Comparing the epigenetic landscape in myonuclei purified with a PCM1 antibody from a fast/glycolytic and a slow/oxidative muscle

Muscle cells have different phenotypes adapted to different usage, and can be grossly divided into fast/glycolytic and slow/oxidative types. While most muscles contain a mixture of such fiber types, we aimed at providing a genome-wide analysis of the epigenetic landscape by ChIP-Seq in two muscle extremes, the fast/glycolytic extensor digitorum longus (EDL) and slow/oxidative soleus muscles. Muscle is a heterogeneous tissue where up to 60% of the nuclei can be of a different origin. Since cellular homogeneity is critical in epigenome-wide association studies we developed a new method for purifying skeletal muscle nuclei from whole tissue, based on the nuclear envelope protein Pericentriolar material 1 (PCM1) being a specific marker for myonuclei. Using antibody labelling and a magnetic-assisted sorting approach, we were able to sort out myonuclei with 95% purity in muscles from mice, rats and humans. The sorting eliminated influence from the other cell types in the tissue and improved the myo-specific signal. A genome-wide comparison of the epigenetic landscape in EDL and soleus reflected the differences in the functional properties of the two muscles, and revealed distinct regulatory programs involving distal enhancers, including a glycolytic super-enhancer in the EDL. The two muscles were also regulated by different sets of transcription factors; e.g. in soleus, binding sites for MEF2C, NFATC2 and PPARA were enriched, while in EDL MYOD1 and SIX1 binding sites were found to be overrepresented. In addition, more novel transcription factors for muscle regulation such as members of the MAF family, ZFX and ZBTB14 were identified.

[1]  I. Dimauro,et al.  Function and Fiber-Type Specific Distribution of Hsp60 and αB-Crystallin in Skeletal Muscles: Role of Physical Exercise , 2021, Biology.

[2]  P. Maire,et al.  Single-nucleus RNA-seq and FISH identify coordinated transcriptional activity in mammalian myofibers , 2020, Nature Communications.

[3]  C. Reggiani,et al.  Muscle hypertrophy and muscle strength: dependent or independent variables? A provocative review , 2020, European journal of translational myology.

[4]  Tingting Fu,et al.  Histone methyltransferase MLL4 controls myofiber identity and muscle performance through MEF2 interaction. , 2020, The Journal of clinical investigation.

[5]  M. Murgia,et al.  Fiber type diversity in skeletal muscle explored by mass spectrometry-based single fiber proteomics. , 2020, Histology and histopathology.

[6]  O. Elemento,et al.  A reference single-cell transcriptomic atlas of human skeletal muscle tissue reveals bifurcated muscle stem cell populations , 2020, bioRxiv.

[7]  S. Sealfon,et al.  Single-cell transcriptional profiles in human skeletal muscle , 2020, Scientific Reports.

[8]  K. Sun,et al.  MyoD induced enhancer RNA interacts with hnRNPL to activate target gene transcription during myogenic differentiation , 2019, Nature Communications.

[9]  M. Kjaer,et al.  The influence of fibrillin‐1 and physical activity upon tendon tissue morphology and mechanical properties in mice , 2019, Physiological reports.

[10]  D. Bishop,et al.  Transcriptomic profiling of skeletal muscle adaptations to exercise and inactivity , 2019, Nature Communications.

[11]  G. Barish,et al.  Dynamic enhancers control skeletal muscle identity and reprogramming , 2019, PLoS biology.

[12]  F. Braet,et al.  Skeletal MyBP-C isoforms tune the molecular contractility of divergent skeletal muscle systems , 2019, Proceedings of the National Academy of Sciences.

[13]  O. Elemento,et al.  Single-cell analysis of the muscle stem cell hierarchy identifies heterotypic communication signals involved in skeletal muscle regeneration , 2019, bioRxiv.

[14]  G. Lanfranchi,et al.  Transcriptomic Analysis of Single Isolated Myofibers Identifies miR-27a-3p and miR-142-3p as Regulators of Metabolism in Skeletal Muscle. , 2019, Cell reports.

[15]  B. Peters,et al.  An ATAC-seq atlas of chromatin accessibility in mouse tissues , 2019, Scientific Data.

[16]  F. Saatcioglu,et al.  Cachexia does not induce loss of myonuclei or muscle fibres during xenografted prostate cancer in mice , 2018, Acta physiologica.

[17]  Kevin A. Murach,et al.  A novel tetracycline-responsive transgenic mouse strain for skeletal muscle-specific gene expression , 2018, Skeletal Muscle.

