Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
Development ; 150(11)2023 06 01.
Article in English | MEDLINE | ID: mdl-37272529

ABSTRACT

The mechanism of pattern formation during limb muscle development remains poorly understood. The canonical view holds that naïve limb muscle progenitor cells (MPCs) invade a pre-established pattern of muscle connective tissue, thereby forming individual muscles. Here, we show that early murine embryonic limb MPCs highly accumulate pSMAD1/5/9, demonstrating active signaling of bone morphogenetic proteins (BMP) in these cells. Overexpression of inhibitory human SMAD6 (huSMAD6) in limb MPCs abrogated BMP signaling, impaired their migration and proliferation, and accelerated myogenic lineage progression. Fewer primary myofibers developed, causing an aberrant proximodistal muscle pattern. Patterning was not disturbed when huSMAD6 was overexpressed in differentiated muscle, implying that the proximodistal muscle pattern depends on BMP-mediated expansion of MPCs before their differentiation. We show that limb MPCs differentially express Hox genes, and Hox-expressing MPCs displayed active BMP signaling. huSMAD6 overexpression caused loss of HOXA11 in early limb MPCs. In conclusion, our data show that BMP signaling controls expansion of embryonic limb MPCs as a prerequisite for establishing the proximodistal muscle pattern, a process that involves expression of Hox genes.


Subject(s)
Bone Morphogenetic Proteins , Muscle, Skeletal , Animals , Humans , Mice , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Genes, Homeobox , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Smad6 Protein/metabolism
2.
PLoS Genet ; 19(6): e1010781, 2023 06.
Article in English | MEDLINE | ID: mdl-37267426

ABSTRACT

Four SIX homeoproteins display a combinatorial expression throughout embryonic developmental myogenesis and they modulate the expression of the myogenic regulatory factors. Here, we provide a deep characterization of their role in distinct mouse developmental territories. We showed, at the hypaxial level, that the Six1:Six4 double knockout (dKO) somitic precursor cells adopt a smooth muscle fate and lose their myogenic identity. At the epaxial level, we demonstrated by the analysis of Six quadruple KO (qKO) embryos, that SIX are required for fetal myogenesis, and for the maintenance of PAX7+ progenitor cells, which differentiated prematurely and are lost by the end of fetal development in qKO embryos. Finally, we showed that Six1 and Six2 are required to establish craniofacial myogenesis by controlling the expression of Myf5. We have thus described an unknown role for SIX proteins in the control of myogenesis at different embryonic levels and refined their involvement in the genetic cascades operating at the head level and in the genesis of myogenic stem cells.


Subject(s)
Homeodomain Proteins , Somites , Mice , Animals , Homeodomain Proteins/metabolism , Cell Differentiation/genetics , Somites/metabolism , Muscle Development/genetics , Gene Expression Regulation, Developmental , Muscle, Skeletal/metabolism
3.
J Cell Sci ; 136(16)2023 08 15.
Article in English | MEDLINE | ID: mdl-37589341

ABSTRACT

Bioenergetic metabolism is a key regulator of cellular function and signaling, but how it can instruct the behavior of cells and their fate during embryonic development remains largely unknown. Here, we investigated the role of glucose metabolism in the development of avian trunk neural crest cells (NCCs), a migratory stem cell population of the vertebrate embryo. We uncovered that trunk NCCs display glucose oxidation as a prominent metabolic phenotype, in contrast to what is seen for cranial NCCs, which instead rely on aerobic glycolysis. In addition, only one pathway downstream of glucose uptake is not sufficient for trunk NCC development. Indeed, glycolysis, mitochondrial respiration and the pentose phosphate pathway are all mobilized and integrated for the coordinated execution of diverse cellular programs, epithelial-to-mesenchymal transition, adhesion, locomotion, proliferation and differentiation, through regulation of specific gene expression. In the absence of glucose, the OXPHOS pathway fueled by pyruvate failed to promote trunk NCC adaptation to environmental stiffness, stemness maintenance and fate-decision making. These findings highlight the need for trunk NCCs to make the most of the glucose pathway potential to meet the high metabolic demands appropriate for their development.


