Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Cell–extracellular matrix mechanotransduction in 3D

Abstract

Mechanical properties of extracellular matrices (ECMs) regulate essential cell behaviours, including differentiation, migration and proliferation, through mechanotransduction. Studies of cell–ECM mechanotransduction have largely focused on cells cultured in 2D, on top of elastic substrates with a range of stiffnesses. However, cells often interact with ECMs in vivo in a 3D context, and cell–ECM interactions and mechanisms of mechanotransduction in 3D can differ from those in 2D. The ECM exhibits various structural features as well as complex mechanical properties. In 3D, mechanical confinement by the surrounding ECM restricts changes in cell volume and cell shape but allows cells to generate force on the matrix by extending protrusions and regulating cell volume as well as through actomyosin-based contractility. Furthermore, cell–matrix interactions are dynamic owing to matrix remodelling. Accordingly, ECM stiffness, viscoelasticity and degradability often play a critical role in regulating cell behaviours in 3D. Mechanisms of 3D mechanotransduction include traditional integrin-mediated pathways that sense mechanical properties and more recently described mechanosensitive ion channel-mediated pathways that sense 3D confinement, with both converging on the nucleus for downstream control of transcription and phenotype. Mechanotransduction is involved in tissues from development to cancer and is being increasingly harnessed towards mechanotherapy. Here we discuss recent progress in our understanding of cell–ECM mechanotransduction in 3D.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Tissue mechanics, ECM components and cell–ECM mechanical interactions.
Fig. 2: Cell–matrix interactions in 3D.
Fig. 3: 2D and 3D mechanotransduction.
Fig. 4: Mechanotransduction in development.
Fig. 5: Mechanotransduction in tissues.

Similar content being viewed by others

References

  1. Levental, I., Georges, P. C. & Janmey, P. A. Soft biological materials and their impact on cell function. Soft Matter 3, 299–306 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Swift, J. et al. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341, 1240104 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Storm, C., Pastore, J. J., MacKintosh, F. C., Lubensky, T. C. & Janmey, P. A. Nonlinear elasticity in biological gels. Nature 435, 191–194 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Discher Dennis, E., Janmey, P. & Wang, Y.-L. Tissue cells feel and respond to the stiffness of their substrate. Science 310, 1139–1143 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Vogel, V. & Sheetz, M. Local force and geometry sensing regulate cell functions. Nat. Rev. Mol. Cell Biol. 7, 265–275 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Wozniak, M. A. & Chen, C. S. Mechanotransduction in development: a growing role for contractility. Nat. Rev. Mol. Cell Biol. 10, 34–43 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. DuFort, C. C., Paszek, M. J. & Weaver, V. M. Balancing forces: architectural control of mechanotransduction. Nat. Rev. Mol. Cell Biol. 12, 308–319 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kechagia, J. Z., Ivaska, J. & Roca-Cusachs, P. Integrins as biomechanical sensors of the microenvironment. Nat. Rev. Mol. Cell Biol. 20, 457–473 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Cukierman, E., Pankov, R., Stevens, D. R. & Yamada, K. M. Taking cell-matrix adhesions to the third dimension. Science 294, 1708–1712 (2001). This articles demonstrates the key differences in the structure and composition of cell–ECM adhesions for fibroblasts between 2D culture, 3D culture and tissues.

    Article  CAS  PubMed  Google Scholar 

  10. Baker, B. M. & Chen, C. S. Deconstructing the third dimension – how 3D culture microenvironments alter cellular cues. J. Cell Sci. 125, 3015–3024 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Von Der Mark, K., Gauss, V., Von Der Mark, H. & MÜLler, P. Relationship between cell shape and type of collagen synthesised as chondrocytes lose their cartilage phenotype in culture. Nature 267, 531–532 (1977).

    Article  PubMed  Google Scholar 

  12. Petersen, O. W., Rønnov-Jessen, L., Howlett, A. R. & Bissell, M. J. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc. Natl Acad. Sci. USA 89, 9064–9068 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gerecht, S. et al. Hyaluronic acid hydrogel for controlled self-renewal and differentiation of human embryonic stem cells. Proc. Natl Acad. Sci. USA 104, 11298–11303 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Fischbach, C. et al. Cancer cell angiogenic capability is regulated by 3D culture and integrin engagement. Proc. Natl Acad. Sci. USA 106, 399–404 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Fratzl, P. in Collagen: Structure and Mechanics (ed. Fratzl, P.) 1–13 (Springer, 2008).

  16. Jokinen, J. et al. Integrin-mediated cell adhesion to type I collagen fibrils. J. Biol. Chem. 279, 31956–31963 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Humphries, J. D., Byron, A. & Humphries, M. J. Integrin ligands at a glance. J. Cell Sci. 119, 3901–3903 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Gautieri, A., Vesentini, S., Redaelli, A. & Buehler, M. J. Hierarchical structure and nanomechanics of collagen microfibrils from the atomistic scale up. Nano Lett. 11, 757–766 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Vader, D., Kabla, A., Weitz, D. & Mahadevan, L. Strain-induced alignment in collagen gels. PLoS ONE 4, e5902 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Proestaki, M., Ogren, A., Burkel, B. & Notbohm, J. Modulus of fibrous collagen at the length scale of a cell. Exp. Mech. 59, 1323–1334 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hotary, K., Allen, E., Punturieri, A., Yana, I. & Weiss, S. J. Regulation of cell invasion and morphogenesis in a three-dimensional type I collagen matrix by membrane-type matrix metalloproteinases 1, 2, and 3. J. Cell Biol. 149, 1309–1323 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Münster, S. et al. Strain history dependence of the nonlinear stress response of fibrin and collagen networks. Proc. Natl Acad. Sci. USA 110, 12197–12202 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Nam, S., Hu, K. H., Butte, M. J. & Chaudhuri, O. Strain-enhanced stress relaxation impacts nonlinear elasticity in collagen gels. Proc. Natl Acad. Sci. USA 113, 5492–5497 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ban, E. et al. Mechanisms of plastic deformation in collagen networks induced by cellular forces. Biophys. J. 114, 450–461 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Collet, J.-P., Shuman, H., Ledger, R. E., Lee, S. & Weisel, J. W. The elasticity of an individual fibrin fiber in a clot. Proc. Natl Acad. Sci. USA 102, 9133–9137 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Brown, A. E. X., Litvinov, R. I., Discher, D. E., Purohit, P. K. & Weisel, J. W. Multiscale mechanics of fibrin polymer: gel stretching with protein unfolding and loss of water. Science 325, 741–744 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yurchenco, P. D. Basement membranes: cell scaffoldings and signaling platforms. Cold Spring Harb. Perspect. Biol. 3, a004911 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Chang, J. & Chaudhuri, O. Beyond proteases: basement membrane mechanics and cancer invasion. J. Cell Biol. 218, 2456–2469 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Li, H., Zheng, Y., Han, Y. L., Cai, S. & Guo, M. Nonlinear elasticity of biological basement membrane revealed by rapid inflation and deflation. Proc. Natl Acad. Sci. USA 118, e2022422118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Stowers, R. S. et al. Extracellular matrix stiffening induces a malignant phenotypic transition in breast epithelial cells. Cell. Mol. Bioeng. 10, 114–123 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Reuten, R. et al. Basement membrane stiffness determines metastases formation. Nat. Mater. 20, 892–903 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Kleinman, H. K. & Martin, G. R. Matrigel: Basement membrane matrix with biological activity. Semin. Cancer Biol. 15, 378–386 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Chopra, A. et al. Augmentation of integrin-mediated mechanotransduction by hyaluronic acid. Biomaterials 35, 71–82 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Wolf, K. J. et al. A mode of cell adhesion and migration facilitated by CD44-dependent microtentacles. Proc. Natl Acad. Sci. USA 117, 11432–11443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wolf, K. J. & Kumar, S. Hyaluronic acid: incorporating the bio into the material. ACS Biomater. Sci. Eng. 5, 3753–3765 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Burdick, J. A. & Prestwich, G. D. Hyaluronic acid hydrogels for biomedical applications. Adv. Mater. 23, H41–H56 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Vogel, V. Mechanotransduction involving multimodular proteins: converting force into biochemical signals. Annu. Rev. Biophys. Biomol. Struct. 35, 459–488 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Kong Hyun, J., Polte Thomas, R., Alsberg, E. & Mooney David, J. FRET measurements of cell-traction forces and nano-scale clustering of adhesion ligands varied by substrate stiffness. Proc. Natl Acad. Sci. USA 102, 4300–4305 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Huebsch, N. et al. Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nat. Mater. 9, 518–526 (2010). Using hydrogels to tune stiffness independently of ligand density, the authors demonstrate that ECM stiffness directs stem cell fate in 3D through integrin clustering.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chaudhuri, O. et al. Hydrogels with tunable stress relaxation regulate stem cell fate and activity. Nat. Mater. 15, 326–334 (2016). By developing hydrogels of the same initial stiffness but independently tunable stress relaxation and using these for 3D cell culture, the authors show that stress relaxation (viscoelasticity) impacted cell spreading, proliferation and differentiation of MSCs.

