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 migration: implications for repair and regeneration in joint disease

Abstract

Connective tissues within the synovial joints are characterized by their dense extracellular matrix and sparse cellularity. With injury or disease, however, tissues commonly experience an influx of cells owing to proliferation and migration of endogenous mesenchymal cell populations, as well as invasion of the tissue by other cell types, including immune cells. Although this process is critical for successful wound healing, aberrant immune-mediated cell infiltration can lead to pathological inflammation of the joint. Importantly, cells of mesenchymal or haematopoietic origin use distinct modes of migration and thus might respond differently to similar biological cues and microenvironments. Furthermore, cell migration in the physiological microenvironment of musculoskeletal tissues differs considerably from migration in vitro. This Review addresses the complexities of cell migration in fibrous connective tissues from three separate but interdependent perspectives: physiology (including the cellular and extracellular factors affecting 3D cell migration), pathophysiology (cell migration in the context of synovial joint autoimmune disease and injury) and tissue engineering (cell migration in engineered biomaterials). Improved understanding of the fundamental mechanisms governing interstitial cell migration might lead to interventions that stop invasion processes that culminate in deleterious outcomes and/or that expedite migration to direct endogenous cell-mediated repair and regeneration of joint tissues.

Key points

  • Interstitial cell migration in the fibrous microenvironments of intra-articular tissues is regulated by biophysical and biochemical factors.

  • Immune cells are recruited to and retained within the synovium by inflammatory cytokines and chemokines in rheumatic disorders.

  • High matrix density and stiffness of adult dense connective tissues restrict the mobility of endogenous cells, impeding wound healing after injury.

  • Early cell migration into biomaterial scaffolds is a critical but challenging step towards engineering functional musculoskeletal tissues.

  • Targeted strategies that limit inflammatory cell invasion while promoting the migration of endogenous reparative cells might enhance joint tissue formation and regeneration.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Connective tissue type and migration capacity.
Fig. 2: Cell migration during joint development, disease and repair.
Fig. 3: Designing engineered scaffolds to promote cell migration and tissue repair.

Similar content being viewed by others

References

  1. Franz, C. M., Jones, G. E. & Ridley, A. J. Cell migration in development and disease. Dev. Cell 2, 153–158 (2002).

    CAS  PubMed  Google Scholar 

  2. Cukierman, E., Pankov, R., Stevens, D. R. & Yamada, K. M. Taking cell-matrix adhesions to the third dimension. Science 294, 1708–1712 (2001).

    CAS  PubMed  Google Scholar 

  3. 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 

  4. Pathak, A. & Kumar, S. Biophysical regulation of tumor cell invasion: moving beyond matrix stiffness. Integr. Biol. 3, 267–278 (2011).

    CAS  Google Scholar 

  5. Friedl, P. & Wolf, K. Plasticity of cell migration: a multiscale tuning model. J. Cell Biol. 188, 11–19 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Stoker, M. & Gherardi, E. Regulation of cell movement: the motogenic cytokines. Biochim. Biophys. Acta 1072, 81–102 (1991).

    CAS  PubMed  Google Scholar 

  8. Walters, N. J. & Gentleman, E. Evolving insights in cell-matrix interactions: elucidating how non-soluble properties of the extracellular niche direct stem cell fate. Acta Biomater. 11, 3–16 (2015).

    CAS  PubMed  Google Scholar 

  9. Petrie, R. J. & Yamada, K. M. At the leading edge of three-dimensional cell migration. J. Cell Sci. 125, 5917–5926 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Kurosaka, S. & Kashina, A. Cell biology of embryonic migration. Birth Defects Res. C Embryo Today 84, 102–122 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Friedl, P., Zanker, K. S. & Brocker, E. B. Cell migration strategies in 3D extracellular matrix: differences in morphology, cell matrix interactions, and integrin function. Microsc. Res. Tech. 43, 369–378 (1998).

    CAS  PubMed  Google Scholar 

  13. Friedl, P. & Brocker, E. B. T cell migration in three-dimensional extracellular matrix: guidance by polarity and sensations. Dev. Immunol. 7, 249–266 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Liu, Y. J. et al. Confinement and low adhesion induce fast amoeboid migration of slow mesenchymal cells. Cell 160, 659–672 (2015).