Subject(s)
Glucose , Neural Crest , Quail , Quail/growth & development , Quail/metabolism , Animals , Neural Crest/growth & development , Neural Crest/metabolism , Glucose/metabolism , Neural Tube/cytology , Cells, Cultured , In Vitro Techniques , Oxidative Phosphorylation , Metabolic Networks and Pathways , Cell Adhesion
4.
Development ; 149(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-35005776

ABSTRACT

The location and regulation of fusion events within skeletal muscles during development remain unknown. Using the fusion marker myomaker (Mymk), named TMEM8C in chicken, as a readout of fusion, we identified a co-segregation of TMEM8C-positive cells and MYOG-positive cells in single-cell RNA-sequencing datasets of limbs from chicken embryos. We found that TMEM8C transcripts, MYOG transcripts and the fusion-competent MYOG-positive cells were preferentially regionalized in central regions of foetal muscles. We also identified a similar regionalization for the gene encoding the NOTCH ligand JAG2 along with an absence of NOTCH activity in TMEM8C+ fusion-competent myocytes. NOTCH function in myoblast fusion had not been addressed so far. We analysed the consequences of NOTCH inhibition for TMEM8C expression and myoblast fusion during foetal myogenesis in chicken embryos. NOTCH inhibition increased myoblast fusion and TMEM8C expression and released the transcriptional repressor HEYL from the TMEM8C regulatory regions. These results identify a regionalization of TMEM8C-dependent fusion and a molecular mechanism underlying the fusion-inhibiting effect of NOTCH in foetal myogenesis. The modulation of NOTCH activity in the fusion zone could regulate the flux of fusion events.


Subject(s)
Avian Proteins/metabolism , Muscle Development , Muscle Proteins/metabolism , Myoblasts/metabolism , Receptors, Notch/metabolism , Animals , Cells, Cultured , Chick Embryo , Membrane Proteins/metabolism , Myoblasts/cytology , Signal Transduction
5.
Nature ; 557(7707): 714-718, 2018 05.
Article in English | MEDLINE | ID: mdl-29795344

ABSTRACT

The cell microenvironment, which is critical for stem cell maintenance, contains both cellular and non-cellular components, including secreted growth factors and the extracellular matrix1-3. Although Notch and other signalling pathways have previously been reported to regulate quiescence of stem cells4-9, the composition and source of molecules that maintain the stem cell niche remain largely unknown. Here we show that adult muscle satellite (stem) cells in mice produce extracellular matrix collagens to maintain quiescence in a cell-autonomous manner. Using chromatin immunoprecipitation followed by sequencing, we identified NOTCH1/RBPJ-bound regulatory elements adjacent to specific collagen genes, the expression of which is deregulated in Notch-mutant mice. Moreover, we show that Collagen V (COLV) produced by satellite cells is a critical component of the quiescent niche, as depletion of COLV by conditional deletion of the Col5a1 gene leads to anomalous cell cycle entry and gradual diminution of the stem cell pool. Notably, the interaction of COLV with satellite cells is mediated by the Calcitonin receptor, for which COLV acts as a surrogate local ligand. Systemic administration of a calcitonin derivative is sufficient to rescue the quiescence and self-renewal defects found in COLV-null satellite cells. This study reveals a Notch-COLV-Calcitonin receptor signalling cascade that maintains satellite cells in a quiescent state in a cell-autonomous fashion, and raises the possibility that similar reciprocal mechanisms act in diverse stem cell populations.