    Article  CAS  PubMed  Google Scholar 

  41. Indana, D., Agarwal, P., Bhutani, N. & Chaudhuri, O. Viscoelasticity and adhesion signaling in biomaterials control human pluripotent stem cell morphogenesis in 3D culture. Adv. Mater. 33, 2101966 (2021).

    Article  CAS  Google Scholar 

  42. Raeber, G. P., Lutolf, M. P. & Hubbell, J. A. Molecularly engineered PEG hydrogels: a novel model system for proteolytically mediated cell migration. Biophys. J. 89, 1374–1388 (2005). This study uses polyethylene glycol-based hydrogels engineered to be susceptible to proteolytic degradation by cell-secreted matrix metalloproteinases to show that increased matrix degradability promotes cell migration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Rowley, J. A., Madlambayan, G. & Mooney, D. J. Alginate hydrogels as synthetic extracellular matrix materials. Biomaterials 20, 45–53 (1999).

    Article  CAS  PubMed  Google Scholar 

  44. Kubow, K. E. et al. Mechanical forces regulate the interactions of fibronectin and collagen I in extracellular matrix. Nat. Commun. 6, 8026 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Baneyx, G., Baugh, L. & Vogel, V. Fibronectin extension and unfolding within cell matrix fibrils controlled by cytoskeletal tension. Proc. Natl Acad. Sci. USA 99, 5139–5143 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Miroshnikova, Y. A. et al. Tissue mechanics promote IDH1-dependent HIF1α–tenascin C feedback to regulate glioblastoma aggression. Nat. Cell Biol. 18, 1336–1345 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Wishart, A. L. et al. Decellularized extracellular matrix scaffolds identify full-length collagen VI as a driver of breast cancer cell invasion in obesity and metastasis. Sci. Adv. 6, eabc3175 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Papanicolaou, M. et al. Temporal profiling of the breast tumour microenvironment reveals collagen XII as a driver of metastasis. Nat. Commun. 13, 4587 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lai, V. K. et al. Swelling of collagen-hyaluronic acid co-gels: an in vitro residual stress model. Ann. Biomed. Eng. 44, 2984–2993 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Burla, F., Tauber, J., Dussi, S., van der Gucht, J. & Koenderink, G. H. Stress management in composite biopolymer networks. Nat. Phys. 15, 549–553 (2019). Lai et al. (2016) and Burla et al. (2019) show the emergent properties of composite materials consisting of col-1 and HA.

    Article  CAS  Google Scholar 

  51. Cocciolone, A. J. et al. Elastin, arterial mechanics, and cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 315, H189–H205 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chaudhuri, O., Cooper-White, J., Janmey, P. A., Mooney, D. J. & Shenoy, V. B. Effects of extracellular matrix viscoelasticity on cellular behaviour. Nature 584, 535–546 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Chen, D. et al. Distinct effects of different matrix proteoglycans on collagen fibrillogenesis and cell-mediated collagen reorganization. Sci. Rep. 10, 19065 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Han Yu, L. et al. Cell contraction induces long-ranged stress stiffening in the extracellular matrix. Proc. Natl Acad. Sci. USA 115, 4075–4080 (2018). Using optical tweezers, the authors demonstrate that cells use their contractile forces to mechanically remodel their local microenvironments into stiffer environments.

    Article  PubMed  PubMed Central  Google Scholar 

  55. van Oosten, A. S. G. et al. Emergence of tissue-like mechanics from fibrous networks confined by close-packed cells. Nature 573, 96–101 (2019). This article demonstrates that a composite of ECM and densely packed cells gives rise to compression stiffening, whereas pure ECM exhibited compression softening, highlighting the role of cells in tissue mechanics.

    Article  PubMed  Google Scholar 

  56. Engler, A. J., Sen, S., Sweeney, H. L. & Discher, D. E. Matrix elasticity directs stem cell lineage specification. Cell 126, 677–689 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Davidson, M. D. et al. Engineered fibrous networks to investigate the influence of fiber mechanics on myofibroblast differentiation. ACS Biomater. Sci. Eng. 5, 3899–3908 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Yamada, K. M. & Sixt, M. Mechanisms of 3D cell migration. Nat. Rev. Mol. Cell Biol. 20, 738–752 (2019).

    Article  CAS  PubMed  Google Scholar 

  59. Paul, C. D., Mistriotis, P. & Konstantopoulos, K. Cancer cell motility: lessons from migration in confined spaces. Nat. Rev. Cancer 17, 131–140 (2017).

    Article  CAS  PubMed  Google Scholar 

  60. Khetan, S. et al. Degradation-mediated cellular traction directs stem cell fate in covalently crosslinked three-dimensional hydrogels. Nat. Mater. 12, 458–465 (2013). In this seminal work, by changing hydrogel degradability and maintaining the same initial stiffness, the authors demonstrate that hydrogel degradability-mediated cell traction directs stem cell fate in 3D microenvironments.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lee, H.-P., Stowers, R. & Chaudhuri, O. Volume expansion and TRPV4 activation regulate stem cell fate in three-dimensional microenvironments. Nat. Commun. 10, 529 (2019). This study identifies a mechanosensitive ion channel-mediated mechanism that MSCs use to sense matrix. Fast stress relaxation promoted reciprocal cell volume expansion and activation of TRPV4 mechanosensitive ion channels to drive nuclear translocation of RUNX2 and osteogenic differentiation.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Yang, B. et al. Enhanced mechanosensing of cells in synthetic 3D matrix with controlled biophysical dynamics. Nat. Commun. 12, 3514 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Engler, A. et al. Substrate compliance versus ligand density in cell on gel responses. Biophys. J. 86, 617–628 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Chen Christopher, S., Mrksich, M., Huang, S., Whitesides George, M. & Ingber Donald, E. Geometric control of cell life and death. Science 276, 1425–1428 (1997).

    Article  Google Scholar 

  65. Gjorevski, N. et al. Tissue geometry drives deterministic organoid patterning. Science 375, eaaw9021 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Wang, M. et al. Regulating mechanotransduction in three dimensions using sub-cellular scale, crosslinkable fibers of controlled diameter, stiffness, and alignment. Adv. Funct. Mater. 29, 1808967 (2019).

    Article  Google Scholar 

  67. Bodor, D. L., Pönisch, W., Endres, R. G. & Paluch, E. K. Of cell shapes and motion: the physical basis of animal cell migration. Dev. Cell 52, 550–562 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  69. Pelham Robert, J. & Wang, Y.-l Cell locomotion and focal adhesions are regulated by substrate flexibility. Proc. Natl Acad. Sci. USA 94, 13661–13665 (1997).

    Article  PubMed Central  Google Scholar 

  70. Gardel, M. L. et al. Traction stress in focal adhesions correlates biphasically with actin retrograde flow speed. J. Cell Biol. 183, 999–1005 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Sunyer, R. et al. Collective cell durotaxis emerges from long-range intercellular force transmission. Science 353, 1157–1161 (2016).