[18]  Joon-Young Park,et al.  A cellular mechanism of muscle memory facilitates mitochondrial remodelling following resistance training , 2018, The Journal of physiology.

[19]  R. Eskeland,et al.  The SUMO protease SENP1 and the chromatin remodeler CHD3 interact and jointly affect chromatin accessibility and gene expression , 2018, The Journal of Biological Chemistry.

[20]  K. Gundersen,et al.  Specific labelling of myonuclei by an antibody against pericentriolar material 1 on skeletal muscle tissue sections , 2018, Acta physiologica.

[21]  H. Wallberg-henriksson,et al.  IL6 and LIF mRNA expression in skeletal muscle is regulated by AMPK and the transcription factors NFYC, ZBTB14, and SP1. , 2018, American journal of physiology. Endocrinology and metabolism.

[22]  Y. Li,et al.  Transcriptional Regulation of the Warburg Effect in Cancer by SIX1. , 2018, Cancer cell.

[23]  P. Farnham,et al.  ZFX acts as a transcriptional activator in multiple types of human tumors by binding downstream from transcription start sites at the majority of CpG island promoters , 2018, Genome research.

[24]  M. Saklayen The Global Epidemic of the Metabolic Syndrome , 2018, Current Hypertension Reports.

[25]  S. Shepherd,et al.  Human Skeletal Muscle Possesses an Epigenetic Memory of Hypertrophy , 2018, Scientific Reports.

[26]  M. Rudnicki,et al.  The myogenic regulatory factors, determinants of muscle development, cell identity and regeneration. , 2017, Seminars in cell & developmental biology.

[27]  Scott A Lewis,et al.  Transcriptional profiling reveals extraordinary diversity among skeletal muscle tissues , 2017, bioRxiv.

[28]  J. Michael Cherry,et al.  The Encyclopedia of DNA elements (ENCODE): data portal update , 2017, Nucleic Acids Res..

[29]  E. Olson,et al.  MED12 regulates a transcriptional network of calcium-handling genes in the heart. , 2017, JCI insight.

[30]  Hao Sun,et al.  MyoD- and FoxO3-mediated hotspot interaction orchestrates super-enhancer activity during myogenic differentiation , 2017, Nucleic acids research.

[31]  E. Lewandowski,et al.  Peroxisome proliferator-activated receptor-α expression induces alterations in cardiac myofilaments in a pressure-overload model of hypertrophy. , 2017, American journal of physiology. Heart and circulatory physiology.

[32]  S. Trappe,et al.  DNA methylation assessment from human slow- and fast-twitch skeletal muscle fibers. , 2017, Journal of applied physiology.

[33]  I. Goldstein,et al.  Transcription factor assisted loading and enhancer dynamics dictate the hepatic fasting response. , 2017, Genome research.

[34]  M. Beltrame,et al.  Nfix Induces a Switch in Sox6 Transcriptional Activity to Regulate MyHC-I Expression in Fetal Muscle , 2016, Cell reports.

[35]  A. Jimeno,et al.  Humanized Mouse Xenograft Models: Narrowing the Tumor-Microenvironment Gap. , 2016, Cancer research.

[36]  K. Gundersen,et al.  Increased hypertrophic response with increased mechanical load in skeletal muscles receiving identical activity patterns. , 2016, American journal of physiology. Cell physiology.

[37]  K. Sakamoto,et al.  Six1 homeoprotein drives myofiber type IIA specialization in soleus muscle , 2016, Skeletal Muscle.

[38]  Stephen R Quake,et al.  Single-cell multimodal profiling reveals cellular epigenetic heterogeneity , 2016, Nature Methods.

[39]  S. Berger,et al.  Epigenetic Mechanisms of Longevity and Aging , 2016, Cell.

[40]  K. Margulies,et al.  Transcription Factor 7-like 2 Mediates Canonical Wnt/&bgr;-Catenin Signaling and c-Myc Upregulation in Heart Failure , 2016, Circulation. Heart failure.

[41]  R. Eskeland,et al.  PIAS1 binds p300 and behaves as a coactivator or corepressor of the transcription factor c-Myb dependent on SUMO-status. , 2016, Biochimica et biophysica acta.

[42]  E. Barrett,et al.  Exercise resistance across the prediabetes phenotypes: Impact on insulin sensitivity and substrate metabolism , 2016, Reviews in Endocrine and Metabolic Disorders.

[43]  C. Stewart,et al.  Does skeletal muscle have an ‘epi’‐memory? The role of epigenetics in nutritional programming, metabolic disease, aging and exercise , 2016, Aging cell.

[44]  Fidel Ramírez,et al.  deepTools2: a next generation web server for deep-sequencing data analysis , 2016, Nucleic Acids Res..

[45]  K. Gundersen Muscle memory and a new cellular model for muscle atrophy and hypertrophy , 2016, Journal of Experimental Biology.

[46]  Y. Ohira,et al.  Differences in histone modifications between slow- and fast-twitch muscle of adult rats and following overload, denervation, or valproic acid administration. , 2015, Journal of applied physiology.

[47]  R. Backofen,et al.  Deciphering the Epigenetic Code of Cardiac Myocyte Transcription. , 2015, Circulation research.

[48]  W. Fan,et al.  PPARs and ERRs: molecular mediators of mitochondrial metabolism. , 2015, Current opinion in cell biology.

[49]  B. Black,et al.  Myocyte enhancer factor 2C function in skeletal muscle is required for normal growth and glucose metabolism in mice , 2015, Skeletal Muscle.

[50]  D. Zheng,et al.  Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice , 2015, Nature.

[51]  C. Glass,et al.  The selection and function of cell type-specific enhancers , 2015, Nature Reviews Molecular Cell Biology.

[52]  Carlo Reggiani,et al.  Single muscle fiber proteomics reveals unexpected mitochondrial specialization , 2015, EMBO reports.

[53]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[54]  R. Blum Activation of Muscle Enhancers by MyoD and Epigenetic Modifiers , 2014, Journal of cellular biochemistry.

[55]  Maureen A. Sartor,et al.  PePr: a peak-calling prioritization pipeline to identify consistent or differential peaks from replicated ChIP-Seq data , 2014, Bioinform..

[56]  J. Zierath,et al.  Dynamic epigenetic responses to muscle contraction. , 2014, Drug discovery today.

[57]  V. Rogozkin,et al.  [Molecular mechanisms of skeletal muscle hypertrophy]. , 2014, Rossiiskii fiziologicheskii zhurnal imeni I.M. Sechenova.

[58]  V. Hakim,et al.  Six Homeoproteins and a linc-RNA at the Fast MYH Locus Lock Fast Myofiber Terminal Phenotype , 2014, PLoS genetics.

[59]  T. Meehan,et al.  An atlas of active enhancers across human cell types and tissues , 2014, Nature.

[60]  R. Irizarry,et al.  Accounting for cellular heterogeneity is critical in epigenome-wide association studies , 2014, Genome Biology.

[61]  K. Gundersen,et al.  A cellular memory mechanism aids overload hypertrophy in muscle long after an episodic exposure to anabolic steroids , 2013, The Journal of physiology.

[62]  S. Fukada,et al.  Isolation, characterization, and molecular regulation of muscle stem cells , 2013, Front. Physiol..

[63]  M. Ramsey,et al.  Insulin responsiveness in metabolic syndrome after eight weeks of cycle training. , 2013, Medicine and science in sports and exercise.

[64]  Stephen C. J. Parker,et al.  Chromatin stretch enhancer states drive cell-specific gene regulation and harbor human disease risk variants , 2013, Proceedings of the National Academy of Sciences.

[65]  Alexandra M. Binder,et al.  Recommendations for the design and analysis of epigenome-wide association studies , 2013, Nature Methods.

[66]  A. Drobek,et al.  LST1/A is a myeloid leukocyte-specific transmembrane adaptor protein recruiting protein tyrosine phosphatases SHP-1 and SHP-2 to the plasma membrane. , 2013, The Journal of Biological Chemistry.

[67]  B. Dynlacht,et al.  The role of MyoD1 and histone modifications in the activation of muscle enhancers , 2013, Epigenetics.

[68]  Rick B. Vega,et al.  Nuclear receptor/microRNA circuitry links muscle fiber type to energy metabolism. , 2013, The Journal of clinical investigation.

[69]  J. Zierath,et al.  Epigenetic flexibility in metabolic regulation: disease cause and prevention? , 2013, Trends in cell biology.

[70]  David A. Orlando,et al.  Master Transcription Factors and Mediator Establish Super-Enhancers at Key Cell Identity Genes , 2013, Cell.

[71]  M. Ramsey,et al.  Slow-twitch fiber proportion in skeletal muscle correlates with insulin responsiveness. , 2013, The Journal of clinical endocrinology and metabolism.