Subject(s)
Calcitonin Receptor-Like Protein/metabolism , Collagen/metabolism , Muscle, Skeletal/cytology , Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction , Stem Cell Niche , Animals , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Collagen/genetics , Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Transcription, Genetic
6.
Exp Cell Res ; 420(1): 113275, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35931143

ABSTRACT

After extensive proliferation during development, the adult skeletal muscle cells remain outside the cell cycle, either as post-mitotic myofibers or as quiescent muscle stem cells (MuSCs, also known as satellite cells). Despite its terminally differentiated state, adult skeletal muscle has a remarkable regeneration potential, driven by MuSCs. Upon injury, MuSC quiescence is reversed to support tissue growth and repair and it is re-established after the completion of muscle regeneration. The distinct cell cycle states and transitions observed in the different myogenic populations are orchestrated by elements of the cell cycle machinery. This consists of i) complexes of cyclins and Cyclin-Dependent Kinases (CDKs) that ensure cell cycle progression and ii) their negative regulators, the Cyclin-Dependent Kinase Inhibitors (CDKIs). In this review we discuss the roles of these factors in developmental and adult myogenesis, with a focus on CDKIs that have emerging roles in stem cell functions.


Subject(s)
Cyclins , Satellite Cells, Skeletal Muscle , Cell Cycle , Cell Differentiation/physiology , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , Muscle Development , Muscle, Skeletal , Stem Cells
7.
PLoS Genet ; 16(11): e1009164, 2020 11.
Article in English | MEDLINE | ID: mdl-33175861

ABSTRACT

The chromosome translocations generating PAX3-FOXO1 and PAX7-FOXO1 chimeric proteins are the primary hallmarks of the paediatric fusion-positive alveolar subtype of Rhabdomyosarcoma (FP-RMS). Despite the ability of these transcription factors to remodel chromatin landscapes and promote the expression of tumour driver genes, they only inefficiently promote malignant transformation in vivo. The reason for this is unclear. To address this, we developed an in ovo model to follow the response of spinal cord progenitors to PAX-FOXO1s. Our data demonstrate that PAX-FOXO1s, but not wild-type PAX3 or PAX7, trigger the trans-differentiation of neural cells into FP-RMS-like cells with myogenic characteristics. In parallel, PAX-FOXO1s remodel the neural pseudo-stratified epithelium into a cohesive mesenchyme capable of tissue invasion. Surprisingly, expression of PAX-FOXO1s, similar to wild-type PAX3/7, reduce the levels of CDK-CYCLIN activity and increase the fraction of cells in G1. Introduction of CYCLIN D1 or MYCN overcomes this PAX-FOXO1-mediated cell cycle inhibition and promotes tumour growth. Together, our findings reveal a mechanism that can explain the apparent limited oncogenicity of PAX-FOXO1 fusion transcription factors. They are also consistent with certain clinical reports indicative of a neural origin of FP-RMS.


Subject(s)
Cell Transdifferentiation/genetics , Cell Transformation, Neoplastic/genetics , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/metabolism , Rhabdomyosarcoma, Alveolar/genetics , Animals , Biopsy , Chick Embryo , Child , Cyclin D1/genetics , Datasets as Topic , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Invasiveness/genetics , Neural Stem Cells/pathology , Neural Tube/cytology , Oncogene Proteins, Fusion/genetics , PAX3 Transcription Factor/genetics , PAX3 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Paired Box Transcription Factors/genetics , Rhabdomyosarcoma, Alveolar/pathology , S Phase/genetics
8.
Development ; 145(21)2018 10 29.
Article in English | MEDLINE | ID: mdl-30266829

ABSTRACT

The transcription factor Nfix belongs to the nuclear factor one family and has an essential role in prenatal skeletal muscle development, where it is a master regulator of the transition from embryonic to foetal myogenesis. Recently, Nfix was shown to be involved in adult muscle regeneration and in muscular dystrophies. Here, we have investigated the signalling that regulates Nfix expression, and show that JunB, a member of the AP-1 family, is an activator of Nfix, which then leads to foetal myogenesis. Moreover, we demonstrate that their expression is regulated through the RhoA/ROCK axis, which maintains embryonic myogenesis. Specifically, RhoA and ROCK repress ERK kinase activity, which promotes JunB and Nfix expression. Notably, the role of ERK in the activation of Nfix is conserved postnatally in satellite cells, which represent the canonical myogenic stem cells of adult muscle. As lack of Nfix in muscular dystrophies rescues the dystrophic phenotype, the identification of this pathway provides an opportunity to pharmacologically target Nfix in muscular dystrophies.