    CAS  PubMed  Google Scholar 

  15. Ruprecht, V. et al. Cortical contractility triggers a stochastic switch to fast amoeboid cell motility. Cell 160, 673–685 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Petrie, R. J., Koo, H. & Yamada, K. M. Generation of compartmentalized pressure by a nuclear piston governs cell motility in a 3D matrix. Science 345, 1062–1065 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Mouw, J. K., Ou, G. & Weaver, V. M. Extracellular matrix assembly: a multiscale deconstruction. Nat. Rev. Mol. Cell. Biol. 15, 771–785 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Lu, P., Takai, K., Weaver, V. M. & Werb, Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol. 3, a005058 (2011).

    PubMed  PubMed Central  Google Scholar 

  20. Friedl, P. & Brocker, E. B. The biology of cell locomotion within three-dimensional extracellular matrix. Cell. Mol. Life Sci. 57, 41–64 (2000).

    CAS  PubMed  Google Scholar 

  21. Friedl, P. & Wolf, K. Proteolytic interstitial cell migration: a five-step process. Cancer Metastasis Rev. 28, 129–135 (2009).

    PubMed  Google Scholar 

  22. Guilak, F., Nims, R., Dicks, A., Wu, C.-L. & Meulenbelt, I. Osteoarthritis as a disease of the cartilage pericellular matrix. Matrix Biol. 71–72, 40–50 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Guilak, F., Tedrow, J. R. & Burgkart, R. Viscoelastic properties of the cell nucleus. Biochem. Biophys. Res. Commun. 269, 781–786 (2000).

    CAS  PubMed  Google Scholar 

  24. Lautscham, L. A. et al. Migration in confined 3D environments is determined by a combination of adhesiveness, nuclear volume, contractility, and cell stiffness. Biophys. J. 109, 900–913 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Friedl, P., Wolf, K. & Lammerding, J. Nuclear mechanics during cell migration. Curr. Opin. Cell Biol. 23, 55–64 (2011).

    CAS  PubMed  Google Scholar 

  26. Denais, C. M. et al. Nuclear envelope rupture and repair during cancer cell migration. Science 352, 353–358 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Rowat, A. C. et al. Nuclear envelope composition determines the ability of neutrophil-type cells to passage through micron-scale constrictions. J. Biol. Chem. 288, 8610–8618 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Pajerowski, J. D., Dahl, K. N., Zhong, F. L., Sammak, P. J. & Discher, D. E. Physical plasticity of the nucleus in stem cell differentiation. Proc. Natl Acad. Sci. USA 104, 15619–15624 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Lammerding, J. et al. Lamins A and C but not lamin B1 regulate nuclear mechanics. J. Biol. Chem. 281, 25768–25780 (2006).

    CAS  PubMed  Google Scholar 

  31. Booth-Gauthier, E. A. et al. Hutchinson-Gilford progeria syndrome alters nuclear shape and reduces cell motility in three dimensional model substrates. Integr. Biol. 5, 569–577 (2013).

    CAS  Google Scholar 

  32. Greiner, A. M. et al. Multifunctional polymer scaffolds with adjustable pore size and chemoattractant gradients for studying cell matrix invasion. Biomaterials 35, 611–619 (2014).

    CAS  PubMed  Google Scholar 

  33. Infante, E. et al. LINC complex-Lis1 interplay controls MT1-MMP matrix digest-on-demand response for confined tumor cell migration. Nat. Commun. 9, 2443 (2018).

    PubMed  PubMed Central  Google Scholar 

  34. Olins, A. L. et al. Nuclear envelope and chromatin compositional differences comparing undifferentiated and retinoic acid- and phorbol ester-treated HL-60 cells. Exp. Cell Res. 268, 115–127 (2001).

