Skip to main content
Log in

Data-Modeling Identifies Conflicting Signaling Axes Governing Myoblast Proliferation and Differentiation Responses to Diverse Ligand Stimuli

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

Abstract

Introduction

Skeletal muscle tissue development and regeneration relies on the proliferation, maturation and fusion of muscle progenitor cells (myoblasts), which arise transiently from muscle stem cells (satellite cells). Following muscle damage, myoblasts proliferate and differentiate in response to temporally-varying inflammatory cytokines, growth factors, and extracellular matrix cues, which stimulate a shared network of intracellular signaling pathways. Here we present an integrated data-modeling approach to elucidate synergies and antagonisms among proliferation and differentiation signaling axes in myoblasts stimulated by regeneration-associated ligands.

Methods

We treated mouse primary myoblasts in culture with combinations of eight regeneration-associated growth factors and cytokines in mixtures that induced additive, synergistic, and antagonistic effects on myoblast proliferation and differentiation responses. For these combinatorial stimuli, we measured the activation dynamics of seven signal transduction pathways using multiplexed phosphoprotein assays and scored proliferation and differentiation responses based on expression of myogenic commitment factors to assemble a cue-signaling-response data compendium. We interrogated the relationship between these signals and responses by partial least-squares (PLS) regression modeling.

Results

Partial least-squares data-modeling accurately predicted response outcomes in cross-validation on the training compendium (cumulative R 2 = 0.96). The PLS model highlighted signaling axes that distinctly govern myoblast proliferation (MEK–ERK, Stat3) and differentiation (JNK) in response to these combinatorial cues, and we confirmed these signal-response associations with small molecule perturbations. Unexpectedly, we observed that a negative feedback circuit involving the phosphatase DUSP6/MKP-3 auto-regulates MEK–ERK signaling in myoblasts.

Conclusion

This data-modeling approach identified conflicting signaling axes that underlie muscle progenitor cell proliferation and 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.

Institutional subscriptions

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

Abbreviations

AUC:

Area-under-the-curve

CSR:

Cue-signal-response

DUSP:

Dual specificity phosphatase

EGF:

Epidermal growth factor

FGF2:

Fibroblast growth factor 2

IGF1:

Insulin-like growth factor 1

IL-1α:

Interleukin-1α

IL-6:

Interleukin-6

LIF:

Leukemia inhibitor factor

MHC:

Myosin heavy chain

OSM:

Oncostatin-M

PC:

Principal component

PLS:

Partial-least squares

TNF-α:

Tumor necrosis factor-α

References

  1. Albeck, J. G., G. MacBeath, F. M. White, P. K. Sorger, D. A. Lauffenburger, and S. Gaudet. Collecting and organizing systematic sets of protein data. Nat. Rev. Mol. Cell Biol. 7(11):803–812, 2006.

    Article  Google Scholar 

  2. Belizario, J. E., C. C. Fontes-Oliveira, J. P. Borges, J. A. Kashiabara, and E. Vannier. Skeletal muscle wasting and renewal: a pivotal role of myokine il-6. Springerplus 5:619, 2016.

    Article  Google Scholar 

  3. Bennett, A. M., and N. K. Tonks. Regulation of distinct stages of skeletal muscle differentiation by mitogen-activated protein kinases. Science 278(5341):1288–1291, 1997.

    Article  Google Scholar 

  4. Bernet, J. D., J. D. Doles, J. K. Hall, K. Kelly Tanaka, T. A. Carter, and B. B. Olwin. P38 mapk signaling underlies a cell-autonomous loss of stem cell self-renewal in skeletal muscle of aged mice. Nat. Med. 20(3):265–271, 2014.

    Article  Google Scholar 

  5. Bliss, C. I. The toxicity of poisins applied jointly. Ann. Appl. Biol. 26(3):585–615, 1939.

    Article  Google Scholar 

  6. Broholm, C., M. J. Laye, C. Brandt, R. Vadalasetty, H. Pilegaard, B. K. Pedersen, and C. Scheele. LIF is a contraction-induced myokine stimulating human myocyte proliferation. J. Appl. Physiol. (1985) 111(1):251–259, 2011.

    Article  Google Scholar 

  7. Cheung, T. H., and T. A. Rando. Molecular regulation of stem cell quiescence. Nat. Rev. Mol. Cell Biol. 14(6):329–340, 2013.

    Article  Google Scholar 

  8. Cosgrove, B. D., L. G. Alexopoulos, T. C. Hang, B. S. Hendriks, P. K. Sorger, L. G. Griffith, and D. A. Lauffenburger. Cytokine-associated drug toxicity in human hepatocytes is associated with signaling network dysregulation. Mol. BioSyst. 6(7):1195–1206, 2010.

    Article  Google Scholar 

  9. Cosgrove, B. D., L. G. Alexopoulos, J. Saez-Rodriguez, L. G. Griffith, and D. A. Lauffenburger. A multipathway phosphoproteomic signaling network model of idiosyncratic drug- and inflammatory cytokine-induced toxicity in human hepatocytes. In: Conf Proc IEEE EMBS, 2009, pp. 5452–5455.

  10. 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(3):255–264, 2014.

    Article  Google Scholar 

  11. Cosgrove, B. D., L. G. Griffith, and D. A. Lauffenburger. Fusing tissue engineering and systems biology toward fulfilling their promise. Cell. Mol. Bioeng. 1(1):33–41, 2008.

    Article  Google Scholar 

  12. Davies, S. P., H. Reddy, M. Caivano, and P. Cohen. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem. J. 351(Pt 1):95–105, 2000.

    Article  Google Scholar 

  13. Deshpande, R. S., and A. A. Spector. Modeling stem cell myogenic differentiation. Sci. Rep. 7:40639, 2017.

    Article  Google Scholar 

  14. Dumont, N. A., C. F. Bentzinger, M. C. Sincennes, and M. A. Rudnicki. Satellite cells and skeletal muscle regeneration. Compr. Physiol. 5(3):1027–1059, 2015.

    Article  Google Scholar 

  15. Fedorov, Y. V., R. S. Rosenthal, and B. B. Olwin. Oncogenic ras-induced proliferation requires autocrine fibroblast growth factor 2 signaling in skeletal muscle cells. J. Cell Biol. 152(6):1301–1305, 2001.

    Article  Google Scholar 

  16. Fu, X., J. Xiao, Y. Wei, S. Li, Y. Liu, J. Yin, K. Sun, H. Sun, H. Wang, Z. Zhang, B. T. Zhang, C. Sheng, H. Wang, and P. Hu. Combination of inflammation-related cytokines promotes long-term muscle stem cell expansion. Cell Res. 25(9):1082–1083, 2015.

    Article  Google Scholar 

  17. Gaudet, S., K. A. Janes, J. G. Albeck, E. A. Pace, D. A. Lauffenburger, and P. K. Sorger. A compendium of signals and responses triggered by prodeath and prosurvival cytokines. Mol. Cell. Proteom. 4(10):1569–1590, 2005.

    Article  Google Scholar 

  18. Heinemann, T., and A. Raue. Model calibration and uncertainty analysis in signaling networks. Curr. Opin. Biotechnol. 39:143–149, 2016.

    Article  Google Scholar 

  19. Janes, K. A. An analysis of critical factors for quantitative immunoblotting. Sci. Signal. 8(371):rs2, 2015.

    Article  Google Scholar 

  20. Janes, K. A., J. G. Albeck, S. Gaudet, P. K. Sorger, D. A. Lauffenburger, and M. B. Yaffe. A systems model of signaling identifies a molecular basis set for cytokine-induced apoptosis. Science 310(5754):1646–1653, 2005.

    Article  Google Scholar 

  21. Janes, K. A., and M. B. Yaffe. Data-driven modelling of signal-transduction networks. Nat. Rev. Mol. Cell Biol. 7(11):820–828, 2006.

    Article  Google Scholar 

  22. Joanisse, S., and G. Parise. Cytokine mediated control of muscle stem cell function. Adv. Exp. Med. Biol. 900:27–44, 2016.

    Article  Google Scholar 

  23. Jones, N. C., Y. V. Fedorov, R. S. Rosenthal, and B. B. Olwin. Erk1/2 is required for myoblast proliferation but is dispensable for muscle gene expression and cell fusion. J. Cell. Physiol. 186(1):104–115, 2001.

    Article  Google Scholar 

  24. Kellogg, R. A., and S. Tay. Noise facilitates transcriptional control under dynamic inputs. Cell 160(3):381–392, 2015.

    Article  Google Scholar 

  25. Kemp, M. L., L. Wille, C. L. Lewis, L. B. Nicholson, and D. A. Lauffenburger. Quantitative network signal combinations downstream of tcr activation can predict il-2 production response. J. Immunol. 178(8):4984–4992, 2007.

    Article  Google Scholar 

  26. Kreeger, P. K. Using partial least squares regression to analyze cellular response data. Sci. Signal. 6(271):tr7, 2013.

    Article  Google Scholar 

  27. Kumar, N., A. Wolf-Yadlin, F. M. White, and D. A. Lauffenburger. Modeling her2 effects on cell behavior from mass spectrometry phosphotyrosine data. PLoS Comput. Biol. 3(1):e4, 2007.

    Article  Google Scholar 

  28. Lagha, M., T. Sato, L. Bajard, P. Daubas, M. Esner, D. Montarras, F. Relaix, and M. Buckingham. Regulation of skeletal muscle stem cell behavior by pax3 and pax7. Cold Spring Harb. Symp. Quant. Biol. 73:307–315, 2008.

    Article  Google Scholar 

  29. Lawlor, M. A., X. Feng, D. R. Everding, K. Sieger, C. E. Stewart, and P. Rotwein. Dual control of muscle cell survival by distinct growth factor-regulated signaling pathways. Mol. Cell. Biol. 20(9):3256–3265, 2000.

    Article  Google Scholar 

  30. Miller-Jensen, K., K. A. Janes, J. S. Brugge, and D. A. Lauffenburger. Common effector processing mediates cell-specific responses to stimuli. Nature 448(7153):604–608, 2007.

    Article  Google Scholar 

  31. Mueck, T., F. Berger, I. Buechsler, R. S. Valchanova, L. Landuzzi, P. L. Lollini, K. Klingel, and B. Munz. Traf6 regulates proliferation and differentiation of skeletal myoblasts. Differentiation 81(2):99–106, 2011.

    Article  Google Scholar 

  32. Munoz-Canoves, P., C. Scheele, B. K. Pedersen, and A. L. Serrano. Interleukin-6 myokine signaling in skeletal muscle: a double-edged sword? FEBS J. 280:4131–4148, 2013.

    Article  Google Scholar 

  33. Nagata, Y., K. Ohashi, E. Wada, Y. Yuasa, M. Shiozuka, Y. Nonomura, and R. Matsuda. Sphingosine-1-phosphate mediates epidermal growth factor-induced muscle satellite cell activation. Exp. Cell Res. 326(1):112–124, 2014.

    Article  Google Scholar 

  34. Ogura, Y., S. M. Hindi, S. Sato, G. Xiong, S. Akira, and A. Kumar. TAK1 modulates satellite stem cell homeostasis and skeletal muscle repair. Nat. Commun. 6:10123, 2015.

    Article  Google Scholar 

  35. Palacios, D., C. Mozzetta, S. Consalvi, G. Caretti, V. Saccone, V. Proserpio, V. E. Marquez, S. Valente, A. Mai, S. V. Forcales, V. Sartorelli, and P. L. Puri. TNF/p38alpha/polycomb signaling to pax7 locus in satellite cells links inflammation to the epigenetic control of muscle regeneration. Cell Stem Cell 7(4):455–469, 2010.

    Article  Google Scholar 

  36. Patterson, K. I., T. Brummer, P. M. O’Brien, and R. J. Daly. Dual-specificity phosphatases: critical regulators with diverse cellular targets. Biochem. J. 418(3):475–489, 2009.

    Article  Google Scholar 

  37. Pawlikowski, B., T. Orion Vogler, K. Gadek, and B. Olwin. Regulation of skeletal muscle stem cells by fibroblast growth factors. Dev. Dyn. 2017. doi:10.1002/dvdy.24495.

    Google Scholar 

  38. Price, F. D., J. von Maltzahn, C. F. Bentzinger, N. A. Dumont, H. Yin, N. C. Chang, D. H. Wilson, J. Frenette, and M. A. Rudnicki. Inhibition of JAK-STAT signaling stimulates adult satellite cell function. Nat. Med. 20(10):1174–1181, 2014.

    Article  Google Scholar 

  39. Puri, P. L., and V. Sartorelli. Regulation of muscle regulatory factors by DNA-binding, interacting proteins, and post-transcriptional modifications. J. Cell. Physiol. 185(2):155–173, 2000.

    Article  Google Scholar 

  40. Rando, T. A., and H. M. Blau. Primary mouse myoblast purification, characterization, and transplantation for cell-mediated gene therapy. J. Cell Biol. 125(6):1275–1287, 1994.

    Article  Google Scholar 

  41. Rudnicki, M. A., F. Le Grand, I. McKinnell, and S. Kuang. The molecular regulation of muscle stem cell function. CSH Symp. Quant. Biol. 73:323–331, 2008.

    Article  Google Scholar 

  42. Serra, C., D. Palacios, C. Mozzetta, S. V. Forcales, I. Morantte, M. Ripani, D. R. Jones, K. Du, U. S. Jhala, C. Simone, and P. L. Puri. Functional interdependence at the chromatin level between the MKK6/p38 and IGF1/PI3K/AKT pathways during muscle differentiation. Mol. Cell 28(2):200–213, 2007.

