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:

Harnessing nanotopography and integrin–matrix interactions to influence stem cell fate

Abstract

Stem cells respond to nanoscale surface features, with changes in cell growth and differentiation mediated by alterations in cell adhesion. The interaction of nanotopographical features with integrin receptors in the cells' focal adhesions alters how the cells adhere to materials surfaces, and defines cell fate through changes in both cell biochemistry and cell morphology. In this Review, we discuss how cell adhesions interact with nanotopography, and we provide insight as to how materials scientists can exploit these interactions to direct stem cell fate and to understand how the behaviour of stem cells in their niche can be controlled. We expect knowledge gained from the study of cell–nanotopography interactions to accelerate the development of next-generation stem cell culture materials and implant interfaces, and to fuel discovery of stem cell therapeutics to support regenerative therapies.

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

Figure 1: The nanoscale structure of focal adhesions, and nanoscale connectivity to the cytoskeleton.
Figure 2: Interaction of cells with RGD groups and nanoscale topography.
Figure 3: Nanoscale disorder and adhesion bridging.
Figure 4: Natural and synthetic nanopatterns that offer templates for MSC differentiation and self-renewal.
Figure 5: Cytoskeletal contraction and intracellular tension is important for MSC differentiation.
Figure 6: Image showing marrow-niche architecture and use of paired nanotopographical controls to aid study of MSC growth and differentiation, and to aid understanding of niche regulation.
Figure 7: A model of direct mechanotransduction considering the MSC as a mechanical unit.

Similar content being viewed by others

References

  1. Harrison, R. On the stereotropism of embryonic cells. Science 34, 279–281 (1911).

    Article  CAS  Google Scholar 

  2. Weiss, P. & Garber, B. Shape and movement of mesenchyme cells as functions of the physical structure of the medium. Proc. Natl Acad. Sci. USA 38, 264–280 (1952).

    Article  CAS  Google Scholar 

  3. Curtis, A. S. G. & Varde, M. Control of cell behaviour: Topological factors. J. Natl Cancer Res. Inst. 33, 15–26 (1964).

    CAS  Google Scholar 

  4. Riveline, D. et al. Focal contacts as mechanosensors: externally applied local mechanical force induces growth of focal contacts by an mDia1-dependent and ROCK-independent mechanism. J. Cell Biol. 153, 1175–1186 (2001).

    Article  CAS  Google Scholar 

  5. Cavalcanti-Adam, E. A. et al. Cell spreading and focal adhesion dynamics are regulated by spacing of integrin ligands. Biophys. J. 92, 2964–2974 (2007).

    Article  CAS  Google Scholar 

  6. Cavalcanti-Adam, E. A., Aydin, D., Hirschfeld-Warneken, V. C. & Spatz, J. P. Cell adhesion and response to synthetic nanopatterned environments by steering receptor clustering and spatial location. HFSP J. 2, 276–285 (2008).

    Article  CAS  Google Scholar 

  7. Schvartzman, M. et al. Nanolithographic control of the spatial organization of cellular adhesion receptors at the single-molecule level. Nano Lett. 11, 1306–1312 (2011).

    Article  CAS  Google Scholar 

  8. Vieu, C. et al. Electron beam lithography: Resolution limits and applications. Appl. Surf. Sci. 164, 111–117 (2000).

    Article  CAS  Google Scholar 

  9. Maheshwari, G., Brown, G., Lauffenburger, D. A., Wells, A. & Griffith, L. G. Cell adhesion and motility depend on nanoscale RGD clustering. J. Cell Sci. 113, 1677–1686 (2000).

    CAS  Google Scholar 

  10. Massia, S. P. & Hubbell, J. A. An RGD spacing of 440 nm is sufficient for integrin αVβ3-mediated fibroblast spreading and 140 nm for focal contact and stress fiber formation. J. Cell Biol. 114, 1089–1100 (1991).

    Article  CAS  Google Scholar 

  11. Kilian, K. A. & Mrksich, M. Directing stem cell fate by controlling the affinity and density of ligand-receptor interactions at the biomaterials interface. Angew. Chem. Int. Ed. 51, 4891–4895 (2012).