    CAS  PubMed  Google Scholar 

  35. Ekpenyong, A. E. et al. Viscoelastic properties of differentiating blood cells are fate- and function-dependent. PLOS ONE 7, e45237 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Graham, D. M. et al. Enucleated cells reveal differential roles of the nucleus in cell migration, polarity, and mechanotransduction. J. Cell Biol. 217, 895–914 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Singh, S. P., Schwartz, M. P., Lee, J. Y., Fairbanks, B. D. & Anseth, K. S. A peptide functionalized poly(ethylene glycol) (PEG) hydrogel for investigating the influence of biochemical and biophysical matrix properties on tumor cell migration. Biomater. Sci. 2, 1024–1034 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Lowin, T. & Straub, R. H. Integrins and their ligands in rheumatoid arthritis. Arthritis Res. Ther. 13, 244 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Loeser, R. F. Integrins and chondrocyte-matrix interactions in articular cartilage. Matrix Biol. 39, 11–16 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Hinz, B. The extracellular matrix and transforming growth factor-β1: tale of a strained relationship. Matrix Biol. 47, 54–65 (2015).

    CAS  PubMed  Google Scholar 

  41. Ni, G. X., Li, Z. & Zhou, Y. Z. The role of small leucine-rich proteoglycans in osteoarthritis pathogenesis. Osteoarthr. Cartil. 22, 896–903 (2014).

    Google Scholar 

  42. Merline, R., Schaefer, R. M. & Schaefer, L. The matricellular functions of small leucine-rich proteoglycans (SLRPs). J. Cell Commun. Signal 3, 323–335 (2009).

    PubMed  PubMed Central  Google Scholar 

  43. Tufvesson, E. & Westergren-Thorsson, G. Tumour necrosis factor-alpha interacts with biglycan and decorin. FEBS Lett. 530, 124–128 (2002).

    CAS  PubMed  Google Scholar 

  44. Dormann, D. & Weijer, C. J. Chemotactic cell movement during development. Curr. Opin. Genet. Dev. 13, 358–364 (2003).

    CAS  PubMed  Google Scholar 

  45. Wu, J. et al. Gradient biomaterials and their influences on cell migration. Interface Focus 2, 337–355 (2012).

    PubMed  PubMed Central  Google Scholar 

  46. Turner, M. D., Nedjai, B., Hurst, T. & Pennington, D. J. Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim. Biophys. Acta 1843, 2563–2582 (2014).

    CAS  PubMed  Google Scholar 

  47. Qu, F. et al. Maturation state and matrix microstructure regulate interstitial cell migration in dense connective tissues. Sci. Rep. 8, 3295 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  50. 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).

    CAS  PubMed  Google Scholar 

  51. Zaman, M. H. et al. Migration of tumor cells in 3D matrices is governed by matrix stiffness along with cell-matrix adhesion and proteolysis. Proc. Natl Acad. Sci. USA 103, 10889–10894 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Pathak, A. & Kumar, S. Independent regulation of tumor cell migration by matrix stiffness and confinement. Proc. Natl Acad. Sci. USA 109, 10334–10339 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Peyton, S. R. et al. Marrow-derived stem cell motility in 3D synthetic scaffold is governed by geometry along with adhesivity and stiffness. Biotechnol. Bioeng. 108, 1181–1193 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Zaman, M. H., Matsudaira, P. & Lauffenburger, D. A. Understanding effects of matrix protease and matrix organization on directional persistence and translational speed in three-dimensional cell migration. Ann. Biomed. Eng. 35, 91–100 (2007).

    PubMed  Google Scholar 

  55. Kim, M. C. et al. Integrating focal adhesion dynamics, cytoskeleton remodeling, and actin motor activity for predicting cell migration on 3D curved surfaces of the extracellular matrix. Integr. Biol. 4, 1386–1397 (2012).

    CAS  Google Scholar 

  56. Provenzano, P. P., Inman, D. R., Eliceiri, K. W., Trier, S. M. & Keely, P. J. Contact guidance mediated three-dimensional cell migration is regulated by Rho/ROCK-dependent matrix reorganization. Biophys. J. 95, 5374–5384 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Petrie, R. J., Doyle, A. D. & Yamada, K. M. Random versus directionally persistent cell migration. Nat. Rev. Mol. Cell. Biol. 10, 538–549 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Dickinson, R. B., Guido, S. & Tranquillo, R. T. Biased cell migration of fibroblasts exhibiting contact guidance in oriented collagen gels. Ann. Biomed. Eng. 22, 342–356 (1994).

