Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Development ; 151(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38240380

ABSTRACT

Skeletal muscle stem cells (MuSCs) are recognised as functionally heterogeneous. Cranial MuSCs are reported to have greater proliferative and regenerative capacity when compared with those in the limb. A comprehensive understanding of the mechanisms underlying this functional heterogeneity is lacking. Here, we have used clonal analysis, live imaging and single cell transcriptomic analysis to identify crucial features that distinguish extraocular muscle (EOM) from limb muscle stem cell populations. A MyogeninntdTom reporter showed that the increased proliferation capacity of EOM MuSCs correlates with deferred differentiation and lower expression of the myogenic commitment gene Myod. Unexpectedly, EOM MuSCs activated in vitro expressed a large array of extracellular matrix components typical of mesenchymal non-muscle cells. Computational analysis underscored a distinct co-regulatory module, which is absent in limb MuSCs, as driver of these features. The EOM transcription factor network, with Foxc1 as key player, appears to be hardwired to EOM identity as it persists during growth, disease and in vitro after several passages. Our findings shed light on how high-performing MuSCs regulate myogenic commitment by remodelling their local environment and adopting properties not generally associated with myogenic cells.


Subject(s)
Muscle, Skeletal , Oculomotor Muscles , Mice , Animals , Muscle, Skeletal/metabolism , Oculomotor Muscles/metabolism , Mice, Inbred C57BL , Cell Proliferation , Stem Cells
2.
Exp Cell Res ; 424(1): 113484, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36693490

ABSTRACT

A major challenge in the study of living systems is understanding how tissues and organs are established, maintained during homeostasis, reconstituted following injury or deteriorated during disease. Most of the studies that interrogate in vivo cell biological properties of cell populations within tissues are obtained through static imaging approaches. However, in vertebrates, little is known about which, when, and how extracellular and intracellular signals are dynamically integrated to regulate cell behaviour and fates, due largely to technical challenges. Intravital imaging of cellular dynamics in mammalian models has exposed surprising properties that have been missed by conventional static imaging approaches. Here we highlight some selected examples of intravital imaging in mouse intestinal stem cells, hematopoietic stem cells, hair follicle stem cells, and neural stem cells in the brain, each of which have distinct features from an anatomical and niche-architecture perspective. Intravital imaging of mouse skeletal muscles is comparatively less advanced due to several technical constraints that will be discussed, yet this approach holds great promise as a complementary investigative method to validate findings obtained by static imaging, as well as a method for discovery.


Subject(s)
Muscle, Skeletal , Neural Stem Cells , Mice , Animals , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/physiology , Hair Follicle , Hematopoietic Stem Cells , Mammals
3.
PLoS Genet ; 16(10): e1009022, 2020 10.
Article in English | MEDLINE | ID: mdl-33125370

ABSTRACT

Adult skeletal muscles are maintained during homeostasis and regenerated upon injury by muscle stem cells (MuSCs). A heterogeneity in self-renewal, differentiation and regeneration properties has been reported for MuSCs based on their anatomical location. Although MuSCs derived from extraocular muscles (EOM) have a higher regenerative capacity than those derived from limb muscles, the molecular determinants that govern these differences remain undefined. Here we show that EOM and limb MuSCs have distinct DNA methylation signatures associated with enhancers of location-specific genes, and that the EOM transcriptome is reprogrammed following transplantation into a limb muscle environment. Notably, EOM MuSCs expressed host-site specific positional Hox codes after engraftment and self-renewal within the host muscle. However, about 10% of EOM-specific genes showed engraftment-resistant expression, pointing to cell-intrinsic molecular determinants of the higher engraftment potential of EOM MuSCs. Our results underscore the molecular diversity of distinct MuSC populations and molecularly define their plasticity in response to microenvironmental cues. These findings provide insights into strategies designed to improve the functional capacity of MuSCs in the context of regenerative medicine.


Subject(s)
Cell Plasticity/genetics , Epigenome/genetics , Stem Cell Transplantation , Transcriptome/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Extremities/growth & development , Genetic Variation/genetics , Humans , Mice , Mice, Inbred C57BL , Muscle Cells/cytology , Muscle Fibers, Skeletal , Muscle, Skeletal/cytology , Myoblasts/cytology , Regeneration/genetics , Stem Cells/cytology , Stem Cells/metabolism
4.
Cell Rep ; 30(10): 3195-3206.e7, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160529

ABSTRACT

Stem cells can be maintained through symmetric cell divisions (SCDs) and asymmetric cell divisions (ACDs). How and when these divisions occur in vivo in vertebrates is poorly understood. Here, we developed a clonogenic cell tracing method that demonstrates the asymmetric distribution of transcription factors along with old and new DNA in mouse muscle stem cells during skeletal muscle regeneration. Combining single-cell tracking and artificial niches ex vivo, we show how cells switch from ACDs to SCDs, suggesting that they are not engaged in an obligate mode of cell division. Further, we generated SNAP-tagged histone H3-reporter mice and find that, unlike fly germline stem cells, differential fate outcomes are associated with a symmetric distribution of the H3.1 and H3.3 histone variants in mouse muscle stem cells. This versatile and efficient H3-SNAP labeling system will allow an investigation of mechanisms underlying the maintenance of epigenomic identity and plasticity in a variety of tissues.


Subject(s)
Asymmetric Cell Division , Muscles/cytology , Stem Cell Niche , Stem Cells/cytology , Animals , Cell Lineage , DNA/metabolism , Genes, Reporter , Histones/metabolism , Mice , Transgenes
5.
EMBO J ; 38(24): e103924, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31797391

ABSTRACT

Ageing is a multi-factorial condition that results in a gradual decline in tissue and organ function. Systemic, local and intrinsic factors play major roles in this process that also results in a decline in stem cell number and function. In this issue of The EMBO Journal, Li et al (2019) show that a subpopulation of mouse muscle stem cells is depleted in aged mice through loss of niche-derived granulocyte colony-stimulating factor (G-CSF).


Subject(s)
Cellular Senescence/physiology , Granulocyte Colony-Stimulating Factor/metabolism , Satellite Cells, Skeletal Muscle/physiology , Animals , Genetic Heterogeneity , Mice , Signal Transduction
6.
Nat Commun ; 10(1): 4361, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31554804

ABSTRACT

Age-related tissue alterations have been associated with a decline in stem cell number and function. Although increased cell-to-cell variability in transcription or epigenetic marks has been proposed to be a major hallmark of ageing, little is known about the molecular diversity of stem cells during ageing. Here we present a single cell multi-omics study of mouse muscle stem cells, combining single-cell transcriptome and DNA methylome profiling. Aged cells show a global increase of uncoordinated transcriptional heterogeneity biased towards genes regulating cell-niche interactions. We find context-dependent alterations of DNA methylation in aged stem cells. Importantly, promoters with increased methylation heterogeneity are associated with increased transcriptional heterogeneity of the genes they drive. These results indicate that epigenetic drift, by accumulation of stochastic DNA methylation changes in promoters, is associated with the degradation of coherent transcriptional networks during stem cell ageing. Furthermore, our observations also shed light on the mechanisms underlying the DNA methylation clock.


Subject(s)
Aging , Cellular Senescence , DNA Methylation , Stem Cells/metabolism , Transcriptome/genetics , Animals , Cells, Cultured , Epigenesis, Genetic , Epigenomics/methods , Gene Expression Profiling/methods , Gene Ontology , Humans , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Muscles/cytology , Promoter Regions, Genetic/genetics , Single-Cell Analysis , Stem Cells/cytology
7.
NPJ Regen Med ; 3: 24, 2018.
Article in English | MEDLINE | ID: mdl-30588332

ABSTRACT

Investigations on developmental and regenerative myogenesis have led to major advances in decrypting stem cell properties and potential, as well as their interactions within the evolving niche. As a consequence, regenerative myogenesis has provided a forum to investigate intrinsic regulators of stem cell properties as well as extrinsic factors, including stromal cells, during normal growth and following injury and disease. Here we review some of the latest advances in the field that have exposed fundamental processes including regulation of stress following trauma and ageing, senescence, DNA damage control and modes of symmetric and asymmetric cell divisions. Recent studies have begun to explore the nature of the niche that is distinct in different muscle groups, and that is altered from prenatal to postnatal stages, and during ageing. We also discuss heterogeneities among muscle stem cells and how distinct properties within the quiescent and proliferating cell states might impact on homoeostasis and regeneration. Interestingly, cellular quiescence, which was thought to be a passive cell state, is regulated by multiple mechanisms, many of which are deregulated in various contexts including ageing. These and other factors including metabolic activity and genetic background can impact on the efficiency of muscle regeneration.

8.
Cell Stem Cell ; 23(6): 859-868.e5, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30416072

ABSTRACT

Critical features of stem cells include anchoring within a niche and activation upon injury. Notch signaling maintains skeletal muscle satellite (stem) cell quiescence by inhibiting differentiation and inducing expression of extracellular components of the niche. However, the complete spectrum of how Notch safeguards quiescence is not well understood. Here, we perform Notch ChIP-sequencing and small RNA sequencing in satellite cells and identify the Notch-induced microRNA-708, which is a mirtron that is highly expressed in quiescent cells and sharply downregulated in activated cells. We employ in vivo and ex vivo functional studies, in addition to live imaging, to show that miR-708 regulates quiescence and self-renewal by antagonizing cell migration through targeting the transcripts of the focal-adhesion-associated protein Tensin3. Therefore, this study identifies a Notch-miR708-Tensin3 axis and suggests that Notch signaling can regulate satellite cell quiescence and transition to the activation state through dynamic regulation of the migratory machinery.


Subject(s)
Cell Movement/genetics , MicroRNAs/genetics , Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction , Stem Cell Niche , Animals , Female , Male , Mice , Mice, Transgenic
9.
Cell Mol Life Sci ; 74(10): 1923-1936, 2017 05.
Article in English | MEDLINE | ID: mdl-28025671

ABSTRACT

Muscle satellite cells are resistant to cytotoxic agents, and they express several genes that confer resistance to stress, thus allowing efficient dystrophic muscle regeneration after transplantation. However, once they are activated, this capacity to resist to aggressive agents is diminished resulting in massive death of transplanted cells. Although cell immaturity represents a survival advantage, the signalling pathways involved in the control of the immature state remain to be explored. Here, we show that incubation of human myoblasts with retinoic acid impairs skeletal muscle differentiation through activation of the retinoic-acid receptor family of nuclear receptor. Conversely, pharmacologic or genetic inactivation of endogenous retinoic-acid receptors improved myoblast differentiation. Retinoic acid inhibits the expression of early and late muscle differentiation markers and enhances the expression of myogenic specification genes, such as PAX7 and PAX3. These results suggest that the retinoic-acid-signalling pathway might maintain myoblasts in an undifferentiated/immature stage. To determine the relevance of these observations, we characterised the retinoic-acid-signalling pathways in freshly isolated satellite cells in mice and in siMYOD immature human myoblasts. Our analysis reveals that the immature state of muscle progenitors is correlated with high expression of several genes of the retinoic-acid-signalling pathway both in mice and in human. Taken together, our data provide evidences for an important role of the retinoic-acid-signalling pathway in the regulation of the immature state of muscle progenitors.


Subject(s)
Cell Differentiation , Muscle Development , Myoblasts/cytology , Myoblasts/metabolism , Tretinoin/metabolism , Adult , Animals , Cells, Cultured , Gene Expression Regulation, Developmental , Humans , Male , Mice , MyoD Protein/genetics , MyoD Protein/metabolism , RNA Interference , Receptors, Retinoic Acid/metabolism , Signal Transduction
10.
Genome Res ; 25(3): 353-67, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614607

ABSTRACT

Genomic imprinting is an epigenetic mechanism that restrains the expression of ∼ 100 eutherian genes in a parent-of-origin-specific manner. The reason for this selective targeting of genes with seemingly disparate molecular functions is unclear. In the present work, we show that imprinted genes are coexpressed in a network that is regulated at the transition from proliferation to quiescence and differentiation during fibroblast cell cycle withdrawal, adipogenesis in vitro, and muscle regeneration in vivo. Imprinted gene regulation is not linked to alteration of DNA methylation or to perturbation of monoallelic, parent-of-origin-dependent expression. Overexpression and knockdown of imprinted gene expression alters the sensitivity of preadipocytes to contact inhibition and adipogenic differentiation. In silico and in cellulo experiments showed that the imprinted gene network includes biallelically expressed, nonimprinted genes. These control the extracellular matrix composition, cell adhesion, cell junction, and extracellular matrix-activated and growth factor-activated signaling. These observations show that imprinted genes share a common biological process that may account for their seemingly diverse roles in embryonic development, obesity, diabetes, muscle physiology, and neoplasm.


Subject(s)
Epigenomics/methods , Genomic Imprinting , Adipogenesis/genetics , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line , Cluster Analysis , Computational Biology/methods , DNA Methylation , Databases, Nucleic Acid , Extracellular Matrix/genetics , Gene Expression Regulation , Gene Regulatory Networks , Mice
11.
Chromosome Res ; 21(3): 225-42, 2013 May.
Article in English | MEDLINE | ID: mdl-23681656

ABSTRACT

In recent years, our views on how DNA and genes are organised and regulated have evolved significantly. One example is provided by reports that single DNA strands in the double helix could carry distinct forms of information. That chromatids carrying old and nascently replicated DNA strands are recognised by the mitotic machinery, then segregated in a concerted way to distinct daughter cells after cell division is remarkable. Notably, this phenomenon in several cases has been associated with the cell fate choice of resulting daughter cells. Here, we review the evidence for asymmetric or template DNA strand segregation in mammals with a focus on skeletal muscle.


Subject(s)
DNA/metabolism , Gene Expression Regulation , Animals , Cell Lineage , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Regeneration , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...