoderm, which forms in the posterior region of the embryo. Using microarrays, we characterize novel mouse presomitic mesoderm (PSM) markers and show that, unlike the abrupt transcriptome reorganization of the PSM, neural tube differentiation is accompanied by progressive transcriptome changes. The early paraxial mesoderm differentiation stages can be efficiently recapitulated in vitro using mouse and human pluripotent stem cells. While Wnt activation alone can induce posterior PSM markers, acquisition of a committed PSM fate and efficient differentiation into anterior PSM Pax3+ identity further requires BMP inhibition to prevent progenitors from drifting to a lateral plate mesoderm fate. When transplanted into injured adult muscle, these precursors generated large numbers of immature muscle fibers. Furthermore, exposing these mouse PSM-like cells to a brief FGF inhibition step followed by culture in horse serum-containing medium allows efficient recapitulation of the myogenic program to generate myotubes and associated Pax7+ cells. This protocol results in improved in vitro differentiation and maturation of mouse muscle fibers over serum-free protocols and enables the study of myogenic cell fusion and satellite cell differentiation.


Subject(s)
Cell Differentiation/genetics , Mesoderm/cytology , Muscle Development/genetics , Muscle, Skeletal/cytology , Pluripotent Stem Cells/cytology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Humans , Immunohistochemistry , Immunophenotyping , In Situ Hybridization , In Vitro Techniques , Mesoderm/metabolism , Mesoderm/physiology , Mice , Muscle Development/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Wnt Signaling Pathway/genetics
4.
Sci Rep ; 8(1): 4208, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523801

ABSTRACT

Skeletal muscle satellite cells are quiescent adult resident stem cells that activate, proliferate and differentiate to generate myofibres following injury. They harbour a robust proliferation potential and self-renewing capacity enabling lifelong muscle regeneration. Although several classes of microRNAs were shown to regulate adult myogenesis, systematic examination of stage-specific microRNAs during lineage progression from the quiescent state is lacking. Here we provide a genome-wide assessment of the expression of small RNAs during the quiescence/activation transition and differentiation by RNA-sequencing. We show that the majority of small RNAs present in quiescent, activated and differentiated muscle cells belong to the microRNA class. Furthermore, by comparing expression in distinct cell states, we report a massive and dynamic regulation of microRNAs, both in numbers and amplitude, highlighting their pivotal role in regulation of quiescence, activation and differentiation. We also identify a number of microRNAs with reliable and specific expression in quiescence including several maternally-expressed miRNAs generated at the imprinted Dlk1-Dio3 locus. Unexpectedly, the majority of class-switching miRNAs are associated with the quiescence/activation transition suggesting a poised program that is actively repressed. These data constitute a key resource for functional analyses of miRNAs in skeletal myogenesis, and more broadly, in the regulation of stem cell self-renewal and tissue homeostasis.


Subject(s)
Cell Lineage/genetics , MicroRNAs/genetics , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Sequence Analysis, RNA , Animals , Cell Self Renewal/genetics , Chromosomes, Mammalian/genetics , Gene Expression Profiling , Homeostasis/genetics , Mice , Muscle Development , Regeneration
5.
Methods Mol Biol ; 1556: 23-39, 2017.
Article in English | MEDLINE | ID: mdl-28247343

ABSTRACT

Isolation of muscle stem cells from skeletal muscle is a critical step for the study of skeletal myogenesis and regeneration. Although stem cell isolation has been performed for decades, the emergence of flow cytometry with defined cell surface markers, or transgenic mouse models, has allowed the efficient isolation of highly enriched stem cell populations. Here, we describe the isolation of mouse muscle stem cells using two different combinations of enzyme treatments allowing the release of mononucleated muscle stem cells from their niche. Mouse muscle stem cells can be further isolated as a highly enriched population by flow cytometry using fluorescent reporters or cell surface markers. We will present advantages and drawbacks of these different approaches.


