Skip to main content
Log in

Tethered Jagged-1 Synergizes with Culture Substrate Stiffness to Modulate Notch-Induced Myogenic Progenitor Differentiation

  • Published:
Cellular and Molecular Bioengineering Aims and scope Submit manuscript

Abstract

Introduction

Notch signaling is amongst the key intrinsic mechanisms regulating satellite cell fate, promoting the transition of activated satellite cells to highly proliferative myogenic progenitor cells and preventing their premature differentiation. Although much is known about the biochemical milieu that drives myogenic progression, less is known about the spatial cues providing spatiotemporal control of skeletal muscle repair in the context of Notch signaling.

Methods

Using a murine injury model, we quantified in vivo biophysical changes that occur within the skeletal muscle during regeneration. Employing tunable poly(ethylene glycol)—based hydrogel substrates, we modeled the measured changes in bulk stiffness in the context of Notch ligand signaling, which are present in the regenerative milieu at the time of injury.

Results

Following injury, there is a transient increase in the bulk stiffness of the tibialis anterior muscle that may be explained in part by changes in extracellular matrix deposition. When presented to primary myoblasts, Jagged-1, Jagged-2, and Dll1 in a tethered format elicited greater degrees of Notch activity compared to their soluble form. Only tethered Jagged-1 effects were tuned by substrate stiffness, with the greatest Notch activation observed on stiff hydrogels matching the stiffness of regenerating muscle. When exposed to tethered Jagged-1 on stiff hydrogels, fewer primary myoblasts expressed myogenin, and pharmacological inhibitor studies suggest this effect is Notch and RhoA dependent.

Conclusion

Our study proposes that tethered Jagged-1 presented in the context of transient tissue stiffening serves to tune Notch activity in myogenic progenitors during skeletal muscle repair and delay differentiation.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1
Figure 2
Figure 3
Figure 4

Similar content being viewed by others

Abbreviations

bFGF:

Basic fibroblast growth factor

Dll1:

Delta-like 1

DM:

Differentiation media

ECM:

Extracellular matrix

GM:

Growth media

IgG1:

Immunoglobulin G subtype 1

Jag1:

Jagged-1

Jag2:

Jagged-2

MyoG:

Myogenin

NICD:

Notch intracellular domain

PEG:

Polyethylene glycol

PBS:

Phosphate buffered saline

ROCK:

Rho-associated, coiled-coil containing protein kinase

TBST:

Tris-buffered saline plus Tween-20

References

  1. Bjornson, C. R. R., T. H. Cheung, L. Liu, P. V. Tripathi, K. M. Steeper, and T. A. Rando. Notch signaling is necessary to maintain quiescence in adult muscle stem cells. Stem Cells 30:232–242, 2012.

    Article  Google Scholar 

  2. Blau, H. M., and C. Webster. Isolation and characterization of human muscle cells. Proc. Natl. Acad. Sci. USA. 78:5623–5627, 1981.

    Article  Google Scholar 

  3. Buas, M. F., and T. Kadesch. Regulation of skeletal myogenesis by Notch. Exp. Cell Res. 316:3028–3033, 2010.

    Article  Google Scholar 

  4. Calve, S., J. Isaac, J. P. Gumucio, and C. L. Mendias. Hyaluronic acid, HAS1, and HAS2 are significantly upregulated during muscle hypertrophy. Am. J. Physiol. Cell Physiol. 303:C577–C588, 2012.

    Article  Google Scholar 

  5. Conboy, I. M., and T. A. Rando. The regulation of Notch signaling controls satellite cell activation and cell fate determination in postnatal myogenesis. Dev. Cell 3:397–409, 2002.

    Article  Google Scholar 

  6. Cordey, M., M. Limacher, S. Kobel, V. Taylor, and M. P. Lutolf. Enhancing the reliability and throughput of neurosphere culture on hydrogel microwell arrays. Stem Cells 26:2586–2594, 2008.

    Article  Google Scholar 

  7. Cosgrove, B. D., P. M. Gilbert, E. Porpiglia, F. Mourkioti, S. P. Lee, S. Y. Corbel, M. E. Llewellyn, S. L. Delp, and H. M. Blau. Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat. Med. 20:255–264, 2014.