Subject(s)
MAP Kinase Signaling System , Muscle Development , Myoblasts/metabolism , NFI Transcription Factors/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Embryo, Mammalian/metabolism , Female , Fetus/metabolism , Gene Expression Regulation, Developmental , Gene Silencing , Male , Mice , NFI Transcription Factors/genetics , Stem Cells/metabolism , Transcription Factors/metabolism , rho-Associated Kinases/metabolism
9.
Mol Ther ; 28(8): 1887-1901, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32470325

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease mainly caused by mutations or deletions in the survival of motor neuron 1 (SMN1) gene and characterized by the degeneration of motor neurons and progressive muscle weakness. A viable therapeutic approach for SMA patients is a gene replacement strategy that restores functional SMN expression using adeno-associated virus serotype 9 (AAV9) vectors. Currently, systemic or intra-cerebrospinal fluid (CSF) delivery of AAV9-SMN is being explored in clinical trials. In this study, we show that the postnatal delivery of an AAV9 that expresses SMN under the control of the neuron-specific promoter synapsin selectively targets neurons without inducing re-expression in the peripheral organs of SMA mice. However, this approach is less efficient in restoring the survival and neuromuscular functions of SMA mice than the systemic or intra-CSF delivery of an AAV9 in which SMN is placed under the control of a ubiquitous promoter. This study suggests that further efforts are needed to understand the extent to which SMN is required in neurons and peripheral organs for a successful therapeutic effect.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Motor Neurons/metabolism , Motor Neurons/virology , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Animals , Disease Models, Animal , Gene Expression , Gene Transfer Techniques , Genetic Therapy , Locomotion , Mice , Muscular Atrophy, Spinal/drug therapy , Phenotype , Prognosis , Promoter Regions, Genetic , Spinal Cord/metabolism , Spinal Cord/pathology , Survival of Motor Neuron 1 Protein/metabolism , Transduction, Genetic , Treatment Outcome
10.
Int J Mol Sci ; 22(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884532

ABSTRACT

Skeletal muscle development and regeneration rely on the successive activation of specific transcription factors that engage cellular fate, promote commitment, and drive differentiation. Emerging evidence demonstrates that epigenetic regulation of gene expression is crucial for the maintenance of the cell differentiation status upon division and, therefore, to preserve a specific cellular identity. This depends in part on the regulation of chromatin structure and its level of condensation. Chromatin architecture undergoes remodeling through changes in nucleosome composition, such as alterations in histone post-translational modifications or exchange in the type of histone variants. The mechanisms that link histone post-translational modifications and transcriptional regulation have been extensively evaluated in the context of cell fate and differentiation, whereas histone variants have attracted less attention in the field. In this review, we discuss the studies that have provided insights into the role of histone variants in the regulation of myogenic gene expression, myoblast differentiation, and maintenance of muscle cell identity.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Genetic Variation , Histones/genetics , Muscle Development , MyoD Protein/metabolism , Animals , Chromatin Assembly and Disassembly , Histones/metabolism , Humans , MyoD Protein/genetics
11.
Development ; 144(15): 2737-2747, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28694257

ABSTRACT

Postnatal growth of skeletal muscle largely depends on the expansion and differentiation of resident stem cells, the so-called satellite cells. Here, we demonstrate that postnatal satellite cells express components of the bone morphogenetic protein (BMP) signaling machinery. Overexpression of noggin in postnatal mice (to antagonize BMP ligands), satellite cell-specific knockout of Alk3 (the gene encoding the BMP transmembrane receptor) or overexpression of inhibitory SMAD6 decreased satellite cell proliferation and accretion during myofiber growth, and ultimately retarded muscle growth. Moreover, reduced BMP signaling diminished the adult satellite cell pool. Abrogation of BMP signaling in satellite cell-derived primary myoblasts strongly diminished cell proliferation and upregulated the expression of cell cycle inhibitors p21 and p57 In conclusion, these results show that BMP signaling defines postnatal muscle development by regulating satellite cell-dependent myofiber growth and the generation of the adult muscle stem cell pool.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Blotting, Western , Bone Morphogenetic Proteins/genetics , Cell Proliferation/genetics , Cell Proliferation/physiology , Cells, Cultured , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology
13.
Dev Biol ; 432(1): 24-33, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28625870

