Skip to main content

Advertisement

Log in

Smooth Muscle Stiffness Sensitivity is Driven by Soluble and Insoluble ECM Chemistry

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

Abstract

Smooth muscle cell (SMC) invasion into plaques and subsequent proliferation is a major factor in the progression of atherosclerosis. During disease progression, SMCs experience major changes in their microenvironment, such as what integrin-binding sites are exposed, the portfolio of soluble factors available, and the elasticity and modulus of the surrounding vessel wall. We have developed a hydrogel biomaterial platform to examine the combined effect of these changes on SMC phenotype. We were particularly interested in how the chemical microenvironment affected the ability of SMCs to sense and respond to modulus. To our surprise, we observed that integrin binding and soluble factors are major drivers of several critical SMC behaviors, such as motility, proliferation, invasion, and differentiation marker expression, and these factors modulated the effect of stiffness on proliferation and migration. Overall, modulus only modestly affected behaviors other than proliferation, relative to integrin binding and soluble factors. Surprisingly, pathological behaviors (proliferation, motility) are not inversely related to SMC marker expression, in direct conflict with previous studies on substrates coupled with single extracellular matrix (ECM) proteins. A high-throughput bead-based ELISA approach and inhibitor studies revealed that differentiation marker expression is mediated chiefly via focal adhesion kinase (FAK) signaling, and we propose that integrin binding and FAK drive the transition from a migratory to a proliferative phenotype. We emphasize the importance of increasing the complexity of in vitro testing platforms to capture these subtleties in cell phenotypes and signaling, in order to better recapitulate important features of in vivo disease and elucidate potential context-dependent therapeutic targets.

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

References

  1. Abou Zeid, N., A.-M. Vallés, and B. Boyer. Serine phosphorylation regulates paxillin turnover during cell migration. Cell Commun. Signal. 4:8, 2006.

    Article  Google Scholar 

  2. Anseth, K. S., C. N. Bowman, and L. Brannon-Peppas. Mechanical properties of hydrogels and their experimental determination. Biomaterials 17(17):1647–1657, 1996.

    Article  Google Scholar 

  3. Arias-Salgado, E. G., S. Lizano, S. Sarkar, J. S. Brugge, M. H. Ginsberg, and S. J. Shattil. Src kinase activation by direct interaction with the integrin beta cytoplasmic domain. Proc. Natl. Acad. Sci. USA 100(23):13298–13302, 2003.

    Article  Google Scholar 

  4. Bellis, S., J. Perrotta, M. Curtis, and C. Turner. Adhesion of fibroblasts to fibronectin stimulates both serine and tyrosine phosphorylation of paxillin. Biochem. J. 381:375–381, 1997.

    Article  Google Scholar 

  5. Bjorkerud, S. Effects of transforming growth factor-beta 1 on human arterial smooth muscle cells in vitro. Arterioscler. Thromb. Vasc. Biol. 11(4):892–902, 1991.

    Article  Google Scholar 

  6. Blank, R. S., and G. K. Owens. Platelet-derived growth factor regulates actin isoform expression and growth state in cultured rat aortic smooth muscle cells. J. Cell. Physiol. 142(3):635–642, 1990.

    Article  Google Scholar 

  7. Bornfeldt, K. E. Intracellular signaling in arterial smooth muscle migration versus proliferation. Trends Cardiovasc. Med. 6(5):143–151, 1996.

    Article  Google Scholar 

  8. Castellot, J. J., L. A. Pukac, B. L. Caleb, T. C. Wright, and M. J. Karnovsky. Heparin selectively inhibits a protein kinase C-dependent mechanism of cell cycle progression in calf aortic smooth muscle cells. J. Cell Biol. 109(6):3147–3155, 1989.

    Article  Google Scholar 

  9. Corjay, M. H., M. M. Thompson, K. R. Lynch, and G. K. Owens. Differential effect of platelet-derived growth factor-versus serum-induced growth on smooth muscle alpha-actin and nonmuscle beta-actin mRNA expression in cultured rat aortic smooth muscle cells. J. Biol. Chem. 264(18):10501–10506, 1989.

    Google Scholar 

  10. De Donatis, A., G. Comito, F. Buricchi, M. C. Vinci, A. Parenti, A. Caselli, G. Camici, G. Manao, G. Ramponi, and P. Cirri. Proliferation versus migration in platelet-derived growth factor signaling: the key role of endocytosis. J. Biol. Chem. 283(29):19948–19956, 2008.

    Article  Google Scholar 

  11. De Donatis, A., F. Ranaldi, and P. Cirri. Reciprocal control of cell proliferation and migration. Cell Commun. Signal. 8:20, 2010.