Subject(s)
Cell Separation/methods , Muscle, Skeletal/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Biomarkers , Flow Cytometry/methods , Immunophenotyping/methods , Mice , Phenotype , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism
6.
PLoS One ; 11(1): e0147198, 2016.
Article in English | MEDLINE | ID: mdl-26807982

ABSTRACT

BACKGROUND: A longstanding goal in regenerative medicine is to reconstitute functional tissues or organs after injury or disease. Attention has focused on the identification and relative contribution of tissue specific stem cells to the regeneration process. Relatively little is known about how the physiological process is regulated by other tissue constituents. Numerous injury models are used to investigate tissue regeneration, however, these models are often poorly understood. Specifically, for skeletal muscle regeneration several models are reported in the literature, yet the relative impact on muscle physiology and the distinct cells types have not been extensively characterised. METHODS: We have used transgenic Tg:Pax7nGFP and Flk1GFP/+ mouse models to respectively count the number of muscle stem (satellite) cells (SC) and number/shape of vessels by confocal microscopy. We performed histological and immunostainings to assess the differences in the key regeneration steps. Infiltration of immune cells, chemokines and cytokines production was assessed in vivo by Luminex®. RESULTS: We compared the 4 most commonly used injury models i.e. freeze injury (FI), barium chloride (BaCl2), notexin (NTX) and cardiotoxin (CTX). The FI was the most damaging. In this model, up to 96% of the SCs are destroyed with their surrounding environment (basal lamina and vasculature) leaving a "dead zone" devoid of viable cells. The regeneration process itself is fulfilled in all 4 models with virtually no fibrosis 28 days post-injury, except in the FI model. Inflammatory cells return to basal levels in the CTX, BaCl2 but still significantly high 1-month post-injury in the FI and NTX models. Interestingly the number of SC returned to normal only in the FI, 1-month post-injury, with SCs that are still cycling up to 3-months after the induction of the injury in the other models. CONCLUSIONS: Our studies show that the nature of the injury model should be chosen carefully depending on the experimental design and desired outcome. Although in all models the muscle regenerates completely, the trajectories of the regenerative process vary considerably. Furthermore, we show that histological parameters are not wholly sufficient to declare that regeneration is complete as molecular alterations (e.g. cycling SCs, cytokines) could have a major persistent impact.


Subject(s)
Models, Animal , Muscle, Skeletal/physiology , Regeneration , Animals , Barium Compounds/toxicity , Chlorides/toxicity , Cobra Cardiotoxin Proteins/toxicity , Cold Injury/pathology , Cold Injury/physiopathology , Cytokines/physiology , Elapid Venoms/toxicity , Fibrosis , Freezing/adverse effects , Green Fluorescent Proteins/analysis , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Development , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Myoblasts/physiology , Necrosis , Neovascularization, Physiologic , Regeneration/immunology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/physiology , Stem Cells/physiology , Vascular Endothelial Growth Factor Receptor-2/analysis
7.
Nat Biotechnol ; 33(9): 962-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26237517

ABSTRACT

During embryonic development, skeletal muscles arise from somites, which derive from the presomitic mesoderm (PSM). Using PSM development as a guide, we establish conditions for the differentiation of monolayer cultures of mouse embryonic stem (ES) cells into PSM-like cells without the introduction of transgenes or cell sorting. We show that primary and secondary skeletal myogenesis can be recapitulated in vitro from the PSM-like cells, providing an efficient, serum-free protocol for the generation of striated, contractile fibers from mouse and human pluripotent cells. The mouse ES cells also differentiate into Pax7(+) cells with satellite cell characteristics, including the ability to form dystrophin(+) fibers when grafted into muscles of dystrophin-deficient mdx mice, a model of Duchenne muscular dystrophy (DMD). Fibers derived from ES cells of mdx mice exhibit an abnormal branched phenotype resembling that described in vivo, thus providing an attractive model to study the origin of the pathological defects associated with DMD.


Subject(s)
Cell Differentiation , Disease Models, Animal , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Pluripotent Stem Cells/pathology , Animals , Cells, Cultured , Mice , Mice, Transgenic
8.
J Cell Sci ; 127(Pt 17): 3840-51, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25002399

ABSTRACT

The midbody remnant (MBR) that is generated after cytokinetic abscission has recently attracted a lot of attention, because it might have crucial consequences for cell differentiation and tumorigenesis in mammalian cells. In these cells, it has been reported that the MBR is either released into the extracellular medium or retracted into one of the two daughter cells where it can be degraded by autophagy. Here, we describe a major alternative pathway in a variety of human and mouse immortalized cells, cancer cells and primary stem cells. Using correlative light and scanning electron microscopy and quantitative assays, we found that sequential abscissions on both sides of the midbody generate free MBRs, which are tightly associated with the cell surface through a Ca(2+)/Mg(2+)-dependent receptor. Surprisingly, MBRs move over the cell surface for several hours, before being eventually engulfed by an actin-dependent phagocytosis-like mechanism. Mathematical modeling combined with experimentation further demonstrates that lysosomal activities fully account for the clearance of MBRs after engulfment. This study changes our understanding of how MBRs are inherited and degraded in mammalian cells and suggests a mechanism by which MBRs might signal over long distances between cells.