    Article  CAS  PubMed  Google Scholar 

  72. Isomursu, A. et al. Directed cell migration towards softer environments. Nat. Mater. 21, 1081–1090 (2022).

    Article  CAS  PubMed  Google Scholar 

  73. Wolf, K. et al. Physical limits of cell migration: Control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J. Cell Biol. 201, 1069–1084 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Harada, T. et al. Nuclear lamin stiffness is a barrier to 3D migration, but softness can limit survival. J. Cell Biol. 204, 669–682 (2014). Wolf et al. (2013) and Harada et al. (2014) show that matrix pore size regulates cell migration, with cells unable to migrate through pores smaller than 3 μm in diameter in elastic non-degradable gels.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Fraley, S. I. et al. Three-dimensional matrix fiber alignment modulates cell migration and MT1-MMP utility by spatially and temporally directing protrusions. Sci. Rep. 5, 14580 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Trappmann, B. et al. Matrix degradability controls multicellularity of 3D cell migration. Nat. Commun. 8, 371 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Wisdom, K. M. et al. Matrix mechanical plasticity regulates cancer cell migration through confining microenvironments. Nat. Commun. 9, 4144 (2018). Using nanoporous hydrogels with tunable mechanical plasticity, the authors discover that cells can migrate through nanoporous matrices independently of proteases if the matrix exhibits sufficient matrix mechanical plasticity.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Lee, H.-P. et al. The nuclear piston activates mechanosensitive ion channels to generate cell migration paths in confining microenvironments. Sci. Adv. 7, eabd4058 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Petrie, R. J., Harlin, H. M., Korsak, L. I. T. & Yamada, K. M. Activating the nuclear piston mechanism of 3D migration in tumor cells. J. Cell Biol. 216, 93–100 (2016).

    Article  PubMed  Google Scholar 

  80. Polacheck, W. J., German, A. E., Mammoto, A., Ingber, D. E. & Kamm, R. D. Mechanotransduction of fluid stresses governs 3D cell migration. Proc. Natl Acad. Sci. USA 111, 2447–2452 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Blache, U., Stevens, M. M. & Gentleman, E. Harnessing the secreted extracellular matrix to engineer tissues. Nat. Biomed. Eng. 4, 357–363 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Loebel, C., Mauck, R. L. & Burdick, J. A. Local nascent protein deposition and remodelling guide mesenchymal stromal cell mechanosensing and fate in three-dimensional hydrogels. Nat. Mater. 18, 883–891 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Ferreira, S. A. et al. Bi-directional cell-pericellular matrix interactions direct stem cell fate. Nat. Commun. 9, 4049 (2018). Loebel et al. (2019) and Ferreira et al. (2018) demonstrate the role of nascent proteins secreted by cells in 3D as a key regulator of various mechanosensing behaviours such as cell spread area, YAP and TAZ nuclear translocation, and osteogenic differentiation.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Lee, H.-P., Gu, L., Mooney, D. J., Levenston, M. E. & Chaudhuri, O. Mechanical confinement regulates cartilage matrix formation by chondrocytes. Nat. Mater. 16, 1243–1251 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Borelli, A. N. et al. Stress relaxation and composition of hydrazone-crosslinked hybrid biopolymer-synthetic hydrogels determine spreading and secretory properties of MSCs. Adv. Healthc. Mater. https://doi.org/10.1002/adhm.202200393 (2022).

    Article  PubMed  Google Scholar 

  86. Sridhar, B. V. et al. Development of a cellularly degradable PEG hydrogel to promote articular cartilage extracellular matrix deposition. Adv. Healthc. Mater. 4, 702–713 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Vining, K. H. & Mooney, D. J. Mechanical forces direct stem cell behaviour in development and regeneration. Nat. Rev. Mol. Cell Biol. 18, 728–742 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Engler, A. J. et al. Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. J. Cell Biol. 166, 877–887 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Fu, J. et al. Mechanical regulation of cell function with geometrically modulated elastomeric substrates. Nat. Methods 7, 733–736 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Wen, J. H. et al. Interplay of matrix stiffness and protein tethering in stem cell differentiation. Nat. Mater. 13, 979–987 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Saha, K. et al. Substrate modulus directs neural stem cell behavior. Biophys. J. 95, 4426–4438 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Gilbert, P. M. et al. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 329, 1078–1081 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Ye, K. et al. Matrix stiffness and nanoscale spatial organization of cell-adhesive ligands direct stem cell fate. Nano Lett. 15, 4720–4729 (2015).

    Article  CAS  PubMed  Google Scholar 

  94. Yang, C. et al. Spatially patterned matrix elasticity directs stem cell fate. Proc. Natl Acad. Sci. USA 113, E4439–E4445 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Charrier, E. E., Pogoda, K., Wells, R. G. & Janmey, P. A. Control of cell morphology and differentiation by substrates with independently tunable elasticity and viscous dissipation. Nat. Commun. 9, 449 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  96. Cameron, A. R., Frith, J. E. & Cooper-White, J. J. The influence of substrate creep on mesenchymal stem cell behaviour and phenotype. Biomaterials 32, 5979–5993 (2011).

    Article  CAS  PubMed  Google Scholar 

  97. Stanton, A. E., Tong, X. & Yang, F. Extracellular matrix type modulates mechanotransduction of stem cells. Acta Biomater. 96, 310–320 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kilian, K. A., Bugarija, B., Lahn, B. T. & Mrksich, M. Geometric cues for directing the differentiation of mesenchymal stem cells. Proc. Natl Acad. Sci. USA 107, 4872–4877 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Muncie, J. M. et al. Mechanical tension promotes formation of gastrulation-like nodes and patterns mesoderm specification in human embryonic stem cells. Dev. Cell 55, 679–694.e611 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Das, R. K., Gocheva, V., Hammink, R., Zouani, O. F. & Rowan, A. E. Stress-stiffening-mediated stem-cell commitment switch in soft responsive hydrogels. Nat. Mater. 15, 318–325 (2016).

    Article  CAS  PubMed  Google Scholar 

  101. Hayward, M.-K., Muncie, J. M. & Weaver, V. M. Tissue mechanics in stem cell fate, development, and cancer. Dev. Cell 56, 1833–1847 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Shao, Y., Sang, J. & Fu, J. On human pluripotent stem cell control: the rise of 3D bioengineering and mechanobiology. Biomaterials 52, 26–43 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Madl, C. M. et al. Maintenance of neural progenitor cell stemness in 3D hydrogels requires matrix remodelling. Nat. Mater. 16, 1233–1242 (2017). This article demonstrates that increased ECM degradability in 3D allows cell–cell contact formation, which helps to maintain neural progenitor cell stemness, in the absence of cytoskeletal tension generation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Vining, K. H. et al. Mechanical checkpoint regulates monocyte differentiation in fibrotic niches. Nat. Mater. 21, 939–950 (2022).

    Article  CAS  PubMed  Google Scholar 

  105. Klein, E. A. et al. Cell-cycle control by physiological matrix elasticity and in vivo tissue stiffening. Curr. Biol. 19, 1511–1518 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Nam, S. et al. Cell cycle progression in confining microenvironments is regulated by a growth-responsive TRPV4-PI3K/Akt-p27Kip1 signaling axis. Sci. Adv. 5, eaaw6171 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Dudaryeva, O. Y. et al. 3D confinement regulates cell life and death. Adv. Funct. Mater. 31, 2104098 (2021).

    Article  CAS  Google Scholar 

  108. Kratochvil, M. J. et al. Engineered materials for organoid systems. Nat. Rev. Mater. 4, 606–622 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Rezakhani, S., Gjorevski, N. & Lutolf, M. P. Extracellular matrix requirements for gastrointestinal organoid cultures. Biomaterials 276, 121020 (2021).

    Article  CAS  PubMed  Google Scholar 

  110. Enemchukwu, N. O. et al. Synthetic matrices reveal contributions of ECM biophysical and biochemical properties to epithelial morphogenesis. J. Cell Biol. 212, 113–124 (2015).

    Article  PubMed  Google Scholar 

  111. Gjorevski, N. et al. Designer matrices for intestinal stem cell and organoid culture. Nature 539, 560–564 (2016).

    Article  CAS  PubMed  Google Scholar 

  112. Cruz-Acuña, R. et al. Synthetic hydrogels for human intestinal organoid generation and colonic wound repair. Nat. Cell Biol. 19, 1326–1335 (2017). Gjorevski et al. (2016) and Cruz-Acuña et al. (2017) demonstrate the use of 3D culture in engineered hydrogels for guiding morphogenesis of stem cells, highlighting the role of hydrogel degradability.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Ranga, A. et al. Neural tube morphogenesis in synthetic 3D microenvironments. Proc. Natl Acad. Sci. USA 113, E6831–E6839 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Blatchley, M. R., Hall, F., Wang, S., Pruitt, H. C. & Gerecht, S. Hypoxia and matrix viscoelasticity sequentially regulate endothelial progenitor cluster-based vasculogenesis. Sci. Adv. 5, eaau7518 (2019). Blatchley et al. (2019) and Indana et al. (2021) demonstrate the use of 3D culture in engineered hydrogels for guiding morphogenesis of stem cells, highlighting the role of hydrogel viscoelasticity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Chrisnandy, A., Blondel, D., Rezakhani, S., Broguiere, N. & Lutolf, M. P. Synthetic dynamic hydrogels promote degradation-independent in vitro organogenesis. Nat. Mater. 21, 479–487 (2022).