    Article  CAS  Google Scholar 

  12. Lo, C. M., Wang, H. B., Dembo, M. & Wang, Y. L. Cell movement is guided by the rigidity of the substrate. Biophys. J. 79, 144–152 (2000).

    Article  CAS  Google Scholar 

  13. 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  Google Scholar 

  14. Discher, D. E., Mooney, D. J. & Zandstra, P. W. Growth factors, matrices, and forces combine and control stem cells. Science 324, 1673–1677 (2009).

    Article  CAS  Google Scholar 

  15. 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  Google Scholar 

  16. Huebsch, N. et al. Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nature Mater. 9, 518–526 (2010).

    Article  CAS  Google Scholar 

  17. Patla, I. et al. Dissecting the molecular architecture of integrin adhesion sites by cryo-electron tomography. Nature Cell Biol. 12, 909–915 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  19. Dalby, M. J., Riehle, M. O., Johnstone, H., Affrossman, S. & Curtis, A. S. Investigating the limits of filopodial sensing: a brief report using SEM to image the interaction between 10 nm high nano-topography and fibroblast filopodia. Cell Biol. Int. 28, 229–236 (2004).

    Article  CAS  Google Scholar 

  20. Teixeira, A. I., Abrams, G. A., Bertics, P. J., Murphy, C. J. & Nealey, P. F. Epithelial contact guidance on well-defined micro- and nanostructured substrates. J. Cell Sci. 116, 1881–1892 (2003).

    Article  CAS  Google Scholar 

  21. Loesberg, W. A. et al. The threshold at which substrate nanogroove dimensions may influence fibroblast alignment and adhesion. Biomaterials 28, 3944–3951 (2007).

    Article  CAS  Google Scholar 

  22. McNamara, L. E. et al. Investigation of the limits of nanoscale filopodial interactions. J. Tissue Eng. (in the press).

  23. Dalby, M. J., Hart, A. & Yarwood, S. J. The effect of the RACK1 signalling protein on the regulation of cell adhesion and cell contact guidance on nanometric grooves. Biomaterials 29, 282–289 (2008).

    Article  CAS  Google Scholar 

  24. Yim, E. K., Darling, E. M., Kulangara, K., Guilak, F. & Leong, K. W. Nanotopography-induced changes in focal adhesions, cytoskeletal organization, and mechanical properties of human mesenchymal stem cells. Biomaterials 31, 1299–1306 (2010).

    Article  CAS  Google Scholar 

  25. Wood, M. A., Bagnaninchi, P. & Dalby, M. J. The β integrins and cytoskeletal nanoimprinting. Exp. Cell Res. 314, 927–935 (2008).

    Article  CAS  Google Scholar 

  26. Curtis, A. S. G., Dalby, M. J. & Gadegaard, N. Nanoprinting onto cells. J. R. Soc. Interface 3, 393–398 (2006).

    Article  CAS  Google Scholar 

  27. Gallagher, J. O., McGhee, K. F., Wilkinson, C. D. & Riehle, M. O. Interaction of animal cells with ordered nanotopography. IEEE Trans. Nanobiosci. 1, 24–28 (2002).

    Article  CAS  Google Scholar 

  28. Dalby, M. J., Gadegaard, N., Riehle, M. O., Wilkinson, C. D. & Curtis, A. S. Investigating filopodia sensing using arrays of defined nano-pits down to 35 nm diameter in size. Int. J. Biochem. Cell Biol. 36, 2015–2025 (2004).

    Article  CAS  Google Scholar 

  29. Anselme, K. et al. The interaction of cells and bacteria with surfaces structured at the nanometre scale. Acta Biomater. 6, 3824–3846 (2010).

    Article  CAS  Google Scholar 

  30. Leven, R. M., Virdi, A. S. & Sumner, D. R. Patterns of gene expression in rat bone marrow stromal cells cultured on titanium alloy discs of different roughness. J. Biomed. Mater. Res. A 70, 391–401 (2004).

    Article  CAS  Google Scholar 

  31. Anselme, K. et al. Qualitative and quantitative study of human osteoblast adhesion on materials with various surface roughnesses. J. Biomed. Mater. Res. 49, 155–166 (2000).