Subject(s)
Cell Membrane/metabolism , Cytokinesis/physiology , Microtubules/metabolism , Organelles/metabolism , Animals , Cell Line , Cell Membrane/ultrastructure , HeLa Cells/cytology , Humans , Microscopy, Electrochemical, Scanning , Microtubules/ultrastructure , Organelles/ultrastructure , Phagocytosis/physiology
9.
J Cell Sci ; 125(Pt 7): 1738-49, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22366456

ABSTRACT

Skeletal muscle stem cell fate in adult mice is regulated by crucial transcription factors, including the determination genes Myf5 and Myod. The precise role of Myf5 in regulating quiescent muscle stem cells has remained elusive. Here we show that most, but not all, quiescent satellite cells express Myf5 protein, but at varying levels, and that resident Myf5 heterozygous muscle stem cells are more primed for myogenic commitment compared with wild-type satellite cells. Paradoxically however, heterotypic transplantation of Myf5 heterozygous cells into regenerating muscles results in higher self-renewal capacity compared with wild-type stem cells, whereas myofibre regenerative capacity is not altered. By contrast, Pax7 haploinsufficiency does not show major modifications by transcriptome analysis. These observations provide a mechanism linking Myf5 levels to muscle stem cell heterogeneity and fate by exposing two distinct and opposing phenotypes associated with Myf5 haploinsufficiency. These findings have important implications for how stem cell fates can be modulated by crucial transcription factors while generating a pool of responsive heterogeneous cells.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Haploinsufficiency/genetics , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Myogenic Regulatory Factor 5/genetics , Animals , Cell Lineage , Mice , Muscle, Skeletal/cytology , Myogenic Regulatory Factor 5/deficiency , Myogenic Regulatory Factor 5/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Phenotype
10.
Cell ; 148(1-2): 112-25, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22265406

ABSTRACT

Satellite cells are adult skeletal muscle stem cells that are quiescent and constitute a poorly defined heterogeneous population. Using transgenic Tg:Pax7-nGFP mice, we show that Pax7-nGFP(Hi) cells are less primed for commitment and have a lower metabolic status and delayed first mitosis compared to Pax7-nGFP(Lo) cells. Pax7-nGFP(Hi) can give rise to Pax7-nGFP(Lo) cells after serial transplantations. Proliferating Pax7-nGFP(Hi) cells exhibit lower metabolic activity, and the majority performs asymmetric DNA segregation during cell division, wherein daughter cells retaining template DNA strands express stem cell markers. Using chromosome orientation-fluorescence in situ hybridization, we demonstrate that all chromatids segregate asymmetrically, whereas Pax7-nGFP(Lo) cells perform random DNA segregation. Therefore, quiescent Pax7-nGFP(Hi) cells represent a reversible dormant stem cell state, and during muscle regeneration, Pax7-nGFP(Hi) cells generate distinct daughter cell fates by asymmetrically segregating template DNA strands to the stem cell. These findings provide major insights into the biology of stem cells that segregate DNA asymmetrically.


Subject(s)
Adult Stem Cells/cytology , Chromosome Segregation , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Division , Female , Mice , Mice, Transgenic , PAX7 Transcription Factor/metabolism , Templates, Genetic
11.
Development ; 138(17): 3647-56, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21828093

