Skip to main content

Advertisement

Log in

Regulation of Macrophages by Extracellular Matrix Composition and Adhesion Geometry

  • Published:
Regenerative Engineering and Translational Medicine Aims and scope Submit manuscript

Abstract

The extracellular matrix is a dynamic structural component of tissue and plays a key role in wound healing by providing adhesive cues that regulate cell behavior during tissue repair. Macrophages are essential regulators of inflammation and tissue remodeling, and adaptively change their function in different microenvironments. Although much is known about how soluble factors including cytokines and chemokines influence immune cell function, much less is known about how insoluble cues including those presented by the matrix regulate their behavior. The goal of this study is to understand the potential role of different adhesive proteins and their geometric presentation in regulating macrophage behavior. Previously, using micropatterning and topographical features, we observed that macrophage elongation helps to promote polarization towards a pro-healing phenotype. In this work, we found that adhesion to different extracellular matrix ligands had only a moderate effect on macrophage cytokine secretion in response to prototypical activating stimuli. However, expression of arginase-1, a marker of pro-healing phenotype, was enhanced when cells were cultured on laminin, Matrigel, and vitronectin when compared to collagen, fibronectin, or fibrinogen. When micropatterned into lines, almost all matrix ligands allowed elongation of macrophages and a concomitant increase in arginase-1 expression. Together, these data demonstrate that extracellular matrix composition and adhesion geometry influence macrophage cell shape and function.

Lay Summary

In this study, we examined the effects of different extracellular matrix proteins on the phenotypic polarization of macrophages, a major innate immune cell involved in defense against pathogens, wound healing, and progression of many diseases. Our results suggest that the cytokine secretion response of macrophages to inflammatory or wound-healing stimuli was largely independent of the type of adhesion protein on which they were cultured, although laminin, Matrigel, and vitronectin promoted the expression of the pro-healing marker arginase-1. Interestingly, these matrix proteins are all prevalent in tumor environments, where pro-healing macrophages are often observed. Macrophages forced to elongate on patterned substrates of nearly all ECMs increased their expression of arginase-1. The extracellular matrix is dynamic in structure and composition during healing after injury and progression of many diseases including tissue fibrosis, cancer, or cardiovascular disease. This work may provide insight to how adhesion to different matrix environments and geometries regulates macrophages, and their impact on disease.

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.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496(7446):445–55. https://doi.org/10.1038/nature12034.

    Article  CAS  Google Scholar 

  2. Anderson JM, Rodriguez A, Chang DT. Foreign body reaction to biomaterials. Semin Immunol. 2008;20(2):86–100. https://doi.org/10.1016/j.smim.2007.11.004.

    Article  CAS  Google Scholar 

  3. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13. https://doi.org/10.12703/P6-13.

    Article  CAS  Google Scholar 

  4. Yu X, Buttgereit A, Lelios I, Utz SG, Cansever D, Becher B, et al. The cytokine TGF-beta promotes the development and homeostasis of alveolar macrophages. Immunity. 2017;47(5):903–12 e4. https://doi.org/10.1016/j.immuni.2017.10.007.

    Article  CAS  Google Scholar 

  5. Clark RAF, DellaPelle P, Manseau E, Lanigan JM, Dvorak HF, Colvin RB. Blood vessel fibronectin increases in conjunction with endothelial cell proliferation and capillary ingrowth during wound healing. J Investig Dermatol. 1982;79(5):269–76. https://doi.org/10.1111/1523-1747.ep12500076.

    Article  CAS  Google Scholar 

  6. O’Toole EA. Extracellular matrix and keratinocyte migration. Clin Exp Dermatol. 2001;26(6):525–30. https://doi.org/10.1046/j.1365-2230.2001.00891.x.

    Article  Google Scholar 

  7. Leight JL, Wozniak MA, Chen S, Lynch ML, Chen CS. Matrix rigidity regulates a switch between TGF-β1-induced apoptosis and epithelial-mesenchymal transition. Mol Biol Cell. 2012;23:781–91. https://doi.org/10.1091/mbc.E11-06-0537.