    CAS  PubMed  Google Scholar 

  60. Riching, K. M. et al. 3D collagen alignment limits protrusions to enhance breast cancer cell persistence. Biophys. J. 107, 2546–2558 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Smith, M. D. The normal synovium. Open Rheumatol. J. 5, 100–106 (2011).

    PubMed  PubMed Central  Google Scholar 

  62. Nevius, E., Gomes, A. C. & Pereira, J. P. Inflammatory cell migration in rheumatoid arthritis: a comprehensive review. Clin. Rev. Allergy Immunol. 51, 59–78 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Tak, P. P. et al. Analysis of the synovial cell infiltrate in early rheumatoid synovial tissue in relation to local disease activity. Arthritis. Rheum. 40, 217–225 (1997).

    CAS  PubMed  Google Scholar 

  64. Mulherin, D., Fitzgerald, O. & Bresnihan, B. Synovial tissue macrophage populations and articular damage in rheumatoid arthritis. Arthritis Rheum. 39, 115–124 (1996).

    CAS  PubMed  Google Scholar 

  65. Mellado, M. et al. T cell migration in rheumatoid arthritis. Front. Immunol. 6, 384 (2015).

    PubMed  PubMed Central  Google Scholar 

  66. Iwamoto, T., Okamoto, H., Toyama, Y. & Momohara, S. Molecular aspects of rheumatoid arthritis: chemokines in the joints of patients. FEBS J. 275, 4448–4455 (2008).

    CAS  PubMed  Google Scholar 

  67. Koelink, P. J. et al. Targeting chemokine receptors in chronic inflammatory diseases: an extensive review. Pharmacol. Ther. 133, 1–18 (2012).

    CAS  PubMed  Google Scholar 

  68. Buckley, C. D. Why does chronic inflammatory joint disease persist? Clin. Med. 3, 361–366 (2003).

    Google Scholar 

  69. Solari, R., Pease, J. E. & Begg, M. Chemokine receptors as therapeutic targets: why aren’t there more drugs? Eur. J. Pharmacol. 746, 363–367 (2015).

    CAS  PubMed  Google Scholar 

  70. Hitchon, C. A. & El-Gabalawy, H. S. The synovium in rheumatoid arthritis. Open Rheumatol. J. 5, 107–114 (2011).

    PubMed  PubMed Central  Google Scholar 

  71. Amin, M. A. et al. Interleukin-18 induces angiogenic factors in rheumatoid arthritis synovial tissue fibroblasts via distinct signaling pathways. Arthritis Rheum. 56, 1787–1797 (2007).

    CAS  PubMed  Google Scholar 

  72. Bruhl, H. et al. Functional expression of the chemokine receptor CCR7 on fibroblast-like synoviocytes. Rheumatology 47, 1771–1774 (2008).

    CAS  Google Scholar 

  73. Garcia-Vicuna, R. et al. CC and CXC chemokine receptors mediate migration, proliferation, and matrix metalloproteinase production by fibroblast-like synoviocytes from rheumatoid arthritis patients. Arthritis Rheum. 50, 3866–3877 (2004).

    CAS  PubMed  Google Scholar 

  74. Nanki, T., Nagasaka, K., Hayashida, K., Saita, Y. & Miyasaka, N. Chemokines regulate IL-6 and IL-8 production by fibroblast-like synoviocytes from patients with rheumatoid arthritis. J. Immunol. 167, 5381–5385 (2001).

    CAS  PubMed  Google Scholar 

  75. Lefevre, S. et al. Disease-specific effects of matrix and growth factors on adhesion and migration of rheumatoid synovial fibroblasts. J. Immunol. 198, 4588–4595 (2017).