ABSTRACT

Distinct cell populations with regenerative capacity have been reported to contribute to myofibres after skeletal muscle injury, including non-satellite cells as well as myogenic satellite cells. However, the relative contribution of these distinct cell types to skeletal muscle repair and homeostasis and the identity of adult muscle stem cells remain unknown. We generated a model for the conditional depletion of satellite cells by expressing a human diphtheria toxin receptor under control of the murine Pax7 locus. Intramuscular injection of diphtheria toxin during muscle homeostasis, or combined with muscle injury caused by myotoxins or exercise, led to a marked loss of muscle tissue and failure to regenerate skeletal muscle. Moreover, the muscle tissue became infiltrated by inflammatory cells and adipocytes. This localised loss of satellite cells was not compensated for endogenously by other cell types, but muscle regeneration was rescued after transplantation of adult Pax7(+) satellite cells alone. These findings indicate that other cell types with regenerative potential depend on the presence of the satellite cell population, and these observations have important implications for myopathic conditions and stem cell-based therapeutic approaches.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , PAX7 Transcription Factor/metabolism , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Diphtheria Toxin/pharmacology , Female , Flow Cytometry , Immunohistochemistry , Male , Mice , Muscle, Skeletal/drug effects , PAX7 Transcription Factor/genetics , Regeneration/drug effects , Reverse Transcriptase Polymerase Chain Reaction
12.
Stem Cells ; 27(11): 2769-80, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19785007

ABSTRACT

Multiple cell types arise from cells in the dermomyotome of the somite that express Pax3 and Pax7, and myogenesis is regulated by Notch signaling. The asymmetric cell fate determinant Numb is thought to promote differentiation of skeletal muscle and other lineages by negatively regulating Notch signaling. We used transgenesis to overexpress Numb spatiotemporally in Pax3(+)/Pax7(+) somitic stem and progenitor cells in mouse embryos using a spatiotemporally regulated enhancer element from the Myf5 locus that can target muscle progenitor cells prior to cell commitment. Molecular analyses as well as examination of dermal and skeletal muscle cell fates in vivo show that although Numb is thought to be associated with muscle differentiation, unexpectedly the common stem/progenitor pool size for these lineages is increased in Numb-transgenic embryos. Prospective isolation of the relevant transgenic cells and analysis by quantitative reverse-transcription polymerase chain reaction demonstrated that, in this context, canonical Notch targets are not significantly downregulated. These findings were corroborated using a Notch reporter mouse during the formation of somites and prior to lineage segregation. Thus, we propose that Numb can regulate the self-renewal of dermal and muscle progenitors during a lineage progression.


Subject(s)
Membrane Proteins/physiology , Muscle Fibers, Skeletal/cytology , Nerve Tissue Proteins/physiology , Somites/cytology , Stem Cells/cytology , Animals , Blotting, Western , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mitosis/genetics , Mitosis/physiology , Muscle Development/genetics , Muscle Development/physiology , Muscle Fibers, Skeletal/enzymology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
13.
Dev Cell ; 16(6): 810-21, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19531352

ABSTRACT

Genetic regulatory networks governing skeletal myogenesis in the body are well understood, yet their hierarchical relationships in the head remain unresolved. We show that either Myf5 or Mrf4 is necessary for initiating extraocular myogenesis. Whereas Mrf4 is dispensable for pharyngeal muscle progenitor fate, Tbx1 and Myf5 act synergistically for governing myogenesis in this location. As in the body, Myod acts epistatically to the initiating cascades in the head. Thus, complementary pathways, governed by Pax3 for body, and Tbx1 for pharyngeal muscles, but absent for extraocular muscles, activate the core myogenic network. These diverse muscle progenitors maintain their respective embryonic regulatory signatures in the adult. However, these signatures are not sufficient to ensure the specific muscle phenotypes, since the expected differentiated phenotype is not manifested when satellite cells are engrafted heterotopically. These findings identify novel genetic networks that may provide insights into myopathies which often affect only subsets of muscles.


Subject(s)
Branchial Region/cytology , Cell Lineage , Eye/cytology , Gene Regulatory Networks , Muscles/cytology , Stem Cells/cytology , Animals , Branchial Region/metabolism , Cell Survival , Eye/metabolism , Eye/transplantation , Gene Expression Regulation, Developmental , Head , Mice , Muscle Development , Muscles/metabolism , Mutation/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Phenotype , Satellite Cells, Skeletal Muscle/cytology , Somites/cytology , Somites/metabolism , Stem Cell Transplantation , Stem Cells/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transplantation, Heterotopic
14.
Regen Med ; 4(2): 293-319, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19317647

ABSTRACT

Tissue development and regeneration share common features, since modules of regulatory pathways and transcription factors that are crucial for prenatal development are redeployed for tissue reconstruction after trauma. Regenerative medicine has therefore gained important insights through the study of developmental and regenerative biology. Moreover, diverse experimental models have been used to investigate the regeneration process in different tissues and organs. Paradoxically, little is known regarding the relative contribution of stem cells with respect to the supporting tissue during tissue regeneration. Particular attention will be given to mouse models using distinct injury paradigms to investigate the regenerative biology of skeletal muscle. An understanding of the response of stem and parenchymal cells is crucial for the development of clinical strategies to combat the normal decline in tissue performance during aging or its reconstitution after trauma and during disease. This review addresses these issues, focusing on muscle regeneration and how different factors, including genes, cells and the environment, impinge on this process.


