Quiescence of human muscle stem cells is favored by culture on natural biopolymeric films

Claire Monge, Nicholas DiStasio, Thomas Rossi, Muriel Sébastien, Hiroshi Sakai, Benoit Kalman, Thomas Boudou, Shahragim Tajbakhsh, Isabelle Marty, Anne Bigot, Vincent Mouly, Catherine Picart, Claire Monge, Nicholas DiStasio, Thomas Rossi, Muriel Sébastien, Hiroshi Sakai, Benoit Kalman, Thomas Boudou, Shahragim Tajbakhsh, Isabelle Marty, Anne Bigot, Vincent Mouly, Catherine Picart

Abstract

Background: Satellite cells are quiescent resident muscle stem cells that present an important potential to regenerate damaged tissue. However, this potential is diminished once they are removed from their niche environment in vivo, prohibiting the long-term study and genetic investigation of these cells. This study therefore aimed to provide a novel biomaterial platform for the in-vitro culture of human satellite cells that maintains their stem-like quiescent state, an important step for cell therapeutic studies.

Methods: Human muscle satellite cells were isolated from two donors and cultured on soft biopolymeric films of controlled stiffness. Cell adhesive phenotype, maintenance of satellite cell quiescence and capacity for gene manipulation were investigated using FACS, western blotting, fluorescence microscopy and electron microscopy.

Results: About 85% of satellite cells cultured in vitro on soft biopolymer films for 3 days maintained expression of the quiescence marker Pax7, as compared with 60% on stiffer films and 50% on tissue culture plastic. The soft biopolymeric films allowed satellite cell culture for up to 6 days without renewing the media. These cells retained their stem-like properties, as evidenced by the expression of stem cell markers and reduced expression of differentiated markers. In addition, 95% of cells grown on these soft biopolymeric films were in the G0/G1 stage of the cell cycle, as opposed to those grown on plastic that became activated and began to proliferate and differentiate.

Conclusions: Our study identifies a new biomaterial made of a biopolymer thin film for the maintenance of the quiescence state of muscle satellite cells. These cells could be activated at any point simply by replating them onto a plastic culture dish. Furthermore, these cells could be genetically manipulated by viral transduction, showing that this biomaterial may be further used for therapeutic strategies.

Keywords: Biomimetism; Culture platform; Layer by layer; Polymeric biomaterial; Quiescence; Satellite cells.