    CAS  PubMed  Google Scholar 

  76. Tolboom, T. C. et al. Invasive properties of fibroblast-like synoviocytes: correlation with growth characteristics and expression of MMP-1, MMP-3, and MMP-10. Ann. Rheum. Dis. 61, 975–980 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Li, D., Xiao, Z., Wang, G. & Song, X. Knockdown of ADAM10 inhibits migration and invasion of fibroblast-like synoviocytes in rheumatoid arthritis. Mol. Med. Rep. 12, 5517–5523 (2015).

    CAS  PubMed  Google Scholar 

  78. Tolboom, T. C. et al. Invasiveness of fibroblast-like synoviocytes is an individual patient characteristic associated with the rate of joint destruction in patients with rheumatoid arthritis. Arthritis Rheum. 52, 1999–2002 (2005).

    PubMed  Google Scholar 

  79. El-Zayadi, A. A. et al. Interleukin-22 drives the proliferation, migration and osteogenic differentiation of mesenchymal stem cells: a novel cytokine that could contribute to new bone formation in spondyloarthropathies. Rheumatology 56, 488–493 (2017).

    CAS  Google Scholar 

  80. Justa, S., Zhou, X. & Sarkar, S. Endogenous IL-22 plays a dual role in arthritis: regulation of established arthritis via IFN-gamma responses. PLOS ONE 9, e93279 (2014).

    PubMed  PubMed Central  Google Scholar 

  81. Qin, Y. et al. Increased CCL19 and CCL21 levels promote fibroblast ossification in ankylosing spondylitis hip ligament tissue. BMC Musculoskelet. Disord. 15, 316 (2014).

    PubMed  PubMed Central  Google Scholar 

  82. Lee, J. et al. Stimulation of osteoclast migration and bone resorption by C-C chemokine ligands 19 and 21. Exp. Mol. Med. 49, e358 (2017).

    PubMed  PubMed Central  Google Scholar 

  83. Meng, X. H. et al. Quantitative evaluation of knee cartilage and meniscus destruction in patients with rheumatoid arthritis using T1rho and T2 mapping. Eur. J. Radiol. 96, 91–97 (2017).

    PubMed  Google Scholar 

  84. Fuhrmann, I. K., Steinhagen, J., Ruther, W. & Schumacher, U. Comparative immunohistochemical evaluation of the zonal distribution of extracellular matrix and inflammation markers in human meniscus in osteoarthritis and rheumatoid arthritis. Acta Histochem. 117, 243–254 (2015).

    CAS  PubMed  Google Scholar 

  85. Lopez-Franco, M. et al. Meniscal degeneration in human knee osteoarthritis: in situ hybridization and immunohistochemistry study. Arch. Orthop. Trauma Surg. 136, 175–183 (2016).

    PubMed  Google Scholar 

  86. van de Sande, M. A., de Groot, J. H. & Rozing, P. M. Clinical implications of rotator cuff degeneration in the rheumatic shoulder. Arthritis Rheum. 59, 317–324 (2008).

    PubMed  Google Scholar 

  87. Meyer, C. et al. Rheumatoid arthritis affecting the upper cervical spine: biomechanical assessment of the stabilizing ligaments. Biomed. Res. Int. 2017, 6131703 (2017).

    PubMed  PubMed Central  Google Scholar 

  88. Puttlitz, C. M. et al. Biomechanical rationale for the pathology of rheumatoid arthritis in the craniovertebral junction. Spine 25, 1607–1616 (2000).

    CAS  PubMed  Google Scholar 

  89. Yang, G., Rothrauff, B. B. & Tuan, R. S. Tendon and ligament regeneration and repair: clinical relevance and developmental paradigm. Birth Defects Res. C Embryo Today 99, 203–222 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. McNulty, A. L., Moutos, F. T., Weinberg, J. B. & Guilak, F. Enhanced integrative repair of the porcine meniscus in vitro by inhibition of interleukin-1 or tumor necrosis factor alpha. Arthritis Rheum. 56, 3033–3042 (2007).

    CAS  PubMed  Google Scholar 

  91. Mohanraj, B. et al. Chondrocyte and mesenchymal stem cell derived engineered cartilage exhibits differential sensitivity to pro-inflammatory cytokines. J. Orthop. Res. 36, 2901–2910 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  92. Arnoczky, S. P. & Warren, R. F. The microvasculature of the meniscus and its response to injury. An experimental study in the dog. Am. J. Sports Med. 11, 131–141 (1983).