ABSTRACT

Transcription factors are key orchestrators of the emergence of neuronal diversity within the developing spinal cord. As such, the two paralogous proteins Pax3 and Pax7 regulate the specification of progenitor cells within the intermediate neural tube, by defining a neat segregation between those fated to form motor circuits and those involved in the integration of sensory inputs. To attain insights into the molecular means by which they control this process, we have performed detailed phenotypic analyses of the intermediate spinal interneurons (IN), namely the dI6, V0D, V0VCG and V1 populations in compound null mutants for Pax3 and Pax7. This has revealed that the levels of Pax3/7 proteins determine both the dorso-ventral extent and the number of cells produced in each subpopulation; with increasing levels leading to the dorsalisation of their fate. Furthermore, thanks to the examination of mutants in which Pax3 transcriptional activity is skewed either towards repression or activation, we demonstrate that this cell diversification process is mainly dictated by Pax3/7 ability to repress gene expression. Consistently, we show that Pax3 and Pax7 inhibit the expression of Dbx1 and of its repressor Prdm12, fate determinants of the V0 and V1 interneurons, respectively. Notably, we provide evidence for the activity of several cis-regulatory modules of Dbx1 to be sensitive to Pax3 and Pax7 transcriptional activity levels. Altogether, our study provides insights into how the redundancy within a TF family, together with discrete dynamics of expression profiles of each member, are exploited to generate cellular diversity. Furthermore, our data supports the model whereby cell fate choices in the neural tube do not rely on binary decisions but rather on inhibition of multiple alternative fates.


Subject(s)
Homeodomain Proteins/physiology , Interneurons/physiology , Nerve Tissue Proteins/physiology , PAX3 Transcription Factor/physiology , PAX7 Transcription Factor/physiology , Spinal Cord/cytology , Animals , Cell Differentiation/physiology , Chick Embryo , Gene Expression Regulation, Developmental , Interneurons/cytology , Mice , Neural Tube/physiology , Spinal Cord/embryology , Stem Cells/cytology , Stem Cells/physiology
14.
Development ; 142(7): 1242-53, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25742797

ABSTRACT

The satellite cells, which serve as adult muscle stem cells, are both located beneath myofiber basement membranes and closely associated with capillary endothelial cells. We observed that 90% of capillaries were associated with pericytes in adult mouse and human muscle. During post-natal growth, newly formed vessels with their neuroglial 2 proteoglycan (NG2)-positive pericytes became progressively associated with the post-natal muscle stem cells, as myofibers increased in size and satellite cells entered into quiescence. In vitro, human muscle-derived pericytes promoted myogenic cell differentiation through insulin-like growth factor 1 (IGF1) and myogenic cell quiescence through angiopoietin 1 (ANGPT1). Diphtheria toxin-induced ablation of muscle pericytes in growing mice led both to myofiber hypotrophy and to impaired establishment of stem cells quiescence. Similar effects were observed following conditional in vivo deletion of pericyte Igf1 and Angpt1 genes, respectively. Our data therefore demonstrate that, by promoting post-natal myogenesis and stem cell quiescence, pericytes play a key role in the microvascular niche of satellite cells.