Figures

Fig. 1
Fig. 1
SC morphology on biopolymeric films in comparison with plastic. Biopolymeric films were crosslinked at different levels: EDC10, EDC50 and EDC70. a Representative images of actin staining after 1 day (D1) or 3 days of culture (D3) on EDC10, EDC50 and EDC70 films and plastic. Scale bars: 50 μm. b Corresponding quantification of the cell spreading area. Data plotted from three independent experiments (n = 200 cells minimum per experimental condition). **p < 0.01. c Scanning electron microscopy imaging of SCs cultured on EDC10 films, EDC70 films and plastic. The white arrows indicate filopodia. Scale bars: 30 μm
Fig. 2
Fig. 2
SC morphology and ECM protein secretion on biopolymeric films. EDC10 and EDC70 films were compared with plastic. a Representative pictures of actin, fibronectin and collagen I staining after culture for 3, 10 and 21 days (respectively D3, D10 and D21) on EDC10 films, and for 3 days on EDC70 films and plastic. Scale bar: 50 μm. b Quantification of fibronectin and collagen I secretion by SCs cultured on EDC10 films (D3, D10, D21), EDC70 films and plastic (D3). Data are mean ± SD from two independent experiments (n = 100 cells minimum per condition). ***p <0.001. c Scanning electron microscopy micrographs of SCs seeded on EDC10 films for 10 days. Scale bars: 30 μm
Fig. 3
Fig. 3
Pax7 and myogenin expression in SCs cultured on biopolymeric films. a Fluorescent labeling of actin, Pax7, myogenin (MyoG) and nucleus (Hoechst) at D3 on EDC10 and EDC70 films in comparison with plastic. Scale bar: 50 μm. bPax7 and c MyoG expression were quantified by immunofluorescence at D1 and D3 in GM. Data from three independent experiments (n = 100 cells minimum per experimental condition). *p < 0.05, ***p < 0.001. d Western blot analysis of Pax7 and MyoG expression and semi-quantitative determination of ePax7 and f MyoG expression as compared with plastic control (the condition “plastic D1” was set to 1). Results of D6 and D10 are only given for the culture on EDC10 films because cell confluence was reached on EDC70 films and plastic as soon as day 3; for these conditions, D6 and D10 are thus referred to as “not applicable” (NA). Data are mean ± SD from three independent experiments. D1, D3, D6, D10 days 1, 3, 6, 10
Fig. 4
Fig. 4
Absence of SC proliferation on EDC10 films up to 10 days. a Quantification of SC proliferation by EdU assay: for D1, D3, D6 and D10 on EDC10 films and only for D1 and D3 on plastic, because confluence was reached (referred as “not applicable” (NA)). Data are box plots from three independent experiments (n = 200 cells in total per experimental condition). ***p < 0.001. b FACS analysis of the cell cycle at D3, D6 and D10 on EDC10 films and at D1 on plastic. Data plotted from two independent experiments (n = 5000 cells per condition). c Western blot of cyclin D1 and semi-quantitative analysis. The condition “plastic D1” was taken as the reference (value set to 1). Data are mean ± SD from three independent experiments. D1, D3, D6, D10 days 1, 3, 6, 10, EdU 5-ethynyl-2′-deoxyuridine
Fig. 5
Fig. 5
Reactivation of cells cultured on EDC10 films. a Schematic of experiment where SCs were precultured on EDC10 films or plastic for 3 days and then replated onto plastic. b Proliferation was assessed by EdU incorporation and c differentiation by the number of MHC+ cells after 3 days in DM. Data are mean ± SD from two different experiments (n = 50 cells per condition in each experiment). d Schematic of experiment where SCs were precultured either on EDC10 films or plastic for 3 days. SCs were then replated on EDC10 films and plastic for another 3 days. The cells were fixed and stained for actin and Pax7. e Representative images of actin staining. Scale bar: 50 μm. f Number of Pax7-positive cells: after the preculture (at D0), on EDC10 films and plastic, after replating for 3 days and depending on the preculture conditions (EDC10 films or plastic). Data are mean ± SD from three independent experiments (n = 100 cells). ***p < 0.001. D–3, D0, D3, days –3, 0, 3, EdU 5-ethynyl-2′-deoxyuridine, MHC skeletal myosin heavy chain
Fig. 6
Fig. 6
Transduction of SCs by lentivirus. a Confocal images of SC-transduced lentivirus GFP (MOI = 120) after 1 day of culture on EDC10 films or plastic. Scale bar: 100 μm. b Quantification of the fluorescence intensity on EDC10 films or plastic, measured 24 h after transduction and 24 h after replating on plastic (following a preculture on EDC10 films). *p < 0.05