    Article  CAS  Google Scholar 

  32. Anselme, K. et al. The relative influence of the topography and chemistry of TiAl6V4 surfaces on osteoblastic cell behaviour. Biomaterials 21, 1567–1577 (2000).

    Article  CAS  Google Scholar 

  33. Anselme, K. & Bigerelle, M. Topography effects of pure titanium substrates on human osteoblast long-term adhesion. Acta Biomater. 1, 211–222 (2005).

    Article  CAS  Google Scholar 

  34. Dalby, M. J. et al. The control of human mesenchymal cell differentiation using nanoscale symmetry and disorder. Nature Mater. 6, 997–1003 (2007).

    Article  CAS  Google Scholar 

  35. Oh, S. et al. Stem cell fate dictated solely by altered nanotube dimension. Proc. Natl Acad. Sci. USA 106, 2130–2135 (2009).

    Article  CAS  Google Scholar 

  36. Dalby, M. J. et al. Nanotopographical stimulation of mechanotransduction and changes in interphase centromere positioning. J. Cell Biochem. 100, 326–338 (2007).

    Article  CAS  Google Scholar 

  37. Biggs, M. J. et al. Interactions with nanoscale topography: adhesion quantification and signal transduction in cells of osteogenic and multipotent lineage. J. Biomed. Mater. Res. A 91, 195–208 (2009).

    Article  CAS  Google Scholar 

  38. 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  Google Scholar 

  39. 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  Google Scholar 

  40. Kwan, A. P., Cummings, C. E., Chapman, J. A. & Grant, M. E. Macromolecular organization of chicken type X collagen in vitro. J. Cell Biol. 114, 597–604 (1991).

    Article  CAS  Google Scholar 

  41. Stephens, M., Kwan, A. P., Bayliss, M. T. & Archer, C. W. Human articular surface chondrocytes initiate alkaline phosphatase and type X collagen synthesis in suspension culture. J. Cell Sci. 103, 1111–1116 (1992).

    CAS  Google Scholar 

  42. Kingham, E., White, K., Gadegaard, N., Dalby, M. J. & Oreffo, R. O. Nanotopographical cues augment mesenchymal differentiation of human embryonic stem cells. Small 9, 2140–2151 (2013).

    Article  CAS  Google Scholar 

  43. Huang, J. et al. Impact of order and disorder in RGD nanopatterns on cell adhesion. Nano Lett. 9, 1111–1116 (2009).

    Article  CAS  Google Scholar 

  44. Biggs, M. J. et al. The use of nanoscale topography to modulate the dynamics of adhesion formation in primary osteoblasts and ERK/MAPK signalling in STRO-1+ enriched skeletal stem cells. Biomaterials 30, 5094–5103 (2009).

    Article  CAS  Google Scholar 

  45. Malmstrom, J. et al. Large area protein patterning reveals nanoscale control of focal adhesion development. Nano Lett. 10, 686–694 (2010).

    Article  CAS  Google Scholar 

  46. Malmstrom, J. et al. Focal complex maturation and bridging on 200 nm vitronectin but not fibronectin patches reveal different mechanisms of focal adhesion formation. Nano Lett. 11, 2264–2271 (2011).

    Article  CAS  Google Scholar 

  47. Biggs, M. J. et al. Adhesion formation of primary human osteoblasts and the functional response of mesenchymal stem cells to 330 nm deep microgrooves. J. R. Soc. Interface 5, 1231–1242 (2008).

    Article  CAS  Google Scholar 

  48. Das, R. K., Zouani, O. F., Labrugere, C., Oda, R. & Durrieu, M. C. Influence of nanohelical shape and periodicity on stem cell fate. ACS Nano 7, 3351–3361 (2013).

    Article  CAS  Google Scholar 

  49. Tsimbouri, P. M. et al. Using nanotopography and metabolomics to identify biochemical effectors of multipotency. ACS Nano 6, 10239–10249 (2012).