    Article  Google Scholar 

  43. Tidball, J. G. Regulation of muscle growth and regeneration by the immune system. Nat. Rev. Immunol. 17(3):165–178, 2017.

    Article  Google Scholar 

  44. Tierney, M. T., T. Aydogdu, D. Sala, B. Malecova, S. Gatto, P. L. Puri, L. Latella, and A. Sacco. Stat3 signaling controls satellite cell expansion and skeletal muscle repair. Nat. Med. 20(10):1182–1186, 2014.

    Article  Google Scholar 

  45. Wales, S., S. Hashemi, A. Blais, and J. C. McDermott. Global MEF2 target gene analysis in cardiac and skeletal muscle reveals novel regulation of DUSP6 by p38MAPK-MEF2 signaling. Nucleic Acids Res. 42(18):11349–11362, 2014.

    Article  Google Scholar 

  46. Xiao, F., H. Wang, X. Fu, Y. Li, K. Ma, L. Sun, X. Gao, and Z. Wu. Oncostatin m inhibits myoblast differentiation and regulates muscle regeneration. Cell Res. 21(2):350–364, 2011.

    Article  Google Scholar 

  47. Yin, H., F. Price, and M. A. Rudnicki. Satellite cells and the muscle stem cell niche. Physiol. Rev. 93(1):23–67, 2013.

    Article  Google Scholar 

Download references

Acknowledgments

This work was financially supported by the National Institute on Aging of the National Institutes of Health under Award R00AG042491 (to B.D.C), a US Department of Education Graduate Assistantship in Areas of National Need under Award P200A150273 (to A.M.L), a Roberta G. and John B. DeVries Graduate Fellowship (to A.M.L.), and Hunter R. Rawlings III Cornell Presidential Research Scholarship (to R.F.K. and J.K.). This work made use of the Nanobiotechnology Center (NBTC) shared research facilities at Cornell University. The authors acknowledge technical assistance from Teresa Porri, Penny Burke, Andrea De Micheli, Hilarie Sit, Muhammad Safwan Jalal, Nancy Mejia, Isabella Mercado, Ryan Ausmus, and Paula Fraczek. The authors thank the anonymous reviewers for their constructive reviews.

Animal Studies

All institutional and national guidelines for the care and use of laboratory animals were followed in a protocol approved by Cornell University’s Institutional Animal Care and Use Committee (IACUC).

Conflicts of interest

A. M. Loiben, S. Soueld-Baumgarten, D. Bhattacharya, R. F. Kopyto, J. C. Kim and B. D. Cosgrove declare that they have no conflicts of interest.

Human Studies

No human studies were carried out by the authors for this article.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Benjamin D. Cosgrove.

Additional information

Associate Editor Richard Waugh oversaw the review of this article.

Benjamin D. Cosgrove

is an Assistant Professor in the Meinig School of Biomedical Engineering at Cornell University in Ithaca, NY, where he directs the Laboratory of Regenerative Systems Biology. His research group, which is currently supported by a NIH R00 Pathway-to-Independence Award, develops and implements systems biology and biomaterials engineering approaches to study how cell–cell communication and intracellular signaling networks regulate stem and progenitor cell function in skeletal muscle homeostasis and regeneration, and how these processes become dysfunctional in aging and muscular dystrophies. Dr. Cosgrove earned a Bachelor’s in Biomedical Engineering at the University of Minnesota and a Ph.D. in Bioengineering at the Massachusetts Institute of Technology. His Ph.D. thesis research, under the joint supervision of Dr. Douglas Lauffenburger and Dr. Linda Griffith, which established on experimental and computational systems biology tools to elucidate signaling network mechanisms regulating liver hepatocyte cell-fate decisions, was supported by a Whitaker Foundation Graduate Research Fellowship and a Biomedical Engineering Society Graduate Research Award. His postdoctoral research with Dr. Helen Blau at Stanford University was supported by a Stanford Molecular Imaging Scholars Fellowship and NIH K99 Pathway-to-Independence Award and was recognized by the Cellular and Molecular Bioengineering Special Interest Group of the Biomedical Engineering Society with a Rising Star award in 2015.

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 (PDF 3868 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Loiben, A.M., Soueid-Baumgarten, S., Kopyto, R.F. et al. Data-Modeling Identifies Conflicting Signaling Axes Governing Myoblast Proliferation and Differentiation Responses to Diverse Ligand Stimuli. Cel. Mol. Bioeng. 10, 433–450 (2017). https://doi.org/10.1007/s12195-017-0508-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-017-0508-5

Keywords

Navigation