    Article  Google Scholar 

  12. Desmouliere, A., L. Rubbia-Brandt, and G. Gabbiani. Modulation of actin isoform expression in cultured arterial smooth muscle cells by heparin and culture conditions. Arterioscler. Thromb. Vasc. Biol. 11(2):244–253, 1991.

    Article  Google Scholar 

  13. Engler, A., L. Bacakova, C. Newman, A. Hategan, M. Griffin, and D. Discher. Substrate compliance versus ligand density in cell on gel responses. Biophys. J. 86(1 Pt 1):617–628, 2004.

    Article  Google Scholar 

  14. Gaudet, C., W. A. Marganski, S. Kim, C. T. Brown, V. Gunderia, M. Dembo, and J. Y. Wong. Influence of type I collagen surface density on fibroblast spreading, motility, and contractility. Biophys. J. 85(5):3329–3335, 2003.

    Article  Google Scholar 

  15. Hadjipanayi, E., V. Mudera, and R. A. Brown. Close dependence of fibroblast proliferation on collagen scaffold matrix stiffness. J. Tissue Eng. Regen. Med. 3(2):77–84, 2009.

    Article  Google Scholar 

  16. Hautmann, M. B., C. S. Madsen, and G. K. Owens. A transforming growth factor beta (TGFbeta) control element drives TGFbeta-induced stimulation of smooth muscle alpha-actin gene expression in concert with two CArG elements. J. Biol. Chem. 272(16):10948–10956, 1997.

    Article  Google Scholar 

  17. Hazeltine, L. B., C. S. Simmons, M. R. Salick, X. Lian, M. G. Badur, W. Han, S. M. Delgado, T. Wakatsuki, W. C. Crone, B. L. Pruitt, and S. P. Palecek. Effects of substrate mechanics on contractility of cardiomyocytes generated from human pluripotent stem cells. Int. J. Cell Biol. 2012:508294, 2012.

    Article  Google Scholar 

  18. Herrick, W. G., T. V. Nguyen, M. Sleiman, S. McRae, T. S. Emrick, and S. R. Peyton. PEG-phosphorylcholine hydrogels as tunable and versatile platforms for mechanobiology. Biomacromolecules 14(7):2294–2304, 2013.

    Article  Google Scholar 

  19. Hollenbeck, S. T., H. Itoh, O. Louie, P. L. Faries, B. Liu, and K. C. Kent. Type I collagen synergistically enhances PDGF-induced smooth muscle cell proliferation through pp60src-dependent crosstalk between the alpha2beta1 integrin and PDGF beta receptor. Biochem. Biophys. Res. Commun. 325(1):328–337, 2004.

    Article  Google Scholar 

  20. Holycross, B. J., R. S. Blank, M. M. Thompson, M. J. Peach, and G. K. Owens. Platelet-derived growth factor-BB-induced suppression of smooth muscle cell differentiation. Circ. Res. 71(6):1525–1532, 1992.

    Article  Google Scholar 

  21. Ishigaki, T., K. Imanaka-Yoshida, N. Shimojo, S. Matsushima, W. Taki, and T. Yoshida. Tenascin-C enhances crosstalk signaling of integrin αvβ3/PDGFR-β complex by SRC recruitment promoting PDGF-induced proliferation and migration in smooth muscle cells. J. Cell. Physiol. 226(10):2617–2624, 2011.

    Article  Google Scholar 

  22. Janat, M. F., W. S. Argraves, and G. Liau. Regulation of vascular smooth muscle cell integrin expression by transforming growth factor beta1 and by platelet-derived growth factor-BB. J. Cell. Physiol. 151(3):588–595, 1992.

    Article  Google Scholar 

  23. Jones, J. I., T. Prevette, A. Gockerman, and D. R. Clemmons. Ligand occupancy of the alpha-V-beta3 integrin is necessary for smooth muscle cells to migrate in response to insulin-like growth factor. Proc. Natl. Acad. Sci. USA 93(6):2482–2487, 1996.

    Article  Google Scholar 

  24. Li, S., S. Sims, Y. Jiao, L. H. Chow, and J. G. Pickering. Evidence from a novel human cell clone that adult vascular smooth muscle cells can convert reversibly between noncontractile and contractile phenotypes. Circ. Res. 85(4):338–348, 1999.

    Article  Google Scholar 

  25. Lindner, V., N. E. Olson, A. W. Clowes, and M. A. Reidy. Inhibition of smooth muscle cell proliferation in injured rat arteries. Interaction of heparin with basic fibroblast growth factor. J. Clin. Invest. 90(5):2044–2049, 1992.