Subject(s)
Muscle, Skeletal/physiology , Regeneration/physiology , Animals , Humans , Muscle Development , Muscle, Skeletal/chemistry , Muscle, Skeletal/cytology , Regenerative Medicine/methods
15.
Methods Mol Biol ; 482: 295-317, 2009.
Article in English | MEDLINE | ID: mdl-19089364

ABSTRACT

Stem cells are present in all tissues and organs, and are crucial for normal regulated growth. How the pool size of stem cells and their progeny is regulated to establish the tissue prenatally, then maintain it throughout life, is a key question in biology and medicine. The ability to precisely locate stem and progenitors requires defining lineage progression from stem to differentiated cells, assessing the mode of cell expansion and self-renewal and identifying markers to assess the different cell states within the lineage. We have shown that during lineage progression from a quiescent adult muscle satellite cell to a differentiated myofibre, both symmetric and asymmetric divisions take place. Furthermore, we provide evidence that a sub-population of label retaining satellite cells co-segregate template DNA strands to one daughter cell. These findings provide a means of identifying presumed stem and progenitor cells within the lineage. In addition, asymmetric segregation of template DNA and the cytoplasmic protein Numb provides a landmark to define cell behaviour as self-renewal and differentiation decisions are being executed.


Subject(s)
Cell Culture Techniques/methods , Cell Division , DNA/metabolism , Muscle, Skeletal/cytology , Stem Cells/cytology , Templates, Genetic , Animals , Antibodies , Bromodeoxyuridine/metabolism , Cell Adhesion , Cell Separation , Clone Cells , Deoxyribonucleases/metabolism , Dissection , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , Membrane Proteins/metabolism , Mice , Microscopy, Video , Mitosis , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , PAX7 Transcription Factor/metabolism , Polylysine , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Stem Cells/metabolism
16.
Dev Biol ; 312(1): 13-28, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17961534

ABSTRACT

The myogenic determination genes Myf5, Myod and Mrf4 direct skeletal muscle cell fate prenatally. In adult myogenesis, Myod has been shown to regulate myoblast differentiation, however, our understanding of satellite cell regulation is incomplete since the roles of Myf5 and Mrf4 had not been clearly defined. Here we examine the function of Myf5 and Mrf4 in the adult using recently generated alleles. Mrf4 is not expressed in normal or Myf5 null satellite cells and myoblasts, therefore excluding a role for this determination gene in adult muscle progenitors. Skeletal muscles of adult Myf5 null mice exhibit a subtle progressive myopathy. Crucially, adult Myf5 null mice exhibit perturbed muscle regeneration with a significant increase in muscle fibre hypertrophy, delayed differentiation, adipocyte accumulation, and fibrosis after freeze-injury. Satellite cell numbers are not significantly altered in Myf5 null animals and they show a modest impaired proliferation under some conditions in vitro. Mice double mutant for Myf5 and Dystrophin were more severely affected than single mutants, with enhanced necrosis and regeneration. Therefore, we show that Myf5 is a regulator of regenerative myogenesis and homeostasis, with functions distinct from those of Myod and Mrf4.