    Article  CAS  PubMed  Google Scholar 

  116. Sorrentino, G. et al. Mechano-modulatory synthetic niches for liver organoid derivation. Nat. Commun. 11, 3416 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Kai, F., Drain, A. P. & Weaver, V. M. The extracellular matrix modulates the metastatic journey. Dev. Cell 49, 332–346 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Provenzano, P. P. et al. Collagen density promotes mammary tumor initiation and progression. BMC Med. 6, 11 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Paszek, M. J. et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005). In this multifaceted work that uses both 2D and 3D cell culture studies, the authors show that increased stiffness and collagen density, as occurs during breast cancer progression, promotes a malignant phenotype in even normal mammary epithelial cells through β1 integrin clustering, Rho and FAK activation.

    Article  CAS  PubMed  Google Scholar 

  120. Levental, K. R. et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139, 891–906 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Wei, S. C. et al. Matrix stiffness drives epithelial–mesenchymal transition and tumour metastasis through a TWIST1–G3BP2 mechanotransduction pathway. Nat. Cell Biol. 17, 678–688 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Chaudhuri, O. et al. Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater. 13, 970–978 (2014).

    Article  CAS  PubMed  Google Scholar 

  123. Tse, J. M. et al. Mechanical compression drives cancer cells toward invasive phenotype. Proc. Natl Acad. Sci. USA 109, 911–916 (2012).

    Article  CAS  PubMed  Google Scholar 

  124. Kim, J. et al. Geometric dependence of 3D collective cancer invasion. Biophys. J. 118, 1177–1182 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Debnath, J. & Brugge, J. S. Modelling glandular epithelial cancers in three-dimensional cultures. Nat. Rev. Cancer 5, 675–688 (2005).

    Article  CAS  PubMed  Google Scholar 

  126. Lee, J. Y. et al. YAP-independent mechanotransduction drives breast cancer progression. Nat. Commun. 10, 1848 (2019). In contrast to the universal role of YAP in mediating mechanotransduction on 2D substrates, this article demonstrates that increased stiffness promotes malignancy in mammary epithelial cells independently of YAP in 3D culture, which is consistent with in vivo analysis of breast cancer, and connected this observation to the lack of focal adhesions, stress fibres and stretched nuclei in 3D.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Stowers, R. S. et al. Matrix stiffness induces a tumorigenic phenotype in mammary epithelium through changes in chromatin accessibility. Nat. Biomed. Eng. 3, 1009–1019 (2019). The authors demonstrate changes in chromatin accessibility in breast cancer cells with a change in ECM stiffness in 3D, and these changes allow binding of the transcription factor Sp1 to promote a malignant phenotype, revealing that chromatin state is a critical mediator of mechanotranduction.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Long, H., Vos, B. E., Betz, T., Baker, B. M. & Trappmann, B. Nonswelling and hydrolytically stable hydrogels uncover cellular mechanosensing in 3D. Adv. Sci. 9, 2105325 (2022).

    Article  Google Scholar 

  129. Lou, J., Stowers, R., Nam, S., Xia, Y. & Chaudhuri, O. Stress relaxing hyaluronic acid-collagen hydrogels promote cell spreading, fiber remodeling, and focal adhesion formation in 3D cell culture. Biomaterials 154, 213–222 (2018).

    Article  CAS  PubMed  Google Scholar 

  130. Caliari, S. R., Vega, S. L., Kwon, M., Soulas, E. M. & Burdick, J. A. Dimensionality and spreading influence MSC YAP/TAZ signaling in hydrogel environments. Biomaterials 103, 314–323 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Nam, S., Lee, J., Brownfield, Doug, G. & Chaudhuri, O. Viscoplasticity enables mechanical remodeling of matrix by cells. Biophys. J. 111, 2296–2308 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Qazi, T. H. et al. Programming hydrogels to probe spatiotemporal cell biology. Cell Stem Cell https://doi.org/10.1016/j.stem.2022.03.013 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  133. Schultz, K. M., Kyburz, K. A. & Anseth, K. S. Measuring dynamic cell–material interactions and remodeling during 3D human mesenchymal stem cell migration in hydrogels. Proc. Natl Acad. Sci. USA 112, E3757–E3764 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Sabeh, F., Shimizu-Hirota, R. & Weiss, S. J. Protease-dependent versus -independent cancer cell invasion programs: three-dimensional amoeboid movement revisited. J. Cell Biol. 185, 11–19 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. McKinnon, D. D., Domaille, D. W., Cha, J. N. & Anseth, K. S. Biophysically defined and cytocompatible covalently adaptable networks as viscoelastic 3D cell culture systems. Adv. Mater. 26, 865–872 (2014).

    Article  CAS  PubMed  Google Scholar 

  136. Nam, S. & Chaudhuri, O. Mitotic cells generate protrusive extracellular forces to divide in three-dimensional microenvironments. Nat. Phys. 14, 621–628 (2018).

    Article  CAS  Google Scholar 

  137. Legant, W. R. et al. Measurement of mechanical tractions exerted by cells in three-dimensional matrices. Nat. Methods 7, 969–971 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Franck, C., Maskarinec, S. A., Tirrell, D. A. & Ravichandran, G. Three-dimensional traction force microscopy: a new tool for quantifying cell-matrix interactions. PLoS ONE 6, e17833 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Steinwachs, J. et al. Three-dimensional force microscopy of cells in biopolymer networks. Nat. Methods 13, 171–176 (2016).

    Article  CAS  PubMed  Google Scholar 

  140. Bell, E., Ivarsson, B. & Merrill, C. Production of a tissue-like structure by contraction of collagen lattices by human fibroblasts of different proliferative potential in vitro. Proc. Natl Acad. Sci. USA 76, 1274–1278 (1979).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Fletcher, D. A. & Mullins, R. D. Cell mechanics and the cytoskeleton. Nature 463, 485–492 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Papalazarou, V. & Machesky, L. M. The cell pushes back: the Arp2/3 complex is a key orchestrator of cellular responses to environmental forces. Curr. Opin. Cell Biol. 68, 37–44 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Gaertner, F. et al. WASp triggers mechanosensitive actin patches to facilitate immune cell migration in dense tissues. Dev. Cell 57, 47–62.e49 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Nam, S., Lin, Y.-H., Kim, T. & Chaudhuri, O. Cellular pushing forces during mitosis drive mitotic elongation in collagen gels. Adv. Sci. 8, 2000403 (2021).

    Article  CAS  Google Scholar 

  145. Hoffmann, E. K., Lambert, I. H. & Pedersen, S. F. Physiology of cell volume regulation in vertebrates. Physiol. Rev. 89, 193–277 (2009).

    Article  CAS  PubMed  Google Scholar 

  146. Ginzberg Miriam, B., Kafri, R. & Kirschner, M. On being the right (cell) size. Science 348, 1245075 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Adar Ram, M. & Safran Samuel, A. Active volume regulation in adhered cells. Proc. Natl Acad. Sci. USA 117, 5604–5609 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Petrie, R. J., Koo, H. & Yamada Kenneth, M. Generation of compartmentalized pressure by a nuclear piston governs cell motility in a 3D matrix. Science 345, 1062–1065 (2014). In contrast to the conventional thinking that the nucleus acts a limiting factor for cell migration in 3D, Petrie et al. (2014) and Lee et al. (2021) drives the nucleus like a piston into the protrusion. This action activates mechanosensitive ion channels to allow an influx of ions that increases osmotic pressure, which then outcompetes hydrostatic pressure to drive protrusion expansion, generating a path for migration in confining 3D microenvironments.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Stroka, K. M. et al. Water permeation drives tumor cell migration in confined microenvironments. Cell 157, 611–623 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Helmlinger, G., Netti, P. A., Lichtenbeld, H. C., Melder, R. J. & Jain, R. K. Solid stress inhibits the growth of multicellular tumor spheroids. Nat. Biotechnol. 15, 778–783 (1997).

    Article  CAS  PubMed  Google Scholar 

  151. Nia, H. T. et al. Solid stress and elastic energy as measures of tumour mechanopathology. Nat. Biomed. Eng. 1, 0004 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  152. Kasza, K. E. et al. The cell as a material. Curr. Opin. Cell Biol. 19, 101–107 (2007).