    Article  CAS  Google Scholar 

  50. McMurray, R. J. et al. Nanoscale surfaces for the long-term maintenance of mesenchymal stem cell phenotype and multipotency. Nature Mater. 10, 637–644 (2011).

    Article  CAS  Google Scholar 

  51. Curran, J. M. et al. Introducing dip pen nanolithography as a tool for controlling stem cell behaviour: unlocking the potential of the next generation of smart materials in regenerative medicine. Lab Chip 10, 1662–1670 (2010).

    Article  CAS  Google Scholar 

  52. Yanes, O. et al. Metabolic oxidation regulates embryonic stem cell differentiation. Nature Chem. Biol. 6, 411–417 (2010).

    Article  CAS  Google Scholar 

  53. Reyes, J. M. et al. Metabolic changes in mesenchymal stem cells in osteogenic medium measured by autofluorescence spectroscopy. Stem Cells 24, 1213–1217 (2006).

    Article  CAS  Google Scholar 

  54. Watt, F. M. & Hogan, B. L. Out of Eden: stem cells and their niches. Science 287, 1427–1430 (2000).

    Article  CAS  Google Scholar 

  55. Ehninger, A. & Trumpp, A. The bone marrow stem cell niche grows up: mesenchymal stem cells and macrophages move in. J. Exp. Med. 208, 421–428 (2011).

    Article  CAS  Google Scholar 

  56. Bianco, P. Bone and the hematopoietic niche: a tale of two stem cells. Blood 117, 5281–5288 (2011).

    Article  CAS  Google Scholar 

  57. Crisan, M. et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 3, 301–313 (2008).

    Article  CAS  Google Scholar 

  58. Ji, L., LaPointe, V. L., Evans, N. D. & Stevens, M. M. Changes in embryonic stem cell colony morphology and early differentiation markers driven by colloidal crystal topographical cues. Eur. Cell Mater. 23, 135–146 (2012).

    Article  CAS  Google Scholar 

  59. Chen, W. et al. Nanotopography influences adhesion, spreading, and self-renewal of human embryonic stem cells. ACS Nano 6, 4094–4103 (2012).

    Article  CAS  Google Scholar 

  60. Kong, Y. P., Tu, C., Donovan, P. J. & Yee, A. F. Expression of Oct4 in human embryonic stem cells is dependent on nanotopographical configuration. Acta Biomater. 9, 6369–6380 (2013).

    Article  CAS  Google Scholar 

  61. Dalby, M. J., Gadegaard, N. & Wilkinson, C. D. The response of fibroblasts to hexagonal nanotopography fabricated by electron beam lithography. J. Biomed. Mater. Res. A 84, 973–979 (2008).

    Article  CAS  Google Scholar 

  62. Kingham, E. & Oreffo, R. O. Embryonic and induced pluripotent stem cells: Understanding, creating, and exploiting the nano-niche for regenerative medicine. ACS Nano 7, 1867–1881 (2013).

    Article  CAS  Google Scholar 

  63. Benoit, D. S., Schwartz, M. P., Durney, A. R. & Anseth, K. S. Small functional groups for controlled differentiation of hydrogel-encapsulated human mesenchymal stem cells. Nature Mater. 7, 816–823 (2008).

    Article  CAS  Google Scholar 

  64. Curran, J. M., Chen, R. & Hunt, J. A. The guidance of human mesenchymal stem cell differentiation in vitro by controlled modifications to the cell substrate. Biomaterials 27, 4783–4793 (2006).

    Article  CAS  Google Scholar 

  65. Curran, J. M., Chen, R. & Hunt, J. A. Controlling the phenotype and function of mesenchymal stem cells in vitro by adhesion to silane-modified clean glass surfaces. Biomaterials 26, 7057–7067 (2005).

    Article  CAS  Google Scholar 

  66. Dalby, M. J., Riehle, M. O., Johnstone, H., Affrossman, S. & Curtis, A. S. In vitro reaction of endothelial cells to polymer demixed nanotopography. Biomaterials 23, 2945–2954 (2002).

    Article  CAS  Google Scholar 

  67. Dalby, M. J. et al. Rapid fibroblast adhesion to 27 nm high polymer demixed nano-topography. Biomaterials 25, 77–83 (2004).