    Article  Google Scholar 

  26. Louis, S. F., and P. Zahradka. Vascular smooth muscle cell motility: from migration to invasion. Exp. Clin. Cardiol. 15(4):e75–e85, 2010.

    Google Scholar 

  27. Macri, L., D. Silverstein, and R. A. F. Clark. Growth factor binding to the pericellular matrix and its importance in tissue engineering. Adv. Drug Deliv. Rev. 59(13):1366–1381, 2007.

    Article  Google Scholar 

  28. Mann, B., and J. West. Cell adhesion peptides alter smooth muscle cell adhesion, proliferation, migration, and matrix protein synthesis on modified surfaces and in polymer scaffolds. J. Biomed. Mater. Res. 60(1):86–93, 2002.

    Article  Google Scholar 

  29. McCaffrey, T. A., D. J. Falcone, C. F. Brayton, L. A. Agarwal, F. G. Welt, and B. B. Weksler. Transforming growth factor-beta activity is potentiated by heparin via dissociation of the transforming growth factor-beta/alpha 2-macroglobulin inactive complex. J. Cell Biol. 109(1):441–448, 1989.

    Article  Google Scholar 

  30. Mitra, S. K., and D. D. Schlaepfer. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr. Opin. Cell Biol. 18(5):516–523, 2006.

    Article  Google Scholar 

  31. Nelson, P. R., S. Yamamura, and K. C. Kent. Extracellular matrix proteins are potent agonists of human smooth muscle cell migration. J. Vasc. Surg. 24(1):25–32, 1996.

    Article  Google Scholar 

  32. Nguyen, T. V., M. Sleiman, T. Moriarty, W. G. Herrick, and S. R. Peyton. Sorafenib resistance and JNK signaling in carcinoma during extracellular matrix stiffening. Biomaterials 35(22):5749–5759, 2014.

    Article  Google Scholar 

  33. Peyton, S. R., P. D. Kim, C. M. Ghajar, D. Seliktar, and A. J. Putnam. The effects of matrix stiffness and RhoA on the phenotypic plasticity of smooth muscle cells in a 3-D biosynthetic hydrogel system. Biomaterials 29(17):2597–2607, 2008.

    Article  Google Scholar 

  34. Peyton, S. R., and A. J. Putnam. Extracellular matrix rigidity governs smooth muscle cell motility in a biphasic fashion. J. Cell. Physiol. 204(1):198–209, 2005.

    Article  Google Scholar 

  35. Peyton, S. R., C. B. Raub, V. P. Keschrumrus, and A. J. Putnam. The use of poly(ethylene glycol) hydrogels to investigate the impact of ECM chemistry and mechanics on smooth muscle cells. Biomaterials 27(28):4881–4893, 2006.

    Article  Google Scholar 

  36. Pirone, D. M., W. F. Liu, S. A. Ruiz, L. Gao, S. Raghavan, C. A. Lemmon, L. H. Romer, and C. S. Chen. An inhibitory role for FAK in regulating proliferation: a link between limited adhesion and RhoA-ROCK signaling. J. Cell Biol. 174(2):277–288, 2006.

    Article  Google Scholar 

  37. Rensen, S. S. M., P. A. F. M. Doevendans, and G. J. J. M. van Eys. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth. Heart J. 15(3):100–108, 2007.

    Article  Google Scholar 

  38. Sazonova, O. V., B. C. Isenberg, J. Herrmann, K. L. Lee, A. Purwada, A. D. Valentine, J. A. Buczek-Thomas, J. Y. Wong, and M. A. Nugent. Extracellular matrix presentation modulates vascular smooth muscle cell mechanotransduction. Matrix Biol. 41:36–43, 2015.

    Article  Google Scholar 

  39. Shi, Z. D., G. Abraham, and J. M. Tarbell. Shear stress modulation of smooth muscle cell marker genes in 2-D and 3-D depends on mechanotransduction by heparan sulfate proteoglycans and ERK1/2. PLoS One 5(8):e12196, 2010.

    Article  Google Scholar 

  40. Shull, K. R., D. Ahn, W.-L. Chen, C. M. Flanigan, and A. J. Crosby. Axisymmetric adhesion tests of soft materials. Macromol. Chem. Phys. 199(4):489–511, 1998.

    Article  Google Scholar 

  41. Skaletz-Rorowski, A., A. Schmidt, G. Breithardt, and E. Buddecke. Heparin-induced overexpression of basic fibroblast growth factor, basic fibroblast growth factor receptor, and cell-associated proteoheparan sulfate in cultured coronary smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 16(8):1063–1069, 1996.