    Article  CAS  PubMed  Google Scholar 

  153. Jung, W., Li, J., Chaudhuri, O. & Kim, T. Nonlinear elastic and inelastic properties of cells. J. Biomech. Eng. https://doi.org/10.1115/1.4046863 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  154. Guo, M. et al. Cell volume change through water efflux impacts cell stiffness and stem cell fate. Proc. Natl Acad. Sci. USA 114, E8618–E8627 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Lomakin, A. J. et al. The nucleus acts as a ruler tailoring cell responses to spatial constraints. Science 370, eaba2894 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Venturini, V. et al. The nucleus measures shape changes for cellular proprioception to control dynamic cell behavior. Science 370, eaba2644 (2020).

    Article  CAS  PubMed  Google Scholar 

  157. Heo, S.-J. et al. Nuclear softening expedites interstitial cell migration in fibrous networks and dense connective tissues. Sci. Adv. 6, eaax5083 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Doyle, A. D., Carvajal, N., Jin, A., Matsumoto, K. & Yamada, K. M. Local 3D matrix microenvironment regulates cell migration through spatiotemporal dynamics of contractility-dependent adhesions. Nat. Commun. 6, 8720 (2015).

    Article  CAS  PubMed  Google Scholar 

  159. Hall Matthew, S. et al. Fibrous nonlinear elasticity enables positive mechanical feedback between cells and ECMs. Proc. Natl Acad. Sci. USA 113, 14043–14048 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Doyle, A. D., Sykora, D. J., Pacheco, G. G., Kutys, M. L. & Yamada, K. M. 3D mesenchymal cell migration is driven by anterior cellular contraction that generates an extracellular matrix prestrain. Dev. Cell 56, 826–841.e824 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Petrie, R. J., Gavara, N., Chadwick, R. S. & Yamada, K. M. Nonpolarized signaling reveals two distinct modes of 3D cell migration. J. Cell Biol. 197, 439–455 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Gong, Z. et al. Recursive feedback between matrix dissipation and chemo-mechanical signaling drives oscillatory growth of cancer cell invadopodia. Cell Rep. 35, 109047 (2021). In this study, the authors use a chemomechanical model to study cell–matrix interactions and find protrusive forces generated by cells through invadopodia deform the matrix and in sufficiently plastic matrices such forces lead to permanent matrix deformations, which in turn promote growth of protrusions and result in larger pores for cell migration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Gong, Z. et al. Matching material and cellular timescales maximizes cell spreading on viscoelastic substrates. Proc. Natl Acad. Sci. USA 115, E2686–E2695 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Blache, U. et al. Notch-inducing hydrogels reveal a perivascular switch of mesenchymal stem cell fate. EMBO Rep. 19, e45964 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  165. Provenzano, P. P., Inman, D. R., Eliceiri, K. W. & Keely, P. J. Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK–ERK linkage. Oncogene 28, 4326–4343 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Wei, Z., Schnellmann, R., Pruitt, H. C. & Gerecht, S. Hydrogel network dynamics regulate vascular morphogenesis. Cell Stem Cell 27, 798–812.e796 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Parekh, S. H. et al. Modulus-driven differentiation of marrow stromal cells in 3D scaffolds that is independent of myosin-based cytoskeletal tension. Biomaterials 32, 2256–2264 (2011).

    Article  CAS  PubMed  Google Scholar 

  168. Bao, M., Xie, J., Piruska, A. & Huck, W. T. S. 3D microniches reveal the importance of cell size and shape. Nat. Commun. 8, 1962 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  169. Agarwal, P. et al. A dysfunctional TRPV4–GSK3β pathway prevents osteoarthritic chondrocytes from sensing changes in extracellular matrix viscoelasticity. Nat. Biomed. Eng. 5, 1472–1484 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Batan, D. et al. Hydrogel cultures reveal transient receptor potential vanilloid 4 regulation of myofibroblast activation and proliferation in valvular interstitial cells. FASEB J. 36, e22306 (2022).

    Article  CAS  PubMed  Google Scholar 

  171. Wong, S. W. et al. Controlled deposition of 3D matrices to direct single cell functions. Adv. Sci. 7, 2001066 (2020).

    Article  CAS  Google Scholar 

  172. Murthy, S. E., Dubin, A. E. & Patapoutian, A. Piezos thrive under pressure: mechanically activated ion channels in health and disease. Nat. Rev. Mol. Cell Biol. 18, 771–783 (2017).

    Article  CAS  PubMed  Google Scholar 

  173. Zhao, R. et al. Cell sensing and decision-making in confinement: the role of TRPM7 in a tug of war between hydraulic pressure and cross-sectional area. Sci. Adv. 5, eaaw7243 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Yankaskas, C. L. et al. The fluid shear stress sensor TRPM7 regulates tumor cell intravasation. Sci. Adv. 7, eabh3457 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Kalukula, Y., Stephens, A. D., Lammerding, J. & Gabriele, S. Mechanics and functional consequences of nuclear deformations. Nat. Rev. Mol. Cell Biol. https://doi.org/10.1038/s41580-022-00480-z (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  176. Cosgrove, B. D. et al. Nuclear envelope wrinkling predicts mesenchymal progenitor cell mechano-response in 2D and 3D microenvironments. Biomaterials 270, 120662 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Darnell, M. et al. Material microenvironmental properties couple to induce distinct transcriptional programs in mammalian stem cells. Proc. Natl Acad. Sci. USA 115, E8368–E8377 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Jang, M. et al. Matrix stiffness epigenetically regulates the oncogenic activation of the Yes-associated protein in gastric cancer. Nat. Biomed. Eng. 5, 114–123 (2021).

    Article  CAS  PubMed  Google Scholar 

  179. Baek, J. et al. Egr1 is a 3D matrix-specific mediator of mechanosensitive stem cell lineage commitment. Sci. Adv. 8, eabm4646 (2022). This article demonstrates upregulation of EGR1, which mediates neural stem cell differentiation, is unique to 3D, highlighting EGR1 as a 3D-specific stem cell mechanoregulatory factor.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Wong, S. W., Lenzini, S., Cooper, M. H., Mooney, D. J. & Shin, J.-W. Soft extracellular matrix enhances inflammatory activation of mesenchymal stromal cells to induce monocyte production and trafficking. Sci. Adv. 6, eaaw0158 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Walker, C. J. et al. Nuclear mechanosensing drives chromatin remodelling in persistently activated fibroblasts. Nat. Biomed. Eng. 5, 1485–1499 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Madl, C. M., LeSavage, B. L., Khariton, M. & Heilshorn, S. C. Neural progenitor cells alter chromatin organization and neurotrophin expression in response to 3D matrix degradability. Adv. Healthc. Mater. 9, 2000754 (2020).

    Article  CAS  Google Scholar 

  183. Stooke-Vaughan, G. A. & Campàs, O. Physical control of tissue morphogenesis across scales. Curr. Opin. Genet. Dev. 51, 111–119 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Serwane, F. et al. In vivo quantification of spatially varying mechanical properties in developing tissues. Nat. Methods 14, 181–186 (2017).

    Article  CAS  PubMed  Google Scholar 

  185. Bedzhov, I. & Zernicka-Goetz, M. Self-organizing properties of mouse pluripotent cells initiate morphogenesis upon implantation. Cell 156, 1032–1044 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Kyprianou, C. et al. Basement membrane remodelling regulates mouse embryogenesis. Nature 582, 253–258 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Rauzi, M. et al. Embryo-scale tissue mechanics during Drosophila gastrulation movements. Nat. Commun. 6, 8677 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Hannezo, E. & Heisenberg, C.-P. Rigidity transitions in development and disease. Trends Cell Biol. 32, 433–444 (2022).

    Article  PubMed  Google Scholar 

  189. Kim, S., Pochitaloff, M., Stooke-Vaughan, G. A. & Campàs, O. Embryonic tissues as active foams. Nat. Phys. 17, 859–866 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Priya, R. et al. Tension heterogeneity directs form and fate to pattern the myocardial wall. Nature 588, 130–134 (2020).

    Article  CAS  PubMed  Google Scholar 

  191. Shellard, A. & Mayor, R. Collective durotaxis along a self-generated stiffness gradient in vivo. Nature 600, 690–694 (2021). This article demonstrates that cells collectively generate stiffness gradients in vivo that result in efficient collective cell migration.

    Article  CAS  PubMed  Google Scholar 

  192. Wang, S., Matsumoto, K., Lish, S. R., Cartagena-Rivera, A. X. & Yamada, K. M. Budding epithelial morphogenesis driven by cell-matrix versus cell-cell adhesion. Cell 184, 3702–3716.e3730 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Hughes, A. J. et al. Engineered tissue folding by mechanical compaction of the mesenchyme. Dev. Cell 44, 165–178.e166 (2018).

    Article  CAS  PubMed  Google Scholar 

  194. Webster, K. D., Ng, W. P. & Fletcher, D. A. Tensional homeostasis in single fibroblasts. Biophys. J. 107, 146–155 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Grinnell, F. Fibroblast biology in three-dimensional collagen matrices. Trends Cell Biol. 13, 264–269 (2003).

    Article  CAS  PubMed  Google Scholar 

  196. Jaalouk, D. E. & Lammerding, J. Mechanotransduction gone awry. Nat. Rev. Mol. Cell Biol. 10, 63–73 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Choudhury, M. I. et al. Kidney epithelial cells are active mechano-biological fluid pumps. Nat. Commun. 13, 2317 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Qian, W. et al. Microskeletal stiffness promotes aortic aneurysm by sustaining pathological vascular smooth muscle cell mechanosensation via Piezo1. Nat. Commun. 13, 512 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Darnell, M. et al. Substrate stress-relaxation regulates scaffold remodeling and bone formation in vivo. Adv. Healthc. Mater. 6, 1601185 (2017).

    Article  Google Scholar 

  200. Phillip, J. M., Aifuwa, I., Walston, J. & Wirtz, D. The mechanobiology of aging. Annu. Rev. Biomed. Eng. 17, 113–141 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Khalilgharibi, N. & Mao, Y. To form and function: on the role of basement membrane mechanics in tissue development, homeostasis and disease. Open. Biol. 11, 200360 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Brun, C. et al. Phenotypic and functional changes in dermal primary fibroblasts isolated from intrinsically aged human skin. Exp. Dermatol. 25, 113–119 (2016).

    Article  CAS  PubMed  Google Scholar 

  203. Phillip, J. M. et al. Fractional re-distribution among cell motility states during ageing. Commun. Biol. 4, 81 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Humphrey, J. D. & Schwartz, M. A. Vascular mechanobiology: homeostasis, adaptation, and disease. Annu. Rev. Biomed. Eng. 23, 1–27 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Hall, C. M., Moeendarbary, E. & Sheridan, G. K. Mechanobiology of the brain in ageing and Alzheimer’s disease. Eur. J. Neurosci. 53, 3851–3878 (2021).

    Article  CAS  PubMed  Google Scholar 

  206. Wynn, T. A. Fibrotic disease and the TH1/TH2 paradigm. Nat. Rev. Immunol. 4, 583–594 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Seo, B. R. et al. Collagen microarchitecture mechanically controls myofibroblast differentiation. Proc. Natl Acad. Sci. USA 117, 11387–11398 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Freeberg, M. A. T. et al. Mechanical feed-forward loops contribute to idiopathic pulmonary fibrosis. Am. J. Pathol. 191, 18–25 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Bissell, M. J. & Hines, W. C. Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat. Med. 17, 320–329 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Piersma, B., Hayward, M.-K. & Weaver, V. M. Fibrosis and cancer: a strained relationship. Biochim. Biophys. Acta Rev. Cancer 1873, 188356 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Acerbi, I. et al. Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr. Biol. 7, 1120–1134 (2015).

    Article  CAS  Google Scholar 

  212. Calvo, F. et al. Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat. Cell Biol. 15, 637–646 (2013).

    Article  CAS  PubMed  Google Scholar 

  213. Maller, O. et al. Tumour-associated macrophages drive stromal cell-dependent collagen crosslinking and stiffening to promote breast cancer aggression. Nat. Mater. 20, 548–559 (2021).

    Article  CAS  PubMed  Google Scholar 

  214. Provenzano, P. P. et al. Collagen reorganization at the tumor-stromal interface facilitates local invasion. BMC Med. 4, 38 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  215. Laklai, H. et al. Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression. Nat. Med. 22, 497–505 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Jiang, H. et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat. Med. 22, 851–860 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Baker, A. M., Bird, D., Lang, G., Cox, T. R. & Erler, J. T. Lysyl oxidase enzymatic function increases stiffness to drive colorectal cancer progression through FAK. Oncogene 32, 1863–1868 (2013).

    Article  CAS  PubMed  Google Scholar 

  218. Peng, D. H. et al. ZEB1 induces LOXL2-mediated collagen stabilization and deposition in the extracellular matrix to drive lung cancer invasion and metastasis. Oncogene 36, 1925–1938 (2017).

    Article  CAS  PubMed  Google Scholar 

  219. Chang, T. T., Thakar, D. & Weaver, V. M. Force-dependent breaching of the basement membrane. Matrix Biol. 57-58, 178–189 (2017).

    Article  CAS  PubMed  Google Scholar 

  220. Mittapalli, V. R. et al. Injury-driven stiffening of the dermis expedites skin carcinoma progression. Cancer Res. 76, 940–951 (2016).

    Article  CAS  PubMed  Google Scholar 

  221. Balleyguier, C. et al. Value of whole breast magnetic resonance elastography added to MRI for lesion characterization. NMR Biomed. 31, e3795 (2018).

    Article  Google Scholar 

  222. Streitberger, K.-J. et al. High-resolution mechanical imaging of glioblastoma by multifrequency magnetic resonance elastography. PLoS ONE 9, e110588 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  223. Garteiser, P. et al. MR elastography of liver tumours: value of viscoelastic properties for tumour characterisation. Eur. Radiol. 22, 2169–2177 (2012).

    Article  PubMed  Google Scholar 

  224. DeForest, C. A. & Tirrell, D. A. A photoreversible protein-patterning approach for guiding stem cell fate in three-dimensional gels. Nat. Mater. 14, 523–531 (2015).

    Article  CAS  PubMed  Google Scholar 

  225. Mao, A. S. et al. Deterministic encapsulation of single cells in thin tunable microgels for niche modelling and therapeutic delivery. Nat. Mater. 16, 236–243 (2017).

    Article  CAS  PubMed  Google Scholar 

  226. Chen, B.-C. et al. Lattice light-sheet microscopy: Imaging molecules to embryos at high spatiotemporal resolution. Science 346, 1257998 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  227. Welch, C. M., Elliott, H., Danuser, G. & Hahn, K. M. Imaging the coordination of multiple signalling activities in living cells. Nat. Rev. Mol. Cell Biol. 12, 749–756 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Han, K. et al. CRISPR screens in cancer spheroids identify 3D growth-specific vulnerabilities. Nature 580, 136–141 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Noskovicova, N. et al. Suppression of the fibrotic encapsulation of silicone implants by inhibiting the mechanical activation of pro-fibrotic TGF-β. Nat. Biomed. Eng. 5, 1437–1456 (2021).

    Article  CAS  PubMed  Google Scholar 

  230. Chen, K. et al. Disrupting mechanotransduction decreases fibrosis and contracture in split-thickness skin grafting. Sci. Transl. Med. 14, eabj9152 (2022).

    Article  CAS  PubMed  Google Scholar 

  231. Umehara, T. et al. Female reproductive life span is extended by targeted removal of fibrotic collagen from the mouse ovary. Sci. Adv. 8, eabn4564 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Winkler, J., Abisoye-Ogunniyan, A., Metcalf, K. J. & Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 11, 5120 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Madl, C. M., Heilshorn, S. C. & Blau, H. M. Bioengineering strategies to accelerate stem cell therapeutics. Nature 557, 335–342 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Yeung, T. et al. Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Motil. 60, 24–34 (2005).

    Article  Google Scholar 

  235. Branco da Cunha, C. et al. Influence of the stiffness of three-dimensional alginate/collagen-I interpenetrating networks on fibroblast biology. Biomaterials 35, 8927–8936 (2014).

    Article  CAS  PubMed  Google Scholar 

  236. Stowers, R. S., Allen, S. C. & Suggs, L. J. Dynamic phototuning of 3D hydrogel stiffness. Proc. Natl Acad. Sci. USA 112, 1953–1958 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Cameron, A. R., Frith, J. E., Gomez, G. A., Yap, A. S. & Cooper-White, J. J. The effect of time-dependent deformation of viscoelastic hydrogels on myogenic induction and Rac1 activity in mesenchymal stem cells. Biomaterials 35, 1857–1868 (2014).

    Article  CAS  PubMed  Google Scholar 

  238. Chaudhuri, O. et al. Substrate stress relaxation regulates cell spreading. Nat. Commun. 6, 6365 (2015).

    Article  CAS  Google Scholar 

  239. Huebsch, N. et al. Matrix elasticity of void-forming hydrogels controls transplanted-stem-cell-mediated bone formation. Nat. Mater. 14, 1269–1277 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Trappmann, B. et al. Extracellular-matrix tethering regulates stem-cell fate. Nat. Mater. 11, 642–649 (2012).

    Article  CAS  PubMed  Google Scholar 

  241. Hakkinen, K. M., Harunaga, J. S., Doyle, A. D. & Yamada, K. M. Direct comparisons of the morphology, migration, cell adhesions, and actin cytoskeleton of fibroblasts in four different three-dimensional extracellular matrices. Tissue Eng. Part. A 17, 713–724 (2011).

    Article  CAS  PubMed  Google Scholar 

  242. McBeath, R., Pirone, D. M., Nelson, C. M., Bhadriraju, K. & Chen, C. S. Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev. Cell 6, 483–495 (2004).

    Article  CAS  PubMed  Google Scholar 

  243. Kassianidou, E. et al. Extracellular matrix geometry and initial adhesive position determine stress fiber network organization during cell spreading. Cell Rep. 27, 1897–1909.e1894 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Ulrich, T. A., de Juan Pardo, E. M. & Kumar, S. The mechanical rigidity of the extracellular matrix regulates the structure, motility, and proliferation of glioma cells. Cancer Res. 69, 4167–4174 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Raab, M. et al. Crawling from soft to stiff matrix polarizes the cytoskeleton and phosphoregulates myosin-II heavy chain. J. Cell Biol. 199, 669–683 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Plotnikov, S. V., Pasapera, A. M., Sabass, B. & Waterman, C. M. Force fluctuations within focal adhesions mediate ECM-rigidity sensing to guide directed cell migration. Cell 151, 1513–1527 (2012).

    Article  CAS  PubMed  Google Scholar 

  247. Ulrich, T. A., Jain, A., Tanner, K., MacKay, J. L. & Kumar, S. Probing cellular mechanobiology in three-dimensional culture with collagen–agarose matrices. Biomaterials 31, 1875–1884 (2010).

    Article  CAS  PubMed  Google Scholar 

  248. Adebowale, K. et al. Enhanced substrate stress relaxation promotes filopodia-mediated cell migration. Nat. Mater. 20, 1290–1299 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Hu, X. et al. Control cell migration by engineering integrin ligand assembly. Nat. Commun. 13, 5002 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Garbett, D. et al. T-Plastin reinforces membrane protrusions to bridge matrix gaps during cell migration. Nat. Commun. 11, 4818 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Reversat, A. et al. Cellular locomotion using environmental topography. Nature 582, 582–585 (2020).

    Article  CAS  PubMed  Google Scholar 

  252. Kloxin, A. M., Kasko, A. M., Salinas, C. N. & Anseth, K. S. Photodegradable hydrogels for dynamic tuning of physical and chemical properties. Science 324, 59–63 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Nikolaev, M. et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature 585, 574–578 (2020).

    Article  PubMed  Google Scholar 

  254. Mih, J. D., Marinkovic, A., Liu, F., Sharif, A. S. & Tschumperlin, D. J. Matrix stiffness reverses the effect of actomyosin tension on cell proliferation. J. Cell Sci. 125, 5974–5983 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Han, W. M. et al. Synthetic matrix enhances transplanted satellite cell engraftment in dystrophic and aged skeletal muscle with comorbid trauma. Sci. Adv. 4, eaar4008 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Vitiello, E. et al. Acto-myosin force organization modulates centriole separation and PLK4 recruitment to ensure centriole fidelity. Nat. Commun. 10, 52 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Moriarty, R. A. & Stroka, K. M. Physical confinement alters sarcoma cell cycle progression and division. Cell Cycle 17, 2360–2373 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Qin, R. et al. Tumor suppressor DAPK1 catalyzes adhesion assembly on rigid but anoikis on soft matrices. Front. Cell Dev. Biol. https://doi.org/10.3389/fcell.2022.959521 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  259. Farrelly, N., Lee, Y.-J., Oliver, J., Dive, C. & Streuli, C. H. Extracellular matrix regulates apoptosis in mammary epithelium through a control on insulin signaling. J. Cell Biol. 144, 1337–1348 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Indra, I. et al. An in vitro correlation of mechanical forces and metastatic capacity. Phys. Biol. 8, 015015 (2011).

    Article  PubMed  Google Scholar 

  261. Kraning-Rush, C. M., Califano, J. P. & Reinhart-King, C. A. Cellular traction stresses increase with increasing metastatic potential. PLoS ONE 7, e32572 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. van den Berg, M. C. W. et al. Proteolytic and opportunistic breaching of the basement membrane zone by immune cells during tumor initiation. Cell Rep. 27, 2837–2846.e2834 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  263. Wolf, K. et al. Multi-step pericellular proteolysis controls the transition from individual to collective cancer cell invasion. Nat. Cell Biol. 9, 893–904 (2007).

    Article  CAS  PubMed  Google Scholar 

  264. Shi, Q. et al. Rapid disorganization of mechanically interacting systems of mammary acini. Proc. Natl Acad. Sci. USA 111, 658–663 (2014).

    Article  CAS  PubMed  Google Scholar 

  265. Lee, J., Abdeen, A. A., Wycislo, K. L., Fan, T. M. & Kilian, K. A. Interfacial geometry dictates cancer cell tumorigenicity. Nat. Mater. 15, 856–862 (2016).

    Article  CAS  PubMed  Google Scholar 

  266. Hushka, E. A., Yavitt, F. M., Brown, T. E., Dempsey, P. J. & Anseth, K. S. Relaxation of extracellular matrix forces directs crypt formation and architecture in intestinal organoids. Adv. Healthc. Mater. 9, 1901214 (2020).

    Article  CAS  Google Scholar 

  267. Wang, W. Y. et al. Direct comparison of angiogenesis in natural and synthetic biomaterials reveals that matrix porosity regulates endothelial cell invasion speed and sprout diameter. Acta Biomater. 135, 260–273 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Wang, C. C. B., Hung, C. T. & Mow, V. C. An analysis of the effects of depth-dependent aggregate modulus on articular cartilage stress-relaxation behavior in compression. J. Biomech. 34, 75–84 (2001).

    Article  CAS  PubMed  Google Scholar 

  269. Charbonier, F., Indana, D. & Chaudhuri, O. Tuning viscoelasticity in alginate hydrogels for 3D cell culture studies. Curr. Protoc. 1, e124 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  270. Gjorevski, N. & Lutolf, M. P. Synthesis and characterization of well-defined hydrogel matrices and their application to intestinal stem cell and organoid culture. Nat. Protoc. 12, 2263–2274 (2017).

    Article  CAS  PubMed  Google Scholar 

  271. Kloxin, A. M., Tibbitt, M. W. & Anseth, K. S. Synthesis of photodegradable hydrogels as dynamically tunable cell culture platforms. Nat. Protoc. 5, 1867–1887 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  272. Serban, M. A., Scott, A. & Prestwich, G. D. Use of hyaluronan-derived hydrogels for three-dimensional cell culture and tumor xenografts. Curr. Protoc. Cell Biol. https://doi.org/10.1002/0471143030.cb1014s40 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  273. Caliari, S. R. & Burdick, J. A. A practical guide to hydrogels for cell culture. Nat. Methods 13, 405–414 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Correa, S. et al. Translational applications of hydrogels. Chem. Rev. 121, 11385–11457 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Chan, C. E. & Odde, D. J. Traction dynamics of filopodia on compliant substrates. Science 322, 1687–1691 (2008).

    Article  CAS  PubMed  Google Scholar 

  276. Elosegui-Artola, A. et al. Mechanical regulation of a molecular clutch defines force transmission and transduction in response to matrix rigidity. Nat. Cell Biol. 18, 540–548 (2016).

    Article  CAS  PubMed  Google Scholar 

  277. Kanchanawong, P. et al. Nanoscale architecture of integrin-based cell adhesions. Nature 468, 580–584 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Kumar, S. et al. Viscoelastic retraction of single living stress fibers and its impact on cell shape, cytoskeletal organization, and extracellular matrix mechanics. Biophys. J. 90, 3762–3773 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. del Rio, A. et al. Stretching single talin rod molecules activates vinculin binding. Science 323, 638–641 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  280. Grashoff, C. et al. Measuring mechanical tension across vinculin reveals regulation of focal adhesion dynamics. Nature 466, 263–266 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  281. Dupont, S. et al. Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183 (2011). The authors reveal the role of the transcription co-regulators YAP and TAZ as central regulators of mechanotransduction in 2D culture, mediating cell spreading, proliferation and apoptosis, and differentiation in response to stiffness.