    Article  CAS  Google Scholar 

  68. Dalby, M. J. et al. Genomic expression of mesenchymal stem cells to altered nanoscale topographies. J. R. Soc. Interface 5, 1055–1065 (2008).

    Article  Google Scholar 

  69. Sjostrom, T. et al. Fabrication of pillar-like titania nanostructures on titanium and their interactions with human skeletal stem cells. Acta Biomater. 5, 1433–1441 (2009).

    Article  CAS  Google Scholar 

  70. Lim, J. Y. et al. The regulation of integrin-mediated osteoblast focal adhesion and focal adhesion kinase expression by nanoscale topography. Biomaterials 28, 1787–1797 (2007).

    Article  CAS  Google Scholar 

  71. Gadegaard, N., Dalby, M. J., Riehle, M. O. & Wilkinson, C. D. W. Optimizing substrate disorder for bone tissue engineering of mesenchymal stem cells. J. Vac. Sci. Technol. B 26, 2554–2557 (2008).

    Article  CAS  Google Scholar 

  72. Chen, C. S., Mrksich, M., Huang, S., Whitesides, G. M. & Ingber, D. E. Geometric control of cell life and death. Science 276, 1425–1428 (1997).

    Article  CAS  Google Scholar 

  73. Ingber, D. E. Mechanical control of tissue growth: Function follows form. Proc. Natl Acad. Sci. USA 102, 11571–11572 (2005).

    Article  CAS  Google Scholar 

  74. Hamamura, K., Jiang, C. & Yokota, H. ECM-dependent mRNA expression profiles and phosphorylation patterns of p130Cas, FAK, ERK and p38 MAPK of osteoblast-like cells. Cell Biol. Int. 34, 1005–1012 (2010).

    Article  CAS  Google Scholar 

  75. Heydarkhan-Hagvall, S. et al. Influence of systematically varied nano-scale topography on cell morphology and adhesion. Cell Commun. Adhes. 14, 181–194 (2007).

    Article  CAS  Google Scholar 

  76. Hamilton, D. W. & Brunette, D. M. The effect of substratum topography on osteoblast adhesion mediated signal transduction and phosphorylation. Biomaterials 28, 1806–1819 (2007).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  78. Rajnicek, A. M., Foubister, L. E. & McCaig, C. D. Alignment of corneal and lens epithelial cells by co-operative effects of substratum topography and DC electric fields. Biomaterials 29, 2082–2095 (2008).

    Article  CAS  Google Scholar 

  79. Lim, S. T. Nuclear FAK: a new mode of gene regulation from cellular adhesions. Mol. Cells 36, 1–6 (2013).

    Article  CAS  Google Scholar 

  80. Lim, S. T. et al. Pyk2 inhibition of p53 as an adaptive and intrinsic mechanism facilitating cell proliferation and survival. J. Biol. Chem. 285, 1743–1753 (2010).

    Article  CAS  Google Scholar 

  81. Lim, S. T. et al. Nuclear FAK promotes cell proliferation and survival through FERM-enhanced p53 degradation. Mol. Cell 29, 9–22 (2008).

    Article  CAS  Google Scholar 

  82. Nikukar, H. et al. Osteogenesis of mesenchymal stem cells by nanoscale mechanotransduction. ACS Nano 7, 2758–2767 (2013).

    Article  CAS  Google Scholar 

  83. Fu, L. et al. Stimulation of osteogenic differentiation and inhibition of adipogenic differentiation in bone marrow stromal cells by alendronate via ERK and JNK activation. Bone 43, 40–47 (2008).

    Article  CAS  Google Scholar 

  84. Xiao, G., Jiang, D., Gopalakrishnan, R. & Franceschi, R. T. Fibroblast growth factor 2 induction of the osteocalcin gene requires MAPK activity and phosphorylation of the osteoblast transcription factor, Cbfa1/Runx2. J. Biol. Chem. 277, 36181–36187 (2002).