[72]  Sean R. Davis,et al.  NCBI GEO: archive for functional genomics data sets—update , 2012, Nucleic Acids Res..

[73]  D. Srivastava,et al.  A role for RNA post-transcriptional regulation in satellite cell activation , 2012, Skeletal Muscle.

[74]  Shane J. Neph,et al.  Circuitry and Dynamics of Human Transcription Factor Regulatory Networks , 2012, Cell.

[75]  Shane J. Neph,et al.  An expansive human regulatory lexicon encoded in transcription factor footprints , 2012, Nature.

[76]  ENCODEConsortium,et al.  An Integrated Encyclopedia of DNA Elements in the Human Genome , 2012, Nature.

[77]  Lee E. Edsall,et al.  A map of the cis-regulatory sequences in the mouse genome , 2012, Nature.

[78]  A. Drobek,et al.  LST1/A Is a Myeloid Leukocyte-specific Transmembrane Adaptor Protein Recruiting Protein Tyrosine Phosphatases SHP-1 and SHP-2 to the Plasma Membrane* , 2012, The Journal of Biological Chemistry.

[79]  M. Sahin,et al.  Endothelial cell-fatty acid binding protein 4 promotes angiogenesis: role of stem cell factor/c-kit pathway , 2012, Angiogenesis.

[80]  M. Lei,et al.  The same pocket in menin binds both MLL and JUND but has opposite effects on transcription , 2012, Nature.

[81]  Carlo Reggiani,et al.  Fiber types in mammalian skeletal muscles. , 2011, Physiological reviews.

[82]  R. Lieber,et al.  Structure and function of the skeletal muscle extracellular matrix , 2011, Muscle & nerve.

[83]  Kristian Gundersen,et al.  Excitation-transcription coupling in skeletal muscle: the molecular pathways of exercise , 2011, Biological reviews of the Cambridge Philosophical Society.

[84]  Matko Bosnjak,et al.  REVIGO Summarizes and Visualizes Long Lists of Gene Ontology Terms , 2011, PloS one.

[85]  G. Lanfranchi,et al.  Microgenomic Analysis in Skeletal Muscle: Expression Signatures of Individual Fast and Slow Myofibers , 2011, PloS one.

[86]  Jun O. Liu,et al.  Structure of p300 bound to MEF2 on DNA reveals a mechanism of enhanceosome assembly , 2011, Nucleic acids research.

[87]  S. Bernard,et al.  Identification of cardiomyocyte nuclei and assessment of ploidy for the analysis of cell turnover. , 2011, Experimental cell research.

[88]  R. Young,et al.  Histone H3K27ac separates active from poised enhancers and predicts developmental state , 2010, Proceedings of the National Academy of Sciences.

[89]  K. Gundersen,et al.  Myonuclei acquired by overload exercise precede hypertrophy and are not lost on detraining , 2010, Proceedings of the National Academy of Sciences.

[90]  Cory Y. McLean,et al.  GREAT improves functional interpretation of cis-regulatory regions , 2010, Nature Biotechnology.

[91]  Timothy L. Bailey,et al.  Motif Enrichment Analysis: a unified framework and an evaluation on ChIP data , 2010, BMC Bioinformatics.

[92]  Julia A. Lasserre,et al.  Histone modification levels are predictive for gene expression , 2010, Proceedings of the National Academy of Sciences.

[93]  R. Bicknell,et al.  Functionally defining the endothelial transcriptome, from Robo4 to ECSCR. , 2009, Biochemical Society transactions.

[94]  Marta García,et al.  NFAT isoforms control activity-dependent muscle fiber type specification , 2009, Proceedings of the National Academy of Sciences.

[95]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[96]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[97]  C. Pandorf,et al.  Differential Epigenetic Modifications of Histones at the Myosin Heavy Chain Genes in Fast and Slow Skeletal Muscle Fibers and in Response to Muscle Unloading , 2009, American journal of physiology. Cell physiology.

[98]  John K. Hall,et al.  Syndecan-4-expressing muscle progenitor cells in the SP engraft as satellite cells during muscle regeneration. , 2009, Cell stem cell.

[99]  Nathaniel D. Heintzman,et al.  Histone modifications at human enhancers reflect global cell-type-specific gene expression , 2009, Nature.

[100]  M. McKee,et al.  Fibrillin assembly requires fibronectin. , 2008, Molecular biology of the cell.

[101]  Brad T. Sherman,et al.  Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists , 2008, Nucleic acids research.

[102]  W. Farrar,et al.  Cancer stem cells, CD200 and immunoevasion. , 2008, Trends in immunology.

[103]  K. Gundersen,et al.  Nuclear domains during muscle atrophy: nuclei lost or paradigm lost? , 2008, The Journal of physiology.

[104]  Kristian Gundersen,et al.  Activity-dependent repression of muscle genes by NFAT , 2008, Proceedings of the National Academy of Sciences.

[105]  K. Gundersen,et al.  In vivo time-lapse microscopy reveals no loss of murine myonuclei during weeks of muscle atrophy. , 2008, The Journal of clinical investigation.

[106]  Haruhiko Koseki,et al.  Ring1-mediated ubiquitination of H2A restrains poised RNA polymerase II at bivalent genes in mouse ES cells , 2007, Nature Cell Biology.

[107]  E. Olson,et al.  MEF2: a central regulator of diverse developmental programs , 2007, Development.

[108]  D. Hardie,et al.  De‐phosphorylation of MyoD is linking nerve‐evoked activity to fast myosin heavy chain expression in rodent adult skeletal muscle , 2007, The Journal of physiology.

[109]  E. Olson,et al.  Histone deacetylase degradation and MEF2 activation promote the formation of slow-twitch myofibers. , 2007, The Journal of clinical investigation.

[110]  A. Buonanno,et al.  PPARδ expression is influenced by muscle activity and induces slow muscle properties in adult rat muscles after somatic gene transfer , 2007, The Journal of physiology.

[111]  Erik K. Malm,et al.  A Human Protein Atlas for Normal and Cancer Tissues Based on Antibody Proteomics* , 2005, Molecular & Cellular Proteomics.

[112]  K. Esser,et al.  Wnt/β-catenin signaling activates growth-control genes during overload-induced skeletal muscle hypertrophy , 2005 .

[113]  Charlotte Collins,et al.  Direct Isolation of Satellite Cells for Skeletal Muscle Regeneration , 2005, Science.

[114]  J. Holloszy,et al.  A potential link between muscle peroxisome proliferator- activated receptor-alpha signaling and obesity-related diabetes. , 2005, Cell metabolism.

[115]  I. Weissman,et al.  Isolation of Adult Mouse Myogenic Progenitors Functional Heterogeneity of Cells within and Engrafting Skeletal Muscle , 2004, Cell.

[116]  Kiyoshi Kawakami,et al.  Six1 and Eya1 Expression Can Reprogram Adult Muscle from the Slow-Twitch Phenotype into the Fast-Twitch Phenotype , 2004, Molecular and Cellular Biology.

[117]  Michael Kjaer,et al.  Role of extracellular matrix in adaptation of tendon and skeletal muscle to mechanical loading. , 2004, Physiological reviews.

[118]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[119]  Xudong Wei,et al.  MEF2C regulates c‐Jun but not TNF‐α gene expression in stimulated mast cells , 2003 .

[120]  M. Ratajczak,et al.  Expression of Functional CXCR4 by Muscle Satellite Cells and Secretion of SDF‐1 by Muscle‐Derived Fibroblasts is Associated with the Presence of Both Muscle Progenitors in Bone Marrow and Hematopoietic Stem/Progenitor Cells in Muscles , 2003, Stem cells.

[121]  G. Dohm,et al.  Muscle fiber type is associated with obesity and weight loss. , 2002, American journal of physiology. Endocrinology and metabolism.

[122]  J. DiMaio,et al.  Activation of MEF2 by muscle activity is mediated through a calcineurin‐dependent pathway , 2001, The EMBO journal.

[123]  B. Olwin,et al.  Syndecan-3 and syndecan-4 specifically mark skeletal muscle satellite cells and are implicated in satellite cell maintenance and muscle regeneration. , 2001, Developmental biology.

[124]  A. Harel-Bellan,et al.  Interaction between Acetylated MyoD and the Bromodomain of CBP and / or p 300 , 2001 .

[125]  H. Beck-Nielsen,et al.  GLUT4 is reduced in slow muscle fibers of type 2 diabetic patients: is insulin resistance in type 2 diabetes a slow, type 1 fiber disease? , 2001, Diabetes.

[126]  Qing Xu,et al.  The Insulin-like Growth Factor-Phosphatidylinositol 3-Kinase-Akt Signaling Pathway Regulates Myogenin Expression in Normal Myogenic Cells but Not in Rhabdomyosarcoma-derived RD Cells* , 2000, The Journal of Biological Chemistry.

[127]  Lukasz Huminiecki,et al.  In Silico Cloning of Novel Endothelial-Specific Genes , 2000 .

[128]  E. Olson,et al.  MEF2 responds to multiple calcium‐regulated signals in the control of skeletal muscle fiber type , 2000, The EMBO journal.

[129]  T. Lømo,et al.  Fast to slow transformation of denervated and electrically stimulated rat muscle , 1998, The Journal of physiology.

[130]  L. Leinwand,et al.  The vertebrate myosin heavy chain: genetics and assembly properties. , 1997, Cell structure and function.

[131]  L. Kedes,et al.  Molecular mechanisms of myogenic coactivation by p300: direct interaction with the activation domain of MyoD and with the MADS box of MEF2C , 1997, Molecular and cellular biology.

[132]  A. Giordano,et al.  p300 is required for MyoD‐dependent cell cycle arrest and muscle‐specific gene transcription , 1997, The EMBO journal.

[133]  C. Reggiani,et al.  Molecular diversity of myofibrillar proteins: gene regulation and functional significance. , 1996, Physiological reviews.

[134]  S. Tapscott,et al.  Selective accumulation of MyoD and myogenin mRNAs in fast and slow adult skeletal muscle is controlled by innervation and hormones. , 1993, Development.

[135]  J. Changeux,et al.  Interaction of nuclear factors with the upstream region of the alpha‐subunit gene of chicken muscle acetylcholine receptor: variations with muscle differentiation and denervation. , 1989, The EMBO journal.

[136]  T. Lømo,et al.  Slow‐to‐fast transformation of denervated soleus muscles by chronic high‐frequency stimulation in the rat. , 1988, The Journal of physiology.

[137]  T. Eken,et al.  Electrical stimulation resembling normal motor‐unit activity: effects on denervated fast and slow rat muscles. , 1988, The Journal of physiology.

[138]  T. Lømo,et al.  Fibre types, calcium‐sequestering proteins and metabolic enzymes in denervated and chronically stimulated muscles of the rat. , 1988, The Journal of physiology.

[139]  I. Bernstein,et al.  Antigen CD34+ marrow cells engraft lethally irradiated baboons. , 1988, The Journal of clinical investigation.

[140]  R. Moss,et al.  Shortening velocity in single fibers from adult rabbit soleus muscles is correlated with myosin heavy chain composition. , 1985, The Journal of biological chemistry.

[141]  S. Perry,et al.  The isoforms of C protein and their distribution in mammalian skeletal muscle , 1985, Journal of Muscle Research & Cell Motility.

[142]  P. Tesch,et al.  Central and peripheral circulation in relation to muscle-fibre composition in normo- and hyper-tensive man. , 1979, Clinical science.

[143]  H. Schmalbruch,et al.  The number of nuclei in adult rat muscles with special reference to satellite cells , 1977, The Anatomical record.

[144]  David D. McDonald,et al.  Programs , 1984, CL.

[145]  R. Eskeland,et al.  The SUMO protease SENP 1 and the chromatin remodeler , 2018 .

[146]  F. Rossi,et al.  Fibro/Adipogenic Progenitors (FAPs): Isolation by FACS and Culture. , 2017, Methods in molecular biology.

[147]  Brad T. Sherman,et al.  Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources , 2008, Nature Protocols.

[148]  K. Esser,et al.  Wnt/beta-catenin signaling activates growth-control genes during overload-induced skeletal muscle hypertrophy. , 2005, American journal of physiology. Cell physiology.

[149]  Xudong Wei,et al.  MEF2C regulates c-Jun but not TNF-alpha gene expression in stimulated mast cells. , 2003, European journal of immunology.

[150]  Alex E. Lash,et al.  Gene Expression Omnibus: NCBI gene expression and hybridization array data repository , 2002, Nucleic Acids Res..

[151]  E. Olson,et al.  MEF2: a calcium-dependent regulator of cell division, differentiation and death. , 2002, Trends in biochemical sciences.

[152]  K Kato,et al.  Isolation and characterization of CD34+ hematopoietic stem cells from human peripheral blood by high-gradient magnetic cell sorting. , 1993, Cytometry.

[153]  J. Lebacq,et al.  Parvalbumin, labile heat and slowing of relaxation in mouse soleus and extensor digitorum longus muscles. , 1992, The Journal of physiology.