Subject(s)
Muscle Development , Muscle, Skeletal/physiology , Myogenic Regulatory Factor 5/genetics , Regeneration , Animals , Cell Count , Cell Differentiation , Cell Proliferation , Mice , Mice, Knockout , Muscle Cells/cytology , Muscle Cells/metabolism , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology
17.
Nat Cell Biol ; 8(7): 677-87, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16799552

ABSTRACT

Satellite cells assure postnatal skeletal muscle growth and repair. Despite extensive studies, their stem cell character remains largely undefined. Using pulse-chase labelling with BrdU to mark the putative stem cell niche, we identify a subpopulation of label-retaining satellite cells during growth and after injury. Strikingly, some of these cells display selective template-DNA strand segregation during mitosis in the muscle fibre in vivo, as well as in culture independent of their niche, indicating that genomic DNA strands are nonequivalent. Furthermore, we demonstrate that the asymmetric cell-fate determinant Numb segregates selectively to one daughter cell during mitosis and before differentiation, suggesting that Numb is associated with self-renewal. Finally, we show that template DNA cosegregates with Numb in label-retaining cells that express the self-renewal marker Pax7. The cosegregation of 'immortal' template DNA strands and their link with the asymmetry apparatus has important implications for stem cell biology and cancer.


Subject(s)
DNA Replication/physiology , Mitosis/physiology , Muscle, Skeletal/metabolism , Regeneration/physiology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Bromodeoxyuridine , Cell Lineage/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , DNA, Single-Stranded/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Satellite Cells, Skeletal Muscle/cytology
18.
J Cell Biol ; 172(1): 91-102, 2006 Jan 02.
Article in English | MEDLINE | ID: mdl-16380438

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.


Subject(s)
Muscle, Skeletal/cytology , MyoD Protein/metabolism , PAX7 Transcription Factor/physiology , Paired Box Transcription Factors/physiology , Satellite Cells, Skeletal Muscle/physiology , Animals , Apoptosis , Cell Cycle , Cell Differentiation/genetics , Cell Survival/physiology , Cells, Cultured , Gene Expression Regulation, Developmental/physiology , Mice , Mutation , PAX3 Transcription Factor , PAX7 Transcription Factor/genetics , Paired Box Transcription Factors/genetics , Satellite Cells, Skeletal Muscle/cytology
19.
Genes Dev ; 19(12): 1426-31, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15964993

ABSTRACT

Skeletal muscle serves as a paradigm for the acquisition of cell fate, yet the relationship between primitive cell populations and emerging myoblasts has remained elusive. We identify a novel population of resident Pax3+/Pax7+, muscle marker-negative cells throughout development. Using mouse mutants that uncouple myogenic progression, we show that these Pax+ cells give rise to muscle progenitors. In the absence of skeletal muscle, they apoptose after down-regulation of Pax7. Furthermore, they mark the emergence of satellite cells during fetal development, and do not require Pax3 function. These findings identify critical cell populations during lineage restriction, and provide a framework for defining myogenic cell states for therapeutic studies.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Myoblasts, Skeletal/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Biomarkers , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Muscle Development/genetics , Muscle Development/physiology , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/cytology , PAX3 Transcription Factor , PAX7 Transcription Factor , Paired Box Transcription Factors , Transcription Factors/deficiency , Transcription Factors/genetics
20.
Nature ; 431(7007): 466-71, 2004 Sep 23.
Article in English | MEDLINE | ID: mdl-15386014

ABSTRACT

In vertebrates, skeletal muscle is a model for the acquisition of cell fate from stem cells. Two determination factors of the basic helix-loop-helix myogenic regulatory factor (MRF) family, Myf5 and Myod, are thought to direct this transition because double-mutant mice totally lack skeletal muscle fibres and myoblasts. In the absence of these factors, progenitor cells remain multipotent and can change their fate. Gene targeting studies have revealed hierarchical relationships between these and the other MRF genes, Mrf4 and myogenin, where the latter are regarded as differentiation genes. Here we show, using an allelic series of three Myf5 mutants that differentially affect the expression of the genetically linked Mrf4 gene, that skeletal muscle is present in the new Myf5:Myod double-null mice only when Mrf4 expression is not compromised. This finding contradicts the widely held view that myogenic identity is conferred solely by Myf5 and Myod, and identifies Mrf4 as a determination gene. We revise the epistatic relationship of the MRFs, in which both Myf5 and Mrf4 act upstream of Myod to direct embryonic multipotent cells into the myogenic lineage.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/deficiency , Gene Deletion , Muscle Proteins/deficiency , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , MyoD Protein/genetics , Myogenic Regulatory Factors/metabolism , Trans-Activators/deficiency , Alleles , Animals , Cell Lineage , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Muscle Proteins/genetics , Muscle, Skeletal/embryology , MyoD Protein/metabolism , Myogenic Regulatory Factor 5 , Myogenin , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...