    Article  CAS  PubMed  Google Scholar 

  282. Elosegui-Artola, A. et al. Force triggers YAP nuclear entry by regulating transport across nuclear pores. Cell 171, 1397–1410.e1314 (2017).

    Article  CAS  PubMed  Google Scholar 

  283. Shiu, J.-Y., Aires, L., Lin, Z. & Vogel, V. Nanopillar force measurements reveal actin-cap-mediated YAP mechanotransduction. Nat. Cell Biol. 20, 262–271 (2018).

    Article  CAS  PubMed  Google Scholar 

  284. Oria, R. et al. Force loading explains spatial sensing of ligands by cells. Nature 552, 219–224 (2017).

    Article  CAS  PubMed  Google Scholar 

  285. Hui, E., Gimeno, K. I., Guan, G. & Caliari, S. R. Spatiotemporal control of viscoelasticity in phototunable hyaluronic acid hydrogels. Biomacromolecules 20, 4126–4134 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Hadden William, J. et al. Stem cell migration and mechanotransduction on linear stiffness gradient hydrogels. Proc. Natl Acad. Sci. USA 114, 5647–5652 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  287. Jain, N. & Vogel, V. Spatial confinement downsizes the inflammatory response of macrophages. Nat. Mater. 17, 1134–1144 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Jain, N., Iyer, K. V., Kumar, A. & Shivashankar, G. V. Cell geometric constraints induce modular gene-expression patterns via redistribution of HDAC3 regulated by actomyosin contractility. Proc. Natl Acad. Sci. USA 110, 11349–11354 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors apologize for not being able to cite all the relevant publications in this Review due to space and reference limitations. O.C. acknowledges support from a National Institutes of Health National Cancer Institute grant (R37 CA214136), a National Science Foundation CAREER award (CMMI 1846367) and National Science Foundation grant MCB 2148041.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to planning, writing and editing of the manuscript.

Corresponding author

Correspondence to Ovijit Chaudhuri.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Molecular Cell Biology thanks Kenneth Yamada, Jennifer Young, who co-reviewed Martin Kiwanuka, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Glossary

Advanced glycation end products

Proteins or lipids that are glycated when exposed to sugars, and are a biomarker implicated in many diseases, such as diabetes and atherosclerosis.

Aggrecan

The major proteoglycan in the articular cartilage which provides hydration to the cartilage.

Arginine–glycine–aspartate (RGD) sequences

A three-peptide cell–matrix adhesion motif derived from extracellular matrix proteins such as fibronectin and vitronectin that serves as a binding site for integrins such as αvβ3, α5β1 and αIIbβ3.

Assay for transposase-accessible chromatin using sequencing

(ATAC-seq). A genome wide assay that identifies accessible DNA regions/genome-wide chromatin accessibility.

Creep

A behaviour of viscoelastic materials, the time-dependent deformation or strain of a material under constant force/stress.

Deviatoric stresses

Distortional mechanical stresses that act to change the shape of an object on which they act, without changing its volume.

Fibrillar adhesions

Cell–extracellular matrix adhesions whose shapes are elliptical, often forming along fibrous extracellular matrix.

Filipodia

Actin-rich protrusions that are long and thin, and can be highly dynamic.

Fluorescence resonance energy transfer

Energy transfer between two light-sensitive molecules. The efficiency of this energy transfer is inversely proportional to the sixth power of the distance between the molecules. This can be used to study small changes in distances between molecules.

Focal adhesion kinase

(FAK). A receptor tyrosine kinase protein that localizes to focal complexes at cell–extracellular matrix adhesion sites and plays a crucial role in the several integrin-dependent mechanotransductive pathways.

Focal adhesions

Large cell–matrix adhesions typically formed by cells cultured on stiff 2D substrates, characterized by clustered integrin receptors, localization of proteins such as paxillin, talin, vinculin, and phosphorylated focal adhesion kinase (FAK) and thick actomyosin stress fibres, mediating strong cell–substrate adhesion.

Hydrostatic stresses

Volumetric or dilational stresses that act to increase or decrease the volume of an object on which they act, without changing its shape.

Invadopodia

Actin-rich structures that are present at the basal surface of cells, and that are thought to degrade and apply forces to extracellular matrix.

Lamellipodia

Thin sheet-like protrusions composed of a branched network of actin filaments. Extension of lamellipodia at the leading edge is often implicated in driving cell migration.

Lysyl oxidase

An enzyme that converts lysine molecules into highly reactive aldehydes that form crosslinks in extracellular matrix proteins such as type I collagen and elastin.

Matrix metalloproteinases

Cell-secreted enzymes that are capable of proteolytically degrading various extracellular matrix components.

Mechanical plasticity

A material property that defines the extent to which the material undergoes permanent or irreversible deformation following the application and release of external deformation or loading.

Mechanosensitive ion channels

Ion channels that open or close in response to cell membrane stretch or tension. TRPV4 and PIEZO1 are examples of mechanosensitive ion channels implicated in mechanotransduction.

Mesenchymal stem cells

(MSCs). Multipotent stem cells that are found in the bone marrow, which have been reported to differentiate into osteoblasts, adipocytes, chondrocytes, myocytes and neurons.

Microtentacles

Microtubule-based membrane protrusions which are often observed in detached circulating tumour cells.

MRTF-A

Myocardin-related transcription factor A, a transcription factor that plays a key role in mediating smooth muscle cell differentiation.

Nonlinear elasticity

Elasticity is the ability of a material to retain its initial shape/configuration following the application and release of external deformation or loading. Nonlinear elasticity is defined as the nonlinear relation between stress and strain for a material, in contrast to linear elasticity, which is defined as the linear relation between stress and strain of a material until the material starts to yield.

Poroelasticity

A material property that describes the interaction between fluid flow and solid deformations within a porous material. Cells and extracellular matrices are usually poroelastic.

Proteoglycans

Supramolecules that posses protein as a core and a side chain of sugars.

Reconstituted BM (rBM) matrices

Commercially available matrices such as Matrigel or Geltrex that are derived from Engelbreth–Holm–Swarm tumour, a mouse sarcoma, and that are commonly used for in vitro cell culture and contain laminin, type IV collagen, entactin, perlecan and other components.

RHAMM

Receptor for hyaluronan-mediated motility, a protein which binds to hyaluronan.

Rho signalling

A cell signalling pathway involved in regulation of a wide variety of cell processes, such as cell spreading, survival, proliferation and adhesion.

Stiffness

The resistance to deformation of a specific structure, which is dependent on the Young’s modulus and geometry of the structure. Hence, stiffness is regarded as a property of a specific structure, whereas Young’s modulus is an inherent property of the material.

Strain

A measure of localized deformation in a material, with uniaxial strain typically defined as the ratio of deformation to the original length of the material.

Stress

A measure of force per unit area, which has the unit of pascals.

Stress relaxation

A behaviour of viscoelastic materials referring to the time-dependent change in stress in a material under constant deformation.

Tenascin C

A multimodular extracellular matrix glycoprotein which is expressed in various tissues during development, disease or injury.

Traction force microscopy

A technique that experimentally measures the force-induced displacement field of a substrate with known mechanical properties and uses this to computationally determine the cell–extracellular matrix forces applied to the substrate.

Versican

A large extracellular matrix proteoglycan that is found in various tissues, such as blood vessels and skin.

Viscoelasticity

A property of materials that exhibit some behaviours characteristic of elastic solids and some of viscous liquids, and is characterized by a time-dependent mechanical response (that is, creep or stress relaxation).

YAP and TAZ

Transcription co-activators that shuttle between the nucleus and the cytoplasm and play an important role in mediating mechanotransduction, particularly in 2D. When translocated to the nucleus, YAP and TAZ do not directly bind to DNA but regulate gene expression through their binding to transcription factors of the TEAD family.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saraswathibhatla, A., Indana, D. & Chaudhuri, O. Cell–extracellular matrix mechanotransduction in 3D. Nat Rev Mol Cell Biol 24, 495–516 (2023). https://doi.org/10.1038/s41580-023-00583-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-023-00583-1

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research