    CAS  PubMed  Google Scholar 

  93. Hiraki, Y. & Shukunami, C. Angiogenesis inhibitors localized in hypovascular mesenchymal tissues: chondromodulin-I and tenomodulin. Connect. Tissue Res. 46, 3–11 (2005).

    CAS  PubMed  Google Scholar 

  94. Manske, P. R., Lesker, P. A., Gelberman, R. H. & Rucinsky, T. E. Intrinsic restoration of the flexor tendon surface in the nonhuman primate. J. Hand Surg. Am. 10, 632–637 (1985).

    CAS  PubMed  Google Scholar 

  95. Sharma, P. & Maffulli, N. Biology of tendon injury: healing, modeling and remodeling. J. Musculoskelet. Neuronal Interact. 6, 181–190 (2006).

    CAS  PubMed  Google Scholar 

  96. Lo, I. K., Chi, S., Ivie, T., Frank, C. B. & Rattner, J. B. The cellular matrix: a feature of tensile bearing dense soft connective tissues. Histol. Histopathol. 17, 523–537 (2002).

    CAS  PubMed  Google Scholar 

  97. Cadby, J. A., Buehler, E., Godbout, C., van Weeren, P. R. & Snedeker, J. G. Differences between the cell populations from the peritenon and the tendon core with regard to their potential implication in tendon repair. PLOS ONE 9, e92474 (2014).

    PubMed  PubMed Central  Google Scholar 

  98. Shen, W. et al. Intra-articular injection of human meniscus stem/progenitor cells promotes meniscus regeneration and ameliorates osteoarthritis through stromal cell-derived factor-1/CXCR4-mediated homing. Stem Cells Transl Med. 3, 387–394 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Mauck, R. L., Martinez-Diaz, G. J., Yuan, X. & Tuan, R. S. Regional multilineage differentiation potential of meniscal fibrochondrocytes: implications for meniscus repair. Anat. Rec. 290, 48–58 (2007).

    CAS  Google Scholar 

  100. Mesiha, M. et al. Pathologic characteristics of the torn human meniscus. Am. J. Sports Med. 35, 103–112 (2007).

    PubMed  Google Scholar 

  101. de Albornoz, P. M. & Forriol, F. The meniscal healing process. Muscles Ligaments Tendons J. 2, 10–18 (2012).

    PubMed  PubMed Central  Google Scholar 

  102. Newman, A. P., Anderson, D. R., Daniels, A. U. & Dales, M. C. Mechanics of the healed meniscus in a canine model. Am. J. Sports Med. 17, 164–175 (1989).

    CAS  PubMed  Google Scholar 

  103. Russo, V. et al. Cellular and molecular maturation in fetal and adult ovine calcaneal tendons. J. Anat. 226, 126–142 (2015).

    CAS  PubMed  Google Scholar 

  104. Clark, C. R. & Ogden, J. A. Development of the menisci of the human knee joint. Morphological changes and their potential role in childhood meniscal injury. J. Bone Joint Surg. Am. 65, 538–547 (1983).

    CAS  PubMed  Google Scholar 

  105. Marturano, J. E., Arena, J. D., Schiller, Z. A., Georgakoudi, I. & Kuo, C. K. Characterization of mechanical and biochemical properties of developing embryonic tendon. Proc. Natl Acad. Sci. USA 110, 6370–6375 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Li, Q. et al. Impacts of maturation on the micromechanics of the meniscus extracellular matrix. J. Biomech. 72, 252–257 (2018).

    PubMed  PubMed Central  Google Scholar 

  107. Ionescu, L. C. et al. Maturation state-dependent alterations in meniscus integration: implications for scaffold design and tissue engineering. Tissue Eng. Part A 17, 193–204 (2011).

    CAS  PubMed  Google Scholar 

  108. Melrose, J., Smith, S., Cake, M., Read, R. & Whitelock, J. Comparative spatial and temporal localisation of perlecan, aggrecan and type I, II and IV collagen in the ovine meniscus: an ageing study. Histochem. Cell Biol. 124, 225–235 (2005).

    CAS  PubMed  Google Scholar 

  109. Di Giancamillo, A., Deponti, D., Addis, A., Domeneghini, C. & Peretti, G. M. Meniscus maturation in the swine model: changes occurring along with anterior to posterior and medial to lateral aspect during growth. J. Cell. Mol. Med. 18, 1964–1974 (2014).

    PubMed  PubMed Central  Google Scholar 

  110. Morales, T. I. Chondrocyte moves: clever strategies? Osteoarthr. Cartil. 15, 861–871 (2007).

    CAS  Google Scholar 

  111. Qu, F., Holloway, J. L., Esterhai, J. L., Burdick, J. A. & Mauck, R. L. Programmed biomolecule delivery to enable and direct cell migration for connective tissue repair. Nat. Commun. 8, 1780 (2017).

    PubMed  PubMed Central  Google Scholar 

  112. Qu, F. et al. Repair of dense connective tissues via biomaterial-mediated matrix reprogramming of the wound interface. Biomaterials 39, 85–94 (2015).

    CAS  PubMed  Google Scholar 

  113. Hunziker, E. B. & Kapfinger, E. Removal of proteoglycans from the surface of defects in articular cartilage transiently enhances coverage by repair cells. J. Bone Joint Surg. Br. 80, 144–150 (1998).

    CAS  PubMed  Google Scholar 

  114. Kim, M., Farrell, M. J., Steinberg, D. R., Burdick, J. A. & Mauck, R. L. Enhanced nutrient transport improves the depth-dependent properties of tri-layered engineered cartilage constructs with zonal co-culture of chondrocytes and MSCs. Acta Biomater. 58, 1–11 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Cheng, N. C., Estes, B. T., Young, T. H. & Guilak, F. Genipin-crosslinked cartilage-derived matrix as a scaffold for human adipose-derived stem cell chondrogenesis. Tissue Eng. Part A 19, 484–496 (2013).

    CAS  PubMed  Google Scholar 

  116. Baker, B. M. et al. The potential to improve cell infiltration in composite fiber-aligned electrospun scaffolds by the selective removal of sacrificial fibers. Biomaterials 29, 2348–2358 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Sundararaghavan, H. G. & Burdick, J. A. Gradients with depth in electrospun fibrous scaffolds for directed cell behavior. Biomacromolecules 12, 2344–2350 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Bhargava, M. M. et al. The effect of cytokines on the proliferation and migration of bovine meniscal cells. Am. J. Sports Med. 27, 636–643 (1999).

    CAS  PubMed  Google Scholar 

  119. Caliari, S. R. & Harley, B. A. The effect of anisotropic collagen-GAG scaffolds and growth factor supplementation on tendon cell recruitment, alignment, and metabolic activity. Biomaterials 32, 5330–5340 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Hannafin, J. A., Attia, E. T., Warren, R. F. & Bhargava, M. M. Characterization of chemotatic migration and growth kinetics of canine knee ligament fibroblasts. J. Orthop. Res. 17, 398–404 (1999).

    CAS  PubMed  Google Scholar 

  121. Kucia, M. et al. CXCR4–SDF-1 signalling, locomotion, chemotaxis and adhesion. J. Mol. Histol. 35, 233–245 (2004).

    CAS  PubMed  Google Scholar 

  122. Moore, K., Macsween, M. & Shoichet, M. Immobilized concentration gradients of neurotrophic factors guide neurite outgrowth of primary neurons in macroporous scaffolds. Tissue Eng. 12, 267–278 (2006).

    CAS  PubMed  Google Scholar 

  123. Wiles, K., Fishman, J. M., De Coppi, P. & Birchall, M. A. The host immune response to tissue-engineered organs: current problems and future directions. Tissue Eng. Part B Rev. 22, 208–219 (2016).

    PubMed  Google Scholar 

  124. Swartz, M. A., Hirosue, S. & Hubbell, J. A. Engineering approaches to immunotherapy. Sci. Transl Med. 4, 148rv149 (2