Subject(s)
Cell Cycle , Muscle Fibers, Skeletal/cytology , Neovascularization, Physiologic , Pericytes/cytology , Satellite Cells, Skeletal Muscle/cytology , Adolescent , Angiopoietin-1/metabolism , Animals , Animals, Newborn , Cell Proliferation , Child , Child, Preschool , Endothelial Cells/cytology , Gene Deletion , Humans , Infant , Insulin-Like Growth Factor I/metabolism , Mice, Inbred C57BL , Muscle Development , Muscle Fibers, Skeletal/metabolism , Pericytes/metabolism , Receptors, Cell Surface/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Stem Cells/cytology , Young Adult
15.
Genesis ; 55(4)2017 04.
Article in English | MEDLINE | ID: mdl-28196404

ABSTRACT

p57Kip2 (p57) is a maternally expressed imprinted gene regulating growth arrest which belongs to the CIP/KIP family of cyclin-dependent kinase inhibitors. While initially identified as a cell cycle arrest protein through inhibition of cyclin and cyclin-dependent kinase complexes, p57 activity has also been linked to differentiation, apoptosis, and senescence. In addition, p57 has recently been shown to be involved in tumorigenesis and cell fate decisions in stem cells. Yet, p57 function in adult tissues remains poorly characterized due to the perinatal lethality of p57 knock-out mice. To analyze p57 tissue-specific activity, we generated a conditional mouse line (p57FL-ILZ/+ ) by flanking the coding exons 2-3 by LoxP sites. To track p57-expressing or mutant cells, the p57FL-ILZ allele also contains an IRES-linked ß-galactosidase reporter inserted in the 3' UTR of the gene. Here, we show that the ß-galactosidase reporter expression pattern recapitulates p57 tissue specificity during development and in postnatal mice. Furthermore, we crossed the p57FL-ILZ/+ mice with PGK-Cre mice to generate p57cKO-ILZ/+ animals with ubiquitous loss of p57. p57cKO-ILZ/+ mice display developmental phenotypes analogous to previously described p57 knock-outs. Thus, p57FL-ILZ/+ is a new genetic tool allowing expression and functional conditional analyses of p57.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/genetics , Gene Targeting/methods , Mutation , Alleles , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Embryo, Mammalian/metabolism , Female , Genes, Reporter , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Phenotype , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
16.
Semin Cell Dev Biol ; 44: 115-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26424495

ABSTRACT

Like other subclasses within the PAX transcription factor family, PAX3 and PAX7 play important roles in the emergence of a number of different tissues during development. PAX3 regulates neural crest and, together with its orthologue PAX7, is also expressed in parts of the central nervous system. In this chapter we will focus on their role in skeletal muscle. Both factors are key regulators of myogenesis where Pax3 plays a major role during early skeletal muscle formation in the embryo while Pax7 predominates during post-natal growth and muscle regeneration in the adult. We review the expression and functions of these factors in the myogenic context. We also discuss mechanistic aspects of PAX3/7 function and modulation of their activity by interaction with other proteins, as well as the post-transcriptional and transcriptional regulation of their expression.


Subject(s)
Muscle Development/physiology , PAX7 Transcription Factor/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Humans , PAX3 Transcription Factor , Paired Box Transcription Factors/physiology
18.
Development ; 141(14): 2780-90, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25005473

ABSTRACT

A central question in development is to define how the equilibrium between cell proliferation and differentiation is temporally and spatially regulated during tissue formation. Here, we address how interactions between cyclin-dependent kinase inhibitors essential for myogenic growth arrest (p21(cip1) and p57(kip2)), the Notch pathway and myogenic regulatory factors (MRFs) orchestrate the proliferation, specification and differentiation of muscle progenitor cells. We first show that cell cycle exit and myogenic differentiation can be uncoupled. In addition, we establish that skeletal muscle progenitor cells require Notch signaling to maintain their cycling status. Using several mouse models combined with ex vivo studies, we demonstrate that Notch signaling is required to repress p21(cip1) and p57(kip2) expression in muscle progenitor cells. Finally, we identify a muscle-specific regulatory element of p57(kip2) directly activated by MRFs in myoblasts but repressed by the Notch targets Hes1/Hey1 in progenitor cells. We propose a molecular mechanism whereby information provided by Hes/Hey downstream of Notch as well as MRF activities are integrated at the level of the p57(kip2) enhancer to regulate the decision between progenitor cell maintenance and muscle differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Homeodomain Proteins/metabolism , Muscle, Skeletal/growth & development , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/metabolism , Receptors, Notch/metabolism , Animals , Cell Cycle Checkpoints , Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Enhancer Elements, Genetic/genetics , Extremities/embryology , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Mice, Transgenic , Muscle Development , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , MyoD Protein/genetics , Myoblasts/cytology , Myoblasts/metabolism , Organ Specificity , PAX7 Transcription Factor/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factor HES-1
19.
FASEB J ; 30(4): 1404-15, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26672000