    Article  Google Scholar 

  8. Davoudi, S., and P. M. Gilbert. Optimization of satellite cell culture through biomaterials. Methods. Mol. Biol. 1556:329–341, 2017.

    Article  Google Scholar 

  9. Engler, A. J., M. A. Griffin, S. Sen, C. G. Bönnemann, H. L. Sweeney, and D. E. Discher. Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. J. Cell Biol. 166:877–887, 2004.

    Article  Google Scholar 

  10. Engler, A. J., S. Sen, H. L. Sweeney, and D. E. Discher. Matrix elasticity directs stem cell lineage specification. Cell 126:677–689, 2006.

    Article  Google Scholar 

  11. Faux, C. H., A. M. Turnley, R. Epa, R. Cappai, and P. F. Bartlett. Interactions between growth factors and Notch regulate neuronal differentiation. J Neurosci. 21:5587–5596, 2001.

    Google Scholar 

  12. Gerdin, B., and R. Hällgren. Dynamic role of hyaluronan (HYA) in connective tissue activation and inflammation. J. Intern. Med. 242:49–55, 1997.

    Article  Google Scholar 

  13. Gilbert, P. M., K. L. Havenstrite, K. E. G. Magnusson, A. Sacco, N. A. Leonardi, P. Kraft, N. K. Nguyen, S. Thrun, M. P. Lutolf, and H. M. Blau. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 329:1078–1081, 2010.

    Article  Google Scholar 

  14. Gordon, W. R., D. Vardar-Ulu, G. Histen, C. Sanchez-Irizarry, J. C. Aster, and S. C. Blacklow. Structural basis for autoinhibition of Notch. Nat Struct Mol Biol 14:295–300, 2007.

    Article  Google Scholar 

  15. Gordon, W. R., B. Zimmerman, L. He, L. J. Miles, J. Huang, K. Tiyanont, D. G. McArthur, J. C. Aster, N. Perrimon, J. J. Loparo, and S. C. Blacklow. Mechanical allostery: evidence for a force requirement in the proteolytic activation of Notch. Dev. Cell 33:729–736, 2015.

    Article  Google Scholar 

  16. Griffith, L. G., and M. A. Swartz. Capturing complex 3D tissue physiology in vitro. Nat Rev Mol Cell Biol 7:211–224, 2006.

    Article  Google Scholar 

  17. Günther, S., J. Kim, S. Kostin, C. Lepper, C. M. Fan, and T. Braun. Myf5-positive satellite cells contribute to Pax7-dependent long-term maintenance of adult muscle stem cells. Cell Stem Cell 13:590–601, 2013.

    Article  Google Scholar 

  18. Hisada, Y., M. Yasunaga, S. Hanaoka, S. Saijou, T. Sugino, A. Tsuji, T. Saga, K. Tsumoto, S. Manabe, J. Kuroda, J. Kuratsu, and Y. Matsumura. Discovery of an uncovered region in fibrin clots and its clinical significance. Sci. Rep. 3:2604, 2013.

    Article  Google Scholar 

  19. Jahnsen, E. D., A. Trindade, H. C. Zaun, S. Lehoux, A. Duarte, and E. A. V. Jones. Notch1 is pan-endothelial at the onset of flow and regulated by flow. PLoS ONE 10:1–14, 2015.

    Article  Google Scholar 

  20. Jiang, W. R., G. Cady, M. M. Hossain, Q. Q. Huang, X. Wang, and J. P. Jin. Mechanoregulation of h2-calponin gene expression and the role of notch signaling. J. Biol. Chem. 289:1617–1628, 2014.

    Article  Google Scholar 

  21. Kondoh, K., K. Sunadome, and E. Nishida. Notch signaling suppresses p38 MAPK activity via induction of MKP-1 in myogenesis. J. Biol. Chem. 282:3058–3065, 2007.

    Article  Google Scholar 

  22. Kuhl, P. R., and L. G. Griffith-Cima. Tethered epidermal growth factor as a paradigm for growth factor–induced stimulation from the solid phase. Nat. Med. 2:1022–1027, 1996.

    Article  Google Scholar 

  23. Lepper, C., S. J. Conway, and C.-M. Fan. Adult satellite cells and embryonic muscle progenitors have distinct genetic requirements. Nature 460:627–631, 2009.

    Article  Google Scholar 

  24. Lutolf, M. P., R. Doyonnas, K. Havenstrite, K. Koleckar, and H. M. Blau. Perturbation of single hematopoietic stem cell fates in artificial niches. Integr Biol 1:59–69, 2009.

    Article  Google Scholar 

  25. Lutolf, M. P., and J. A. Hubbell. Synthesis and physicochemical characterization of end-linked poly(ethylene glycol)-co-peptide hydrogels formed by Michael-type addition. Biomacromolecules 4:713–722, 2003.

    Article  Google Scholar 

  26. Mauro, A., and W. R. Adams. The structure of the sarcolemma of the frog skeletal muscle fiber. J. Biophys. Biochem. Cytol. 10:177–185, 1961.

    Article  Google Scholar 

  27. Morrissey, J. B., R. Y. Cheng, S. Davoudi, and P. M. Gilbert. Biomechanical origins of muscle stem cell signal transduction. J. Mol. Biol. 428:1441–1454, 2015.

    Article  Google Scholar 

  28. Quarta, M., J. O. Brett, R. DiMarco, A. De Morree, S. C. Boutet, R. Chacon, M. C. Gibbons, V. A. Garcia, J. Su, J. B. Shrager, S. Heilshorn, and T. A. Rando. An artificial niche preserves the quiescence of muscle stem cells and enhances their therapeutic efficacy. Nat. Biotechnol. 34:752–759, 2016.

    Article  Google Scholar 

  29. Seo, D., K. M. Southard, J. Kim, J. Cheon, Z. J. Gartner, Y. Jun, D. Seo, K. M. Southard, J. Kim, H. J. Lee, J. Farlow, and J. Lee. A mechanogenetic toolkit for interrogating cell signaling in space and time. Cell 165:1507–1518, 2016.

    Article  Google Scholar 

  30. Tierney, M. T., and A. Sacco. Inducing and Evaluating Skeletal Muscle Injury by Notexin and Barium Chloride. Methods in Molecular Biology, Clifton: Springer, 2016, pp. 53–60.

    Google Scholar 

  31. Trensz, F., F. Lucien, V. Couture, T. Söllrald, G. Drouin, A.-J. Rouleau, M. Grandbois, G. Lacraz, and G. Grenier. Increased microenvironment stiffness in damaged myofibers promotes myogenic progenitor cell proliferation. Skelet. Muscle 5:5, 2015.

    Article  Google Scholar 

  32. Tsivitse, S. Notch and Wnt signaling, physiological stimuli and postnatal myogenesis. Int. J. Biol. Sci. 6:268–281, 2010.

    Article  Google Scholar 

  33. Tu, J., Y. Li, and Z. Hu. Notch1 and 4 signaling responds to an increasing vascular wall shear stress in a rat model of arteriovenous malformations. Biomed. Res. Int. 2014. doi:10.1155/2014/368082.

    Google Scholar 

  34. Urciuolo, A., M. Quarta, V. Morbidoni, F. Gattazzo, S. Molon, P. Grumati, F. Montemurro, F. S. Tedesco, B. Blaauw, G. Cossu, G. Vozzi, T. A. Rando, and P. Bonaldo. Collagen VI regulates satellite cell self-renewal and muscle regeneration. Nat. Commun. 4:1964, 2013.

    Article  Google Scholar 

  35. Varnum-Finney, B., L. Wu, M. Yu, C. Brashem-Stein, S. Staats, D. Flowers, J. D. Griffin, and I. D. Bernstein. Immobilization of Notch ligand, Delta-1, is required for induction of notch signaling. J. Cell Sci. 113(Pt 23):4313–4318, 2000.

    Google Scholar 

  36. Vieira, N. M., I. Elvers, M. S. Alexander, Y. B. Moreira, A. Eran, J. P. Gomes, J. L. Marshall, E. K. Karlsson, S. Verjovski-Almeida, K. Lindblad-Toh, L. M. Kunkel, and M. Zatz. Jagged 1 rescues the duchenne muscular dystrophy phenotype. Cell 163:1204–1213, 2015.

    Article  Google Scholar 

  37. von Maltzahn, J., N. C. Chang, C. F. Bentzinger, and M. A. Rudnicki. Wnt signaling in myogenesis. Trends Cell Biol. 22:602–609, 2012.

    Article  Google Scholar 

  38. von Maltzahn, J., A. E. Jones, R. J. Parks, and M. A. Rudnicki. Pax7 is critical for the normal function of satellite cells in adult skeletal muscle. Proc. Natl. Acad. Sci. USA 110:16474–16479, 2013.

    Article  Google Scholar 

  39. Wang, X., and T. Ha. Defining single molecular forces required to activate integrin and notch signaling. Science 340:991–994, 2013.

    Article  Google Scholar 

  40. Yin, H., F. Price, and M. A. Rudnicki. Satellite cells and the muscle stem cell niche. Physiol. Rev. 93:23–67, 2013.

    Article  Google Scholar 

  41. Zhu, J.-H., C.-L. Chen, S. Flavahan, J. Harr, B. Su, and N. A. Flavahan. Cyclic stretch stimulates vascular smooth muscle cell alignment by redox-dependent activation of Notch3. Am. J. Physiol. Heart Circ. Physiol. 300:H1770–H1780, 2011.

    Article  Google Scholar 

Download references

Acknowledgments

We are grateful to Dr. Yasuhiro Matsumura (National Cancer Center Hospital, Kashiwa City, Japan) for providing us with his custom antibody to analyze fibrin clot formation in our study.

Author Contributions

HS, MAB, CAS, and PMG conceived the study and designed experiments. HS, SD, RYC, AJM, and EWL performed experiments, analyzed data, performed statistical analyses, and prepared figures. HS, MAB, SD, RYC, AJM, EWL, CAS, and PMG wrote, assembled, and revised the manuscript. All authors reviewed and approved the submission.

Funding

This study was funded by the Natural Sciences and Engineering Research Council (USRA fellowship to H.S., CREATE ToEP fellowship to S.D., RGPIN 327627-06 to C.A.S., RGPIN 435724-13 and Canada Research Chair 950-231201 to P.M.G.); Toronto Musculoskeletal Centre (Graduate Scholarships to M.A.B. and R.Y.C.); Barbara and Frank Milligan Foundation (R.Y.C.); Ontario Provincial Government (OGS-visa to M.A.B., 31390 and ER15-11-073 to P.M.G.); Canada Foundation for Innovation (31390 to P.M.G.); Krembil Foundation (Scholarship to M.A.B.); Toronto Western Arthritis Program (to P.M.G.); and Canadian Institutes of Health Research (MOP-302041 to C.A.S. and ONM-137370 to P.M.G.)

Conflict of interest

All authors declared that they have no conflict of interest.

Ethical Approval

All applicable international, national, and/or institutional guidelines for the care and use of animals were followed. This article does not contain any studies with human participants performed by any of the authors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Penney M. Gilbert.

Additional information

Associate Editor Alyssa Panitch oversaw the review of this article.

Penney Gilbert is an Assistant Professor at the University of Toronto in the Institute of Biomaterials and Biomedical Engineering. She holds cross-appointments in the Department of Biochemistry and the Donnelly Centre for Cellular and Biomolecular Research. Gilbert received her PhD from the University of Pennsylvania under the mentorship of Valerie Weaver and then pursued Postdoctoral studies with Helen Blau at Stanford University. Her research team is focused on engineering human skeletal muscle culture models and uncovering niche cues that drive muscle stem cell fate changes in vivo. She is recipient of an Ontario Early Researcher Award and a BMES-CMBE Conference ‘Rising Star’ Award. She holds a Tier II Canada Research Chair in Endogenous Repair.

figure a

This article is part of the 2017 CMBE Young Innovators special issue.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 469 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Safaee, H., Bakooshli, M.A., Davoudi, S. et al. Tethered Jagged-1 Synergizes with Culture Substrate Stiffness to Modulate Notch-Induced Myogenic Progenitor Differentiation. Cel. Mol. Bioeng. 10, 501–513 (2017). https://doi.org/10.1007/s12195-017-0506-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-017-0506-7

Keywords

Navigation