    Article  CAS  Google Scholar 

  8. Hinz B. The extracellular matrix and transforming growth factor-beta1: tale of a strained relationship. Matrix Biol. 2015;47:54–65. https://doi.org/10.1016/j.matbio.2015.05.006.

    Article  CAS  Google Scholar 

  9. Duffy RM, Sun Y, Feinberg AW. Understanding the role of ECM protein composition and geometric micropatterning for engineering human skeletal muscle. Ann Biomed Eng. 2016;44(6):2076–89. https://doi.org/10.1007/s10439-016-1592-8.

    Article  Google Scholar 

  10. Frangogiannis NG. The extracellular matrix in myocardial injury, repair, and remodeling. J Clin Invest. 2017;127(5):1600–12. https://doi.org/10.1172/JCI87491.

    Article  Google Scholar 

  11. Jennewein C, Tran N, Paulus P, Ellinghaus P, Eble JA, Zacharowski K. Novel aspects of fibrin(ogen) fragments during inflammation. Mol Med. 2011;17(5–6):568–73. https://doi.org/10.2119/molmed.2010.00146.

    Article  CAS  Google Scholar 

  12. Chow S, Di Girolamo N. Vitronectin: a migration and wound healing factor for human corneal epithelial cells. Invest Ophthalmol Vis Sci. 2014;55(10):6590–600. https://doi.org/10.1167/iovs.14-15054.

    Article  CAS  Google Scholar 

  13. Barker TH, Engler AJ. The provisional matrix: setting the stage for tissue repair outcomes. Matrix Biol. 2017;60-61:1–4. https://doi.org/10.1016/j.matbio.2017.04.003.

    Article  CAS  Google Scholar 

  14. Giussani M, Merlino G, Cappelletti V, Tagliabue E, Daidone MG. Tumor-extracellular matrix interactions: identification of tools associated with breast cancer progression. Semin Cancer Biol. 2015;35:3–10. https://doi.org/10.1016/j.semcancer.2015.09.012.

    Article  CAS  Google Scholar 

  15. Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 2014;15:1243–53. https://doi.org/10.15252/embr.201439246.

    Article  CAS  Google Scholar 

  16. Slattery ML, John E, Torres-Mejia G, Stern M, Lundgreen A, Hines L, et al. Matrix metalloproteinase genes are associated with breast cancer risk and survival: the Breast Cancer Health Disparities Study. PLoS One. 2013;8(5):e63165. https://doi.org/10.1371/journal.pone.0063165.

    Article  CAS  Google Scholar 

  17. Sorensen HT, Friis S, Olsen JH, Thulstrup AM, Mellemkjaer L, Linet M, et al. Risk of liver and other types of cancer in patients with cirrhosis: a nationwide cohort study in Denmark. Hepatology. 1998;28(4):921–5.

    Article  CAS  Google Scholar 

  18. Neglia JP, Fitzsimmons SC, Maisonneuve P, Schoni MH, Schoni-Affolter F, Corey M, et al. The risk of cancer among patients with cystic fibrosis. NEJM. 1995;332(8):494–9.

    Article  CAS  Google Scholar 

  19. Du J, Wang Y, Jia L. ECM and atherosclerosis, in Atherosclerosis: risks, mechanisms, and therapies. In: Wang H, Patterson C, editors. Atherosclerosis: risks, mechanisms, and therapies. 1st ed. Hoboken: John Wiley & Sons, Inc; 2015. p. 343–51.

    Google Scholar 

  20. RTA M, MGA E, JPM C, MJE K, PHM S, Merkx M, et al. Imaging collagen in intact viable healthy and atherosclerotic arteries using fluorescently labeled CNA35 and two-photon laser scanning microscopy. Mol Imaging. 2007;6(4):247–60. https://doi.org/10.2310/7290.2007.00021.

    Article  CAS  Google Scholar 

  21. Wesley RB II, Meng X, Godin D, Galis ZS. Extracellular matrix modulates macrophage functions characteristic to atheroma: collagen type I enhances acquisition of resident macrophage traits by human peripheral blood monocytes in vitro. Arterioscler Thromb Vasc Biol. 1998;18(3):432–40.