ABSTRACT

Understanding the regulation of the stem cell fate is fundamental for designing novel regenerative medicine strategies. Previous studies have suggested that pharmacological treatments with small molecules provide a robust and reversible regulation of the stem cell program. Previously, we showed that treatment with a vanadium compound influences muscle cell fatein vitro In this study, we demonstrate that treatment with the phosphotyrosine phosphatase inhibitor bisperoxovanadium (BpV) drives primary muscle cells to a poised stem cell stage, with enhanced function in muscle regenerationin vivofollowing transplantation into injured muscles. Importantly, BpV-treated cells displayed increased self-renewal potentialin vivoand replenished the niche in both satellite and interstitial cell compartments. Moreover, we found that BpV treatment induces specific activating chromatin modifications at the promoter regions of genes associated with stem cell fate, includingSca-1andPw1 Thus, our findings indicate that BpV resets the cell fate program by specific epigenetic regulations, such that the committed myogenic cell fate is redirected to an earlier progenitor cell fate stage, which leads to an enhanced regenerative stem cell potential.-Smeriglio, P., Alonso-Martin, S., Masciarelli, S., Madaro, L., Iosue, I., Marrocco, V., Relaix, F., Fazi, F., Marazzi, G., Sassoon, D. A., Bouché, M. Phosphotyrosine phosphatase inhibitor bisperoxovanadium endows myogenic cells with enhanced muscle stem cell functionsviaepigenetic modulation of Sca-1 and Pw1 promoters.


Subject(s)
Antigens, Ly/genetics , Epigenesis, Genetic , Kruppel-Like Transcription Factors/genetics , Membrane Proteins/genetics , Muscle Cells/drug effects , Myoblasts, Skeletal/drug effects , Promoter Regions, Genetic/genetics , Vanadium Compounds/pharmacology , Animals , Blotting, Western , Cell Line , Cells, Cultured , Gene Expression/drug effects , Mice, Nude , Mice, Transgenic , Microscopy, Fluorescence , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/physiopathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Regeneration/drug effects , Reverse Transcriptase Polymerase Chain Reaction
20.
Proc Natl Acad Sci U S A ; 111(24): 8844-9, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24927569

ABSTRACT

Multipotent Pax3-positive (Pax3(+)) cells in the somites give rise to skeletal muscle and to cells of the vasculature. We had previously proposed that this cell-fate choice depends on the equilibrium between Pax3 and Foxc2 expression. In this study, we report that the Notch pathway promotes vascular versus skeletal muscle cell fates. Overactivating the Notch pathway specifically in Pax3(+) progenitors, via a conditional Pax3(NICD) allele, results in an increase of the number of smooth muscle and endothelial cells contributing to the aorta. At limb level, Pax3(+) cells in the somite give rise to skeletal muscles and to a subpopulation of endothelial cells in blood vessels of the limb. We now demonstrate that in addition to the inhibitory role of Notch signaling on skeletal muscle cell differentiation, the Notch pathway affects the Pax3:Foxc2 balance and promotes the endothelial versus myogenic cell fate, before migration to the limb, in multipotent Pax3(+) cells in the somite of the mouse embryo.


Subject(s)
Endothelial Cells/cytology , Extremities/embryology , Gene Expression Regulation, Developmental , Paired Box Transcription Factors/genetics , Receptors, Notch/metabolism , Somites/embryology , Alleles , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Female , Forkhead Transcription Factors/genetics , Genetic Vectors , Male , Mice , Mice, Transgenic , Muscle Development/physiology , Muscle, Skeletal/metabolism , PAX3 Transcription Factor , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL