Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4333, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37468485

ABSTRACT

Skeletal muscle fibers express distinct gene programs during development and maturation, but the underlying gene regulatory networks that confer stage-specific myofiber properties remain unknown. To decipher these distinctive gene programs and how they respond to neural activity, we generated a combined multi-omic single-nucleus RNA-seq and ATAC-seq atlas of mouse skeletal muscle development at multiple stages of embryonic, fetal, and postnatal life. We found that Myogenin, Klf5, and Tead4 form a transcriptional complex that synergistically activates the expression of muscle genes in developing myofibers. During myofiber maturation, the transcription factor Maf acts as a transcriptional switch to activate the mature fast muscle gene program. In skeletal muscles of mutant mice lacking voltage-gated L-type Ca2+ channels (Cav1.1), Maf expression and myofiber maturation are impaired. These findings provide a transcriptional atlas of muscle development and reveal genetic links between myofiber formation, maturation, and contraction.


Subject(s)
Muscle Fibers, Skeletal , Muscle, Skeletal , Mice , Animals , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Cell Differentiation
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.
Nat Commun ; 13(1): 1039, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35210422

ABSTRACT

The contractile properties of adult myofibers are shaped by their Myosin heavy chain isoform content. Here, we identify by snATAC-seq a 42 kb super-enhancer at the locus regrouping the fast Myosin genes. By 4C-seq we show that active fast Myosin promoters interact with this super-enhancer by DNA looping, leading to the activation of a single promoter per nucleus. A rainbow mouse transgenic model of the locus including the super-enhancer recapitulates the endogenous spatio-temporal expression of adult fast Myosin genes. In situ deletion of the super-enhancer by CRISPR/Cas9 editing demonstrates its major role in the control of associated fast Myosin genes, and deletion of two fast Myosin genes at the locus reveals an active competition of the promoters for the shared super-enhancer. Last, by disrupting the organization of fast Myosin, we uncover positional heterogeneity within limb skeletal muscles that may underlie selective muscle susceptibility to damage in certain myopathies.


Subject(s)
Muscle Fibers, Skeletal , Myosins , Animals , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosins/genetics , Myosins/metabolism , Phenotype
4.
STAR Protoc ; 2(3): 100694, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34382019

ABSTRACT

Single-nucleus RNA sequencing allows the profiling of gene expression in isolated nuclei. Here, we describe a step-by-step protocol optimized for adult mouse skeletal muscles. This protocol provides two main advantages compared to the widely used single-cell protocol. First, it allows us to sequence the myonuclei of the multinucleated myofibers. Second, it circumvents the cell-dissociation-induced transcriptional modifications. For complete details on the use and execution of this protocol, please refer to Dos Santos et al. (2020) and Machado, Geara et al. (2021).


Subject(s)
Cell Nucleus/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Cell Culture Techniques/methods , Cell Separation/methods , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , RNA/chemistry , RNA/isolation & purification , Satellite Cells, Skeletal Muscle/metabolism
5.
Mol Metab ; 51: 101228, 2021 09.
Article in English | MEDLINE | ID: mdl-33798773

ABSTRACT

OBJECTIVE: The metabolic master-switch AMP-activated protein kinase (AMPK) mediates insulin-independent glucose uptake in muscle and regulates the metabolic activity of brown and beige adipose tissue (BAT). The regulatory AMPKγ3 isoform is uniquely expressed in skeletal muscle and potentially in BAT. Herein, we investigated the role that AMPKγ3 plays in mediating skeletal muscle glucose uptake and whole-body glucose clearance in response to small-molecule activators that act on AMPK via distinct mechanisms. We also assessed whether γ3 plays a role in adipose thermogenesis and browning. METHODS: Global AMPKγ3 knockout (KO) mice were generated. A systematic whole-body, tissue, and molecular phenotyping linked to glucose homeostasis was performed in γ3 KO and wild-type (WT) mice. Glucose uptake in glycolytic and oxidative skeletal muscle ex vivo as well as blood glucose clearance in response to small molecule AMPK activators that target the nucleotide-binding domain of the γ subunit (AICAR) and allosteric drug and metabolite (ADaM) site located at the interface of the α and ß subunit (991, MK-8722) were assessed. Oxygen consumption, thermography, and molecular phenotyping with a ß3-adrenergic receptor agonist (CL-316,243) treatment were performed to assess BAT thermogenesis, characteristics, and function. RESULTS: Genetic ablation of γ3 did not affect body weight, body composition, physical activity, and parameters associated with glucose homeostasis under chow or high-fat diet. γ3 deficiency had no effect on fiber-type composition, mitochondrial content and components, or insulin-stimulated glucose uptake in skeletal muscle. Glycolytic muscles in γ3 KO mice showed a partial loss of AMPKα2 activity, which was associated with reduced levels of AMPKα2 and ß2 subunit isoforms. Notably, γ3 deficiency resulted in a selective loss of AICAR-, but not MK-8722-induced blood glucose-lowering in vivo and glucose uptake specifically in glycolytic muscle ex vivo. We detected γ3 in BAT and found that it preferentially interacts with α2 and ß2. We observed no differences in oxygen consumption, thermogenesis, morphology of BAT and inguinal white adipose tissue (iWAT), or markers of BAT activity between WT and γ3 KO mice. CONCLUSIONS: These results demonstrate that γ3 plays a key role in mediating AICAR- but not ADaM site binding drug-stimulated blood glucose clearance and glucose uptake specifically in glycolytic skeletal muscle. We also showed that γ3 is dispensable for ß3-adrenergic receptor agonist-induced thermogenesis and browning of iWAT.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Blood Glucose/metabolism , Muscle Fibers, Skeletal/metabolism , AMP-Activated Protein Kinases/genetics , Adipose Tissue, Brown/metabolism , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/analogs & derivatives , Animals , Benzimidazoles/administration & dosage , Diet, High-Fat , Female , Glucose Tolerance Test , Insulin/metabolism , Male , Metabolic Clearance Rate/drug effects , Mice , Mice, Knockout , Models, Animal , Pyridines/administration & dosage , Ribonucleotides/administration & dosage , Thermogenesis/drug effects
6.
Cell Stem Cell ; 28(6): 1125-1135.e7, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33609440

ABSTRACT

Tissue damage dramatically alters how cells interact with their microenvironment. These changes in turn dictate cellular responses, such as stem cell activation, yet early cellular responses in vivo remain ill defined. We generated single-cell and nucleus atlases from intact, dissociated, and injured muscle and liver and identified a common stress response signature shared by multiple cell types across these organs. This prevalent stress response was detected in published datasets across a range of tissues, demonstrating high conservation but also a significant degree of data distortion in single-cell reference atlases. Using quiescent muscle stem cells as a paradigm of cell activation following injury, we captured early cell activation following muscle injury and found that an essential ERK1/2 primary proliferation signal precedes initiation of the Notch-regulated myogenic program. This study defines initial events in response to tissue perturbation and identifies a broadly conserved transcriptional stress response that acts in parallel with cell-specific adaptive alterations.


Subject(s)
Satellite Cells, Skeletal Muscle , Cell Proliferation , Muscle Development , Muscles , Stem Cells
7.
Nat Commun ; 11(1): 5102, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037211

ABSTRACT

Skeletal muscle fibers are large syncytia but it is currently unknown whether gene expression is coordinately regulated in their numerous nuclei. Here we show by snRNA-seq and snATAC-seq that slow, fast, myotendinous and neuromuscular junction myonuclei each have different transcriptional programs, associated with distinct chromatin states and combinations of transcription factors. In adult mice, identified myofiber types predominantly express either a slow or one of the three fast isoforms of Myosin heavy chain (MYH) proteins, while a small number of hybrid fibers can express more than one MYH. By snRNA-seq and FISH, we show that the majority of myonuclei within a myofiber are synchronized, coordinately expressing only one fast Myh isoform with a preferential panel of muscle-specific genes. Importantly, this coordination of expression occurs early during post-natal development and depends on innervation. These findings highlight a previously undefined mechanism of coordination of gene expression in a syncytium.


Subject(s)
Cell Nucleus/genetics , Gene Expression Regulation , In Situ Hybridization, Fluorescence/methods , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Sequence Analysis, RNA/methods , Animals , Female , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Myosin Heavy Chains/genetics , Neuromuscular Junction/cytology , Single-Cell Analysis , Tendons/cytology , Transcription, Genetic
8.
Semin Cell Dev Biol ; 104: 51-64, 2020 08.
Article in English | MEDLINE | ID: mdl-32247726

ABSTRACT

SIX homeoproteins were first described in Drosophila, where they participate in the Pax-Six-Eya-Dach (PSED) network with eyeless, eyes absent and dachsund to drive synergistically eye development through genetic and biochemical interactions. The role of the PSED network and SIX proteins in muscle formation in vertebrates was subsequently identified. Evolutionary conserved interactions with EYA and DACH proteins underlie the activity of SIX transcriptional complexes (STC) both during embryogenesis and in adult myofibers. Six genes are expressed throughout muscle development, in embryonic and adult proliferating myogenic stem cells and in fetal and adult post-mitotic myofibers, where SIX proteins regulate the expression of various categories of genes. In vivo, SIX proteins control many steps of muscle development, acting through feedforward mechanisms: in the embryo for myogenic fate acquisition through the direct control of Myogenic Regulatory Factors; in adult myofibers for their contraction/relaxation and fatigability properties through the control of genes involved in metabolism, sarcomeric organization and calcium homeostasis. Furthermore, during development and in the adult, SIX homeoproteins participate in the genesis and the maintenance of myofibers diversity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/growth & development , Drosophila/genetics , Homeodomain Proteins/metabolism , Muscle Development/genetics , Muscle, Skeletal/metabolism , Animals , Muscle, Skeletal/cytology
9.
Stem Cell Res Ther ; 9(1): 258, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30261919

ABSTRACT

BACKGROUND: Skeletal muscle regeneration is possible thanks to unipotent stem cells, which are satellite cells connected to the myofibers. Populations of stem cells other than muscle-specific satellite cells are considered as sources of cells able to support skeletal muscle reconstruction. Among these are bone marrow-derived mesenchymal stem cells (BM-MSCs), which are multipotent, self-renewing stem cells present in the bone marrow stroma. Available data documenting the ability of BM-MSCs to undergo myogenic differentiation are not definitive. In the current work, we aimed to check if the satellite cell niche could impact the ability of bone marrow-derived cells to follow a myogenic program. METHODS: We established a new in-vitro method for the coculture of bone marrow-derived cells (BMCs) that express CXCR4 (CXCR4+BMCs; the stromal-derived factor-1 (Sdf-1) receptor) with myofibers. Using various tests, we analyzed the myogenic identity of BMCs and their ability to fuse with myoblasts in vitro and in vivo. RESULTS: We showed that Sdf-1 treatment increased the number of CXCR4+BMCs able to bind the myofiber and occupy the satellite cell niche. Moreover, interaction with myofibers induced the expression of myogenic regulatory factors (MRFs) in CXCR4+BMCs. CXCR4+BMCs, pretreated by the coculture with myofibers and Sdf-1, participated in myotube formation in vitro and also myofiber reconstruction in vivo. We also showed that Sdf-1 overexpression in vivo (in injured and regenerating muscles) supported the participation of CXCR4+BMCs in new myofiber formation. CONCLUSION: We showed that CXCR4+BMC interaction with myofibers (that is, within the satellite cell niche) induced CXCR4+BMC myogenic commitment. CXCR4+BMCs, pretreated using such a method of culture, were able to participate in skeletal muscle regeneration.


Subject(s)
Mesenchymal Stem Cells/metabolism , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Coculture Techniques , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Primary Cell Culture , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction
10.
Rheumatol Int ; 38(Suppl 1): 379-386, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29637354

ABSTRACT

The Juvenile Arthritis Multidimensional Assessment Report (JAMAR) is a new parent/patient reported outcome measure that enables a thorough assessment of the disease status in children with juvenile idiopathic arthritis (JIA). We report the results of the cross-cultural adaptation and validation of the parent and patient versions of the JAMAR in the Swiss French language. The reading comprehension of the questionnaire was tested in ten JIA parents and patients. Each participating centre was asked to collect demographic, clinical data, and the JAMAR in 100 consecutive JIA patients or all consecutive patients seen in a 6-month period and to administer the JAMAR to 100 healthy children and their parents. The statistical validation phase explored descriptive statistics and the psychometric issues of the JAMAR: the three Likert assumptions, floor/ceiling effects, internal consistency, Cronbach's alpha, interscale correlations, test-retest reliability, and construct validity (convergent and discriminant validity). A total of 98 JIA patients (3.1% systemic, 43.9% oligoarticular, 16.3% RF negative polyarthritis, 36.7% other categories), and 64 healthy children were enrolled in a paediatric rheumatology centre. The JAMAR components discriminated well healthy subjects from JIA patients. All JAMAR components revealed good psychometric performances. In conclusion, the Swiss French version of the JAMAR is a valid tool for the assessment of children with JIA and is suitable for use both in routine clinical practice and clinical research.


Subject(s)
Arthritis, Juvenile/diagnosis , Disability Evaluation , Patient Reported Outcome Measures , Rheumatology/methods , Adolescent , Age of Onset , Arthritis, Juvenile/physiopathology , Arthritis, Juvenile/psychology , Arthritis, Juvenile/therapy , Case-Control Studies , Child , Child, Preschool , Cultural Characteristics , Female , Health Status , Humans , Male , Parents/psychology , Patients/psychology , Predictive Value of Tests , Prognosis , Psychometrics , Quality of Life , Reproducibility of Results , Switzerland , Translating
11.
Sci Rep ; 8(1): 2075, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391529

ABSTRACT

The hereditary neurodegenerative disorder spinal muscular atrophy (SMA) is characterized by the loss of spinal cord motor neurons and skeletal muscle atrophy. SMA is caused by mutations of the survival motor neuron (SMN) gene leading to a decrease in SMN protein levels. The SMN deficiency alters nuclear body formation and whether it can contribute to the disease remains unclear. Here we screen a series of small-molecules on SMA patient fibroblasts and identify flunarizine that accumulates SMN into Cajal bodies, the nuclear bodies important for the spliceosomal small nuclear RNA (snRNA)-ribonucleoprotein biogenesis. Using histochemistry, real-time RT-PCR and behavioural analyses in a mouse model of SMA, we show that along with the accumulation of SMN into Cajal bodies of spinal cord motor neurons, flunarizine treatment modulates the relative abundance of specific spliceosomal snRNAs in a tissue-dependent manner and can improve the synaptic connections and survival of spinal cord motor neurons. The treatment also protects skeletal muscles from cell death and atrophy, raises the neuromuscular junction maturation and prolongs life span by as much as 40 percent (p < 0.001). Our findings provide a functional link between flunarizine and SMA pathology, highlighting the potential benefits of flunarizine in a novel therapeutic perspective against neurodegenerative diseases.


Subject(s)
Coiled Bodies/drug effects , Flunarizine/pharmacology , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Animals , Cell Line , Coiled Bodies/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Flunarizine/therapeutic use , HeLa Cells , Humans , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/drug therapy , Small Molecule Libraries/pharmacology
12.
Skelet Muscle ; 6(1): 30, 2016.
Article in English | MEDLINE | ID: mdl-27597886

ABSTRACT

BACKGROUND: Adult skeletal muscles are composed of slow and fast myofiber subtypes which each express selective genes required for their specific contractile and metabolic activity. Six homeoproteins are transcription factors regulating muscle cell fate through activation of myogenic regulatory factors and driving fast-type gene expression during embryogenesis. RESULTS: We show here that Six1 protein accumulates more robustly in the nuclei of adult fast-type muscles than in adult slow-type muscles, this specific enrichment takes place during perinatal growth. Deletion of Six1 in soleus impaired fast-type myofiber specialization during perinatal development, resulting in a slow phenotype and a complete lack of Myosin heavy chain 2A (MyHCIIA) expression. Global transcriptomic analysis of wild-type and Six1 mutant myofibers identified the gene networks controlled by Six1 in adult soleus muscle. This analysis showed that Six1 is required for the expression of numerous genes encoding fast-type sarcomeric proteins, glycolytic enzymes and controlling intracellular calcium homeostasis. Parvalbumin, a key player of calcium buffering, in particular, is a direct target of Six1 in the adult myofiber. CONCLUSIONS: This analysis revealed that Six1 controls distinct aspects of adult muscle physiology in vivo, and acts as a main determinant of fast-fiber type acquisition and maintenance.


Subject(s)
Homeodomain Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Calcium/metabolism , Gene Deletion , Gene Regulatory Networks , Glycolysis , Homeodomain Proteins/genetics , Male , Mice , Muscle Fibers, Skeletal/cytology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phenotype , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...