    Article  CAS  Google Scholar 

  22. Armstrong JW, Chapes SK. Effects of extracellular matrix proteins on macrophage differentiation, growth, and function: comparison of liquid and agar culture systems. J Exp Zool. 1994;269(3):178–87. https://doi.org/10.1002/jez.1402690303.

    Article  CAS  Google Scholar 

  23. Toral D, Zaveri JSL, Dolgova NV, Clare-Salzler MJ, Keselowsky BG. Integrin-directed modulation of macrophage responses to biomaterials. Biomaterials. 2014;35:3504–15. https://doi.org/10.1016/j.biomaterials.2014.01.007.

    Article  Google Scholar 

  24. McWhorter FY, Wang T, Nguyen P, Chung T, Liu WF. Modulation of macrophage phenotype by cell shape. PNAS. 2013;110(43):17253–8. https://doi.org/10.1073/pnas.1308887110.

    Article  Google Scholar 

  25. Wang T, Luu TU, Chen A, Khine M, Liu WF. Topographical modulation of macrophage phenotype by shrink-film multi-scale wrinkles. Biomater Sci. 2016;4(6):948–52. https://doi.org/10.1039/c6bm00224b.

    Article  CAS  Google Scholar 

  26. Luu TU, Gott SC, Woo BWK, Rao MP, Liu WF. Micro- and nanopatterned topographical cues for regulating macrophage cell shape and phenotype. ACS Appl Mater Interfaces. 2015;7:28665–72. https://doi.org/10.1021/acsami.5b10589.

    Article  CAS  Google Scholar 

  27. Waldo SW, Li Y, Buono C, Zhao B, Billings EM, Chang J, et al. Heterogeneity of human macrophages in culture and in atherosclerotic plaques. Am J Pathol. 2008;172(4):1112–26. https://doi.org/10.2353/ajpath.2008.070513.

    Article  Google Scholar 

  28. Neubrand VE, Pedreno M, Caro M, Forte-Lago I, Delgado M, Gonzalez-Rey E. Mesenchymal stem cells induce the ramification of microglia via the small RhoGTPases Cdc42 and Rac1. Glia. 2014;62(12):1932–42. https://doi.org/10.1002/glia.22714.

    Article  Google Scholar 

  29. Chen S, Jones JA, Xu Y, Low HY, Anderson JM, Leong KW. Characterization of topographical effects on macrophage behavior in a foreign body response model. Biomaterials. 2010;31(13):3479–91. https://doi.org/10.1016/j.biomaterials.2010.01.074.

    Article  CAS  Google Scholar 

  30. Biswas S, Bachay G, Chu J, Hunter DD, Brunken WJ. Laminin-dependent interaction between astrocytes and microglia: a role in retinal angiogenesis. Am J Pathol. 2017;187(9):2112–27. https://doi.org/10.1016/j.ajpath.2017.05.016.

    Article  CAS  Google Scholar 

  31. Fridman R, Giaccone G, Kanemoto T, Martin GR, Gazdar AF, Mulshine JL. Reconstituted basement membrane (matrigel) and laminin can enhance the tumorgenicity and the drug resistance of small cell lung cancer cell lines. Proc Natl Acad Sci U S A. 1990;87:6698–702.

    Article  CAS  Google Scholar 

  32. Hao W, Zhang X, Xiu B, Yang X, Hu S, Liu Z, et al. Vitronectin: a promising breast cancer serum biomarker for early diagnosis of breast cancer in patients. Tumour Biol. 2016;37(7):8909–16. https://doi.org/10.1007/s13277-015-4750-y.

    Article  CAS  Google Scholar 

  33. Murata K, Motayama T, Kotake C. Collagen types in various layers of the human aorta and their changes with the atherosclerotic process. Atherosclerosis. 1986;60:251–62. https://doi.org/10.1016/0021-9150(86)90172-3.

    Article  CAS  Google Scholar 

  34. Stary HC, Chandler AB, Dinsmore RE, Fuster V, Glagov S, Insull W, et al. A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis. Arterioscler Thromb Vasc Biol. 1995;15:1512–31. https://doi.org/10.1161/01.CIR.92.5.1355.

    Article  CAS  Google Scholar 

  35. Shekhonin BV, Domogatsky SP, Idelson GL, Koteliansky VE, Rukosuev VS. Relative distribution of fibronectin and type I, III, IV, V collagens in normal and atherosclerotic intima of human arteries. Atherosclerosis. 1987;67:9–16. https://doi.org/10.1016/0021-9150(87)90259-0.

    Article  CAS  Google Scholar 

  36. Colin S, Chinetti-Gbaguidi G, Staels B. Macrophage phenotypes in atherosclerosis. Immmunol Rev. 2014;262:153–66.

    Article  CAS  Google Scholar 

  37. Chinetti-Gbaguidi G, Colin S, Staels B. Macrophage subsets in atherosclerosis. Nat Rev Cardiol. 2015;12(1):10–7. https://doi.org/10.1038/nrcardio.2014.173.

    Article  CAS  Google Scholar 

  38. Mirza R, DiPietro LA, Koh TJ. Selective and specific macrophage ablation is detrimental to wound healing in mice. Am J Pathol. 2009;175(6):2454–62. https://doi.org/10.2353/ajpath.2009.090248.

    Article  CAS  Google Scholar 

  39. Xue M, Jackson CJ. Extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv Wound Care (New Rochelle). 2015;4(3):119–36. https://doi.org/10.1089/wound.2013.0485.

    Article  Google Scholar 

  40. Gauglitz GG, Korting HC, Pavicic T, Ruzicka T, Jeschke MG. Hypertrophic scarring and keloids: pathomechanisms and current and emerging treatment strategies. Mol Med. 2011;17(1–2):113–25. https://doi.org/10.2119/molmed.2009.00153.

    Article  CAS  Google Scholar 

  41. Hsieh JY, Smith TD, Meli VS, Tran TN, Botvinick EL, Liu WF. Differential regulation of macrophage inflammatory activation by fibrin and fibrinogen. Acta Biomater. 2017;47:14–24. https://doi.org/10.1016/j.actbio.2016.09.024.

    Article  CAS  Google Scholar 

  42. Blakney AK, Swartzlander MD, Bryant SJ. The effects of substrate stiffness on the in vitro activation of macrophages and in vivo host response to poly(ethylene glycol)-based hydrogels. J Biomed Mater Res A. 2012;100(6):1375–86. https://doi.org/10.1002/jbm.a.34104.

    Article  CAS  Google Scholar 

  43. Swartzlander MD, Lynn AD, Blakney AK, Kyriakides TR, Bryant SJ. Understanding the host response to cell-laden poly(ethylene glycol)-based hydrogels. Biomaterials. 2013;34(4):952–64. https://doi.org/10.1016/j.biomaterials.2012.10.037.

    Article  CAS  Google Scholar 

  44. Previtera ML, Sengupta A. Substrate stiffness regulates proinflammatory mediator production through TLR4 activity in macrophages. PLoS One. 2015;10(12):e0145813. https://doi.org/10.1371/journal.pone.0145813.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Praveen Krishna Veerasubramanian and Linda McCarthy for technical assistance in the laboratory.

Funding

This work was supported by the National Institutes of Health (NIH) National Institute of Dental and Craniofacial Research (NIDCR) Grant DP2DE023319 and National Institute of Allergy and Infectious Diseases (NIAID) Grant 1R21AI128519-01A1.. T. U. L. was supported by a California Institute of Regenerative Medicine (CIRM) Training Fellowship (TG2-01152).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wendy F. Liu.

Ethics declarations

All protocols involving animals were approved by University of California Irvine’s Institutional Animal Care and Use Committee, which is accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care International (AAALACi).

Electronic supplementary material

Table S1

(PDF 414 kb)

Table S2

(PDF 638 kb)

Supplemental Methods

(PDF 65.2 kb)

Figure S1

(PDF 822 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luu, T.U., Liu, W.F. Regulation of Macrophages by Extracellular Matrix Composition and Adhesion Geometry. Regen. Eng. Transl. Med. 4, 238–246 (2018). https://doi.org/10.1007/s40883-018-0065-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40883-018-0065-z

Keywords

Navigation