    Article  Google Scholar 

  42. Stegemann, J. P., and R. M. Nerem. Altered response of vascular smooth muscle cells to exogenous biochemical stimulation in two- and three-dimensional culture. Exp. Cell Res. 283(2):146–155, 2003.

    Article  Google Scholar 

  43. Stephan, S., S. G. Ball, M. Williamson, D. V. Bax, A. Lomas, C. A. Shuttleworth, and C. M. Kielty. Cell-matrix biology in vascular tissue engineering. J. Anat. 209(4):495–502, 2006.

    Article  Google Scholar 

  44. Ucuzian, A. A., L. P. Brewster, A. T. East, Y. Pang, A. A. Gassman, and H. P. Greisler. Characterization of the chemotactic and mitogenic response of SMCs to PDGF-BB and FGF-2 in fibrin hydrogels. J. Biomed. Mater. Res. Part A 94(3):988–996, 2010.

    Google Scholar 

  45. van Eys, G. J., P. M. Niessen, and S. S. Rensen. Smoothelin in vascular smooth muscle cells. Trends Cardiovasc. Med. 17(1):26–30, 2007.

    Article  Google Scholar 

  46. Winder, S. J., B. G. Allen, E. D. Fraser, H. M. Kang, G. J. Kargacin, and M. P. Walsh. Calponin phosphorylation in vitro and in intact muscle. Biochem. J. 296:827–836, 1993.

    Article  Google Scholar 

  47. Wong, J. Y., A. Velasco, P. Rajagopalan, and Q. Pham. Directed movement of vascular smooth muscle cells on gradient-compliant hydrogels. Langmuir 19(9):1908–1913, 2003.

    Article  Google Scholar 

  48. Worth, N. F., B. E. Rolfe, J. Song, and G. R. Campbell. Vascular smooth muscle cell phenotypic modulation in culture is associated with reorganisation of contractile and cytoskeletal proteins. Cell Motil. Cytoskelet. 49(3):130–145, 2001.

    Article  Google Scholar 

  49. Zheng, B., C. Duan, and D. R. Clemmons. The effect of extracellular matrix proteins on porcine smooth muscle cell insulin-like growth factor (IGF) binding protein-5 synthesis and responsiveness to IGF-I. J. Biol. Chem. 273:8994–9000, 1998.

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by a Grant in Aid from the American Heart Association (13GRNT16190013), a Barry and Afsaneh Siadat Career Development Award, grants from the National Science Foundation and the National Cancer Institute (DMR-1234852 and DMR-1304724), start-up funds from the University of Massachusetts Amherst, and the UMass MRSEC on Polymers (DMR-0820506). WGH was supported by a fellowship from the Institute of Cellular Engineering IGERT at UMass (DGE-0654128). SRP is a Pew Biomedical Scholar supported by the Pew Charitable Trusts. We are also grateful to Dr. Nele Van Dessel for helpful discussions.

Conflict of interests

William G. Herrick, Shruti Rattan, Thuy V. Nguyen, Michael S. Grunwald, Christopher W. Barney, Alfred J. Crosby, and Shelly R. Peyton declare no conflicts of interest. No human studies were carried out by the authors for this article. No animal studies were carried out by the authors for this article.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shelly R. Peyton.

Additional information

Associate Editor Cynthia A. Reinhart-King oversaw the review of this article.

This article is part of the 2015 Young Innovators Issue.

Shelly R. Peyton is the Barry and Afsaneh Siadat Assistant Professor of Chemical Engineering at the University of Massachusetts, Amherst. She received her B.S. in Chemical Engineering from Northwestern University in 2002 and went on to obtain her MS and Ph.D. in Chemical Engineering from the University of California, Irvine. She was then an NIH Kirschstein post-doctoral fellow in the Biological Engineering department at MIT before starting her academic appointment at UMass in 2011. Her research interests are in biomaterial design and understanding how cell-material interactions contribute to cancer aggressiveness, cardiovascular disease progression, and regenerative medicine. Since arriving at UMass she has been named a Pew Biomedical Scholar, received a New Innovator Award from the NIH, and she was recently awarded a CAREER grant from the NSF.

figure a

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 65 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Herrick, W.G., Rattan, S., Nguyen, T.V. et al. Smooth Muscle Stiffness Sensitivity is Driven by Soluble and Insoluble ECM Chemistry. Cel. Mol. Bioeng. 8, 333–348 (2015). https://doi.org/10.1007/s12195-015-0397-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-015-0397-4

Keywords

Navigation