References

    1. Schultz E, McCormick KM. Skeletal muscle satellite cells. Rev Physiol Biochem Pharmacol. 1994;123:213–57. doi: 10.1007/BFb0030904.
    1. Charge SBP, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev. 2004;84:209–38. doi: 10.1152/physrev.00019.2003.
    1. Hawke TJ, Garry DJ. Myogenic satellite cells: physiology to molecular biology. J Appl Physiol. 2001;91:534–51.
    1. Peault B, Rudnicki M, Torrente Y, Cossu G, Tremblay JP, Partridge T, Gussoni E, Kunkel LM, Huard J. Stem and progenitor cells in skeletal muscle development, maintenance, and therapy. Mol Ther. 2007;15:867–77. doi: 10.1038/mt.sj.6300145.
    1. Seale P, Sabourin LA, Girgis-Gabardo A, Mansouri A, Gruss P, Rudnicki MA. Pax7 is required for the specification of myogenic satellite cells. Cell. 2000;102:777–86. doi: 10.1016/S0092-8674(00)00066-0.
    1. Latil M, Rocheteau P, Chatre L, Sanulli S, Memet S, Ricchetti M, Tajbakhsh S, Chretien F. Skeletal muscle stem cells adopt a dormant cell state post mortem and retain regenerative capacity. Nat Commun. 2012;3:12. doi: 10.1038/ncomms1890.
    1. Negroni E, Gidaro T, Bigot A, Butler-Browne G, Mouly V, Trollet C. Stem cells and muscle diseases: advances in cell therapy strategies. Neuropathol Appl Neurobiol. 2015;41:270-87.
    1. Camirand G, Rousseau J, Ducharme ME, Rothstein DM, Tremblay JP. Novel Duchenne muscular dystrophy treatment through myoblast transplantation tolerance with anti-CD45RB, anti-CD154 and mixed chimerism. Am J Transplant. 2004;4:1255–65. doi: 10.1111/j.1600-6143.2004.00501.x.
    1. Tedesco F, Cossu G. Stem cell therapies for muscle disorders. Curr Opin Neurol. 2012;25:597–603. doi: 10.1097/WCO.0b013e328357f288.
    1. Perie S, Mamchaoui K, Mouly V, Blot S, Bouazza B, Thornell LE, St Guily JL, Butler-Browne G. Premature proliferative arrest of cricopharyngeal myoblasts in oculo-pharyngeal muscular dystrophy: therapeutic perspectives of autologous myoblast transplantation. Neuromuscul Disord. 2006;16:770–81. doi: 10.1016/j.nmd.2006.07.022.
    1. Perie S, Trollet C, Mouly V, Vanneaux V, Mamchaoui K, Bouazza B, Marolleau JP, Laforet P, Chapon F, Eymard B, Butler-Browne G, Larghero J, St Guily JL. Autologous myoblast transplantation for oculopharyngeal muscular dystrophy: a phase I/IIa clinical study. Mol Ther. 2014;22:219–25. doi: 10.1038/mt.2013.155.
    1. Aziz A, Sebastian S, Dilworth F. The origin and fate of muscle satellite cells. Stem Cell Rev. 2012;8:609–22. doi: 10.1007/s12015-012-9352-0.
    1. Gilbert PM, Havenstrite KL, Magnusson KEG, Sacco A, Leonardi NA, Kraft P, Nguyen NK, Thrun S, Lutolf MP, Blau HM. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science. 2010;329:1078–81. doi: 10.1126/science.1191035.
    1. Moyle LA, Zammit PS. Isolation, culture and immunostaining of skeletal muscle fibres to study myogenic progression in satellite cells. Methods Mol Biol. 2014;1210:63–78. doi: 10.1007/978-1-4939-1435-7_6.
    1. Sousa-Victor P, Gutarra S, Garcia-Prat L, Rodriguez-Ubreva J, Ortet L, Ruiz-Bonilla V, Jardi M, Ballestar E, Gonzalez S, Serrano AL, Perdiguero E, Munoz-Canoves P. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature. 2014;506:316–21. doi: 10.1038/nature13013.
    1. Snow MH. The effects of aging on satellite cells in skeletal muscles of mice and rats. Cell Tissue Res. 1977;185:399–408. doi: 10.1007/BF00220299.
    1. Kuang S, Gillespie MA, Rudnicki MA. Niche regulation of muscle satellite cell self-renewal and differentiation. Cell Stem Cell. 2008;2:22–31. doi: 10.1016/j.stem.2007.12.012.
    1. Pannerec A, Marazzi G, Sassoon D. Stem cells in the hood: the skeletal muscle niche. Trends Mol Med. 2012;18:599–606. doi: 10.1016/j.molmed.2012.07.004.
    1. Motohashi N, Asakura Y, Asakura A. Isolation, culture, and transplantation of muscle satellite cells. J Vis Exp. 2014. 86.
    1. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126:677–89. doi: 10.1016/j.cell.2006.06.044.
    1. Bentzinger CF, Wang YX, von Maltzahn J, Soleimani VD, Yin H, Rudnicki MA, Rudnicki A. Fibronectin regulates Wnt7a signaling and satellite cell expansion. Cell Stem Cell. 2013;12:75–87. doi: 10.1016/j.stem.2012.09.015.
    1. Trappmann B, Gautrot JE, Connelly JT, Strange DG, Li Y, Oyen ML, Cohen Stuart MA, Boehm H, Li B, Vogel V, Spatz JP, Watt FM, Huck WT. Extracellular-matrix tethering regulates stem-cell fate. Nat Mater. 2012;11:642–9. doi: 10.1038/nmat3339.
    1. Chaudhuri O, Koshy ST, da Cunha CB, Shin JW, Verbeke CS, Allison KH, Mooney DJ. Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat Mater. 2014;13:970–8. doi: 10.1038/nmat4009.
    1. Wen JH, Vincent LG, Fuhrmann A, Choi YS, Hribar KC, Taylor-Weiner H, Chen SC, Engler AJ. Interplay of matrix stiffness and protein tethering in stem cell differentiation. Nat Mater. 2014;13:979–87. doi: 10.1038/nmat4051.
    1. Danoviz M, Yablonka-Reuveni Z. Skeletal muscle satellite cells: background and methods for isolation and analysis in a primary culture system. Methods Mol Biol. 2012;798:21–52. doi: 10.1007/978-1-61779-343-1_2.
    1. Calve S, Odelberg SJ, Simon HG. A transitional extracellular matrix instructs cell behavior during muscle regeneration. Dev Biol. 2010;344:259–71. doi: 10.1016/j.ydbio.2010.05.007.
    1. Lukjanenko L, Jung MJ, Hegde N, Perruisseau-Carrier C, Migliavacca E, Rozo M, Karaz S, Jacot G, Schmidt M, Li L, Metairon S, Raymond F, Lee U, Sizzano F, Wilson DH, Dumont NA, Palini A, Fassler R, Steiner P, Descombes P, Rudnicki MA, Fan CM, von Maltzahn J, Feige JN, Bentzinger CF. Loss of fibronectin from the aged stem cell niche affects the regenerative capacity of skeletal muscle in mice. Nat Med. 2016;22:897–905. doi: 10.1038/nm.4126.
    1. Tierney MT, Gromova A, Sesillo FB, Sala D, Spenle C, Orend G, Sacco A. Autonomous extracellular matrix remodeling controls a progressive adaptation in muscle stem cell regenerative capacity during development. Cell Rep. 2016;14:1940–52. doi: 10.1016/j.celrep.2016.01.072.
    1. Engler AJ, Griffin MA, Sen S, Bonnetnann CG, Sweeney HL, Discher DE. Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. J Cell Biol. 2004;166:877–87. doi: 10.1083/jcb.200405004.
    1. Gribova V, Auzely-Velty R, Picart C. Polyelectrolyte multilayer assemblies on materials surfaces: from cell adhesion to tissue engineering. Chem Mat. 2012;24:854–69. doi: 10.1021/cm2032459.
    1. Ren K, Crouzier T, Roy C, Picart C. Polyelectrolyte multilayer films of controlled stiffness modulate myoblast cell differentiation. Adv Funct Mater. 2008;18:1378–89. doi: 10.1002/adfm.200701297.
    1. Monge C, Ren K, Berton K, Guillot R, Peyrade D, Picart C. Engineering muscle tissues on microstructured polyelectrolyte multilayer films. Tissue Eng A. 2012;18:1664–76. doi: 10.1089/ten.tea.2012.0079.
    1. Schneider A, Francius G, Obeid R, Schwinte P, Hemmerle J, Frisch B, Schaaf P, Voegel JC, Senger B, Picart C. Polyelectrolyte multilayers with a tunable Young’s modulus: influence of film stiffness on cell adhesion. Langmuir. 2006;22:1193–200. doi: 10.1021/la0521802.
    1. Boudou T, Crouzier T, Auzely-Velty R, Glinel K, Picart C. Internal composition versus the mechanical properties of polyelectrolyte multilayer films: the influence of chemical cross-linking. Langmuir. 2009;25:13809–19. doi: 10.1021/la9018663.
    1. Boudou T, Crouzier T, Nicolas C, Ren K, Picart C. Polyelectrolyte multilayer nanofilms used as thin materials for cell mechano-sensitivity studies. Macromol Biosci. 2011;11:77–89. doi: 10.1002/mabi.201000301.
    1. Bigot A, Klein AF, Gasnier E, Jacquemin V, Ravassard P, Butler-Browne G, Mouly V, Furling D. Large CTG repeats trigger p16-dependent premature senescence in myotonic dystrophy type 1 muscle precursor cells. Am J Pathol. 2009;174:1435–42. doi: 10.2353/ajpath.2009.080560.