    Article  CAS  Google Scholar 

  85. Ge, C., Xiao, G., Jiang, D. & Franceschi, R. T. Critical role of the extracellular signal-regulated kinase-MAPK pathway in osteoblast differentiation and skeletal development. J. Cell Biol. 176, 709–718 (2007).

    Article  CAS  Google Scholar 

  86. Nikonova, A. S., Astsaturov, I., Serebriiskii, I. G., Dunbrack, R. L. Jr & Golemis, E. A. Aurora A kinase (AURKA) in normal and pathological cell division. Cell Mol. Life Sci. 70, 661–687 (2012).

    Article  CAS  Google Scholar 

  87. Zhu, C. & Jiang, W. Cell cycle-dependent translocation of PRC1 on the spindle by Kif4 is essential for midzone formation and cytokinesis. Proc. Natl Acad. Sci. USA 102, 343–348 (2005).

    Article  CAS  Google Scholar 

  88. Amati, B. & Vlach, J. Kip1 meets SKP2: new links in cell-cycle control. Nature Cell Biol. 1, E91–E93 (1999).

    Article  CAS  Google Scholar 

  89. Brunet, S. et al. Meiotic regulation of TPX2 protein levels governs cell cycle progression in mouse oocytes. PLoS ONE 3, e3338 (2008).

    Article  CAS  Google Scholar 

  90. Ingber, D. E. Tensegrity I. Cell structure and hierarchical systems biology. J. Cell Sci. 116, 1157–1173 (2003).

    Article  CAS  Google Scholar 

  91. Mammoto, T. & Ingber, D. E. Mechanical control of tissue and organ development. Development 137, 1407–1420 (2010).

    Article  CAS  Google Scholar 

  92. Ingber, D. Cellular tensegrity: defining new rules of biological design that govern the cytoskeleton. J. Cell Sci. 104, 613–627 (1993).

    Google Scholar 

  93. Ingber, D. E. Cellular mechanotransduction: putting all the pieces together again. FASEB J. 20, 811–827 (2006).

    Article  CAS  Google Scholar 

  94. Wang, N. et al. Mechanical behavior in living cells consistent with the tensegrity model. Proc. Natl Acad. Sci. USA 98, 7765–7770 (2001).

    Article  CAS  Google Scholar 

  95. Guarda, A., Bolognese, F., Bonapace, I. M. & Badaracco, G. Interaction between the inner nuclear membrane lamin B receptor and the heterochroma–tic methyl binding protein, MeCP2. Exp. Cell Res. 315, 1895–1903 (2009).

    Article  CAS  Google Scholar 

  96. Tzur, Y. B., Wilson, K. L. & Gruenbaum, Y. SUN-domain proteins: 'Velcro' that links the nucleoskeleton to the cytoskeleton. Nature Rev. Mol. Cell Biol. 7, 782–788 (2006).

    Article  CAS  Google Scholar 

  97. Ostlund, C. et al. Dynamics and molecular interactions of linker of nucleoskeleton and cytoskeleton (LINC) complex proteins. J. Cell Sci. 122, 4099–4108 (2009).

    Article  CAS  Google Scholar 

  98. Cremer, T. & Cremer, C. Chromosome territories, nuclear architecture and gene regulation in mammalian cells. Nature Rev. Genet. 2, 292–301 (2001).

    Article  CAS  Google Scholar 

  99. Boyle, S. et al. The spatial organization of human chromosomes within the nuclei of normal and emerin-mutant cells. Hum. Mol. Genet. 10, 211–219 (2001).

    Article  CAS  Google Scholar 

  100. Bolzer, A. et al. Three-dimensional maps of all chromosomes in human male fibroblast nuclei and prometaphase rosettes. PLoS Biol. 3, e157 (2005).

    Article  CAS  Google Scholar 

  101. Heslop-Harrison, J. S., Leitch, A. R. & Schwarzacher, T. in The Chromosome (eds Heslop-Harrison, J. S. & Flavell, R. B.) 221–232 (Bios, 1993).

    Google Scholar 

  102. Felsenfeld, G. & Groudine, M. Controlling the double helix. Nature 421, 448–453 (2003).

    Article  CAS  Google Scholar 

  103. Wang, N. & Suo, Z. Long-distance propagation of forces in a cell. Biochem. Biophys. Res. Commun. 328, 1133–1138 (2005).