    1. Crouzier T, Fourel L, Boudou T, Albiges-Rizo C, Picart C. Presentation of BMP-2 from a soft biopolymeric film unveils its activity on cell adhesion and migration. Adv Mater. 2011;23:H111–8. doi: 10.1002/adma.201004637.
    1. Pozarowski P, Darzynkiewicz Z. Analysis of cell cycle by flow cytometry. Methods Mol Biol. 2004;281:301–11.
    1. Mann CJ, Perdiguero E, Kharraz Y, Aguilar S, Pessina P, Serrano AL, Munoz-Canoves P. Aberrant repair and fibrosis development in skeletal muscle. Skelet Muscle. 2011;1:21. doi: 10.1186/2044-5040-1-21.
    1. Alexakis C, Partridge T, Bou-Gharios G. Implication of the satellite cell in dystrophic muscle fibrosis: a self-perpetuating mechanism of collagen overproduction. Am J Phys Cell Phys. 2007;293:C661–9. doi: 10.1152/ajpcell.00061.2007.
    1. Zammit PS, Relaix F, Nagata Y, Ruiz AP, Collins CA, Partridge TA, Beauchamp JR. Pax7 and myogenic progression in skeletal muscle satellite cells. J Cell Sci. 2006;119:1824–32. doi: 10.1242/jcs.02908.
    1. Yin H, Price F, Rudnicki MA. Satellite cells and the muscle stem cell niche. Physiol Rev. 2013;93:23–67. doi: 10.1152/physrev.00043.2011.
    1. Yuan J, Tang ZL, Yang SL, Li K. CRABP2 promotes myoblast differentiation and is modulated by the transcription factors MyoD and Sp1 in C2C12 cells. Plos One. 2013;8:9. doi: 10.1371/annotation/20cbe947-8308-4bcc-8fcb-d77c4dc96a78.
    1. Yang K, Hitomi M, Stacey DW. Variations in cyclin D1 levels through the cell cycle determine the proliferative fate of a cell. Cell Div. 2006;1:8. doi: 10.1186/1747-1028-1-32.
    1. Montarras D, L’Honore A, Buckingham M. Lying low but ready for action: the quiescent muscle satellite cell. Febs J. 2013;280:4036–50. doi: 10.1111/febs.12372.
    1. Rodgers JT, King KY, Brett JO, Cromie MJ, Charville GW, Maguire KK, Brunson C, Mastey N, Liu L, Tsai CR, Goodell MA, Rando TA. mTORC1 controls the adaptive transition of quiescent stem cells from G(0) to G(Alert) Nature. 2014;510:393–6.
    1. Tang AH, Rando TA. Induction of autophagy supports the bioenergetic demands of quiescent muscle stem cell activation. Embo J. 2014;33:2782–97. doi: 10.15252/embj.201488278.
    1. Quarta M, Brett JO, DiMarco R, De Morree A, Boutet SC, Chacon R, Gibbons MC, Garcia VA, Su J, Shrager JB, Heilshorn S, Rando TA. An artificial niche preserves the quiescence of muscle stem cells and enhances their therapeutic efficacy. Nat Biotechnol. 2016;34:752–9. doi: 10.1038/nbt.3576.
    1. Fukada SI, Uezumi A, Ikemoto M, Masuda S, Segawa M, Tanimura N, Yamamoto H, Miyagoe-Suzuki Y, Takedaa S. Molecular signature of quiescent satellite cells in adult skeletal muscle. Stem Cells. 2007;25:2448–59. doi: 10.1634/stemcells.2007-0019.
    1. Otto A, Collins-Hooper H, Patel A, Dash PR, Patel K. Adult skeletal muscle stem cell migration is mediated by a blebbing/amoeboid mechanism. Rejuv Res. 2011;14:249–60. doi: 10.1089/rej.2010.1151.
    1. Pelham RJ, Wang YL. Cell locomotion and focal adhesions are regulated by substrate flexibility. Proc Natl Acad Sci U S A. 1997;94:13661–5. doi: 10.1073/pnas.94.25.13661.
    1. Collinsworth AM, Torgan CE, Nagda SN, Rajalingam RJ, Kraus WE, Truskey GA. Orientation and length of mammalian skeletal myocytes in response to a unidirectional stretch. Cell Tissue Res. 2000;302:243–51. doi: 10.1007/s004410000224.
    1. Sellathurai J, Cheedipudi S, Dhawan J, Schroder HD. A novel in vitro model for studying quiescence and activation of primary isolated human myoblasts. Plos One. 2013;8:16. doi: 10.1371/journal.pone.0064067.
    1. Collins CA, Olsen I, Zammit PS, Heslop L, Petrie A, Partridge TA, Morgan JE. Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell. 2005;122:289–301. doi: 10.1016/j.cell.2005.05.010.
    1. Han WM, Jang YC, Garcia AJ. Engineered matrices for skeletal muscle satellite cell engraftment and function. Matrix Biol. 2016. [Epub ahead of print]

Source: PubMed

3
Sottoscrivi