    Article  CAS  Google Scholar 

  104. Wang, N. et al. Mechanical behavior in living cells consistent with the tensegrity model. Proc. Natl Acad. Sci. USA 98, 7765–7770 (2001).

    Article  CAS  Google Scholar 

  105. Dalby, M. J. et al. Nanomechanotransduction and interphase nuclear organization influence on genomic control. J. Cell Biochem. 102, 1234–1244 (2007).

    Article  CAS  Google Scholar 

  106. Tsimbouri, P. M. et al. A genomics approach in determining nanotopographical effects on MSC phenotype. Biomaterials 34, 2177–2184 (2013).

    Article  CAS  Google Scholar 

  107. Osborne, C. S. et al. Active genes dynamically colocalize to shared sites of ongoing transcription. Nature Genet. 36, 1065–1071 (2004).

    Article  CAS  Google Scholar 

  108. Maniotis, A. J., Chen, C. S. & Ingber, D. E. Demonstration of mechanical connections between integrins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure. Proc. Natl Acad. Sci. USA 94, 849–854 (1997).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  110. Hench, L. L. & Polak, J. M. Third-generation biomedical materials. Science 295, 1014–1017 (2002).

    Article  CAS  Google Scholar 

  111. Lavenus, S., Ricquier, J. C., Louarn, G. & Layrolle, P. Cell interaction with nanopatterned surface of implants. Nanomedicine 5, 937–947 (2010).

    Article  CAS  Google Scholar 

  112. Bigerelle, M. & Anselme, K. Bootstrap analysis of the relation between initial adhesive events and long-term cellular functions of human osteoblasts cultured on biocompatible metallic substrates. Acta Biomater. 1, 499–510 (2005).

    Article  CAS  Google Scholar 

  113. Sjostrom, T., McNamara, L. E., Meek, R. M., Dalby, M. J. & Su, B. 2D and 3D nanopatterning of titanium for enhancing osteoinduction of stem cells at implant surfaces. Adv. Healthcare Mater. 2, 1285–1293 (2013).

    Article  CAS  Google Scholar 

  114. Downing, T. L. et al. Biophysical regulation of epigenetic state and cell reprogramming. Nature Mater. 12, 1154–1162 (2013).

    Article  CAS  Google Scholar 

  115. Anderson, D. G., Levenberg, S. & Langer, R. Nanoliter-scale synthesis of arrayed biomaterials and application to human embryonic stem cells. Nature Biotechnol. 22, 863–866 (2004).

    Article  CAS  Google Scholar 

  116. Khan, F., Tare, R. S., Kanczler, J. M., Oreffo, R. O. & Bradley, M. Strategies for cell manipulation and skeletal tissue engineering using high-throughput polymer blend formulation and microarray techniques. Biomaterials 31, 2216–2228 (2010).

    Article  CAS  Google Scholar 

  117. Unadkat, H. V. et al. An algorithm-based topographical biomaterials library to instruct cell fate. Proc. Natl Acad. Sci. USA 108, 16565–16570 (2011).

    Article  CAS  Google Scholar 

  118. Gobaa, S. et al. Artificial niche microarrays for probing single stem cell fate in high throughput. Nature Methods 8, 949–955 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

M.J.D, N.G. and R.O.C.O. are funded by grants from BBSRC, EPSRC and MRC. We gratefully acknowledge the encouragement and support of M. Riehle, A. Curtis and the late C. Wilkinson. We thank C. Ranson (Glasgow School of Art) for illustrations of the MSC niche and mechanotransduction. We thank R. Fraser and A. Warren (University of Sydney) for interesting discussions on, and images of, fenestrae. Finally, we thank W. Cushley, C. Berry and J. Dalby for critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Matthew J. Dalby.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dalby, M., Gadegaard, N. & Oreffo, R. Harnessing nanotopography and integrin–matrix interactions to influence stem cell fate. Nature Mater 13, 558–569 (2014). https://doi.org/10.1038/nmat3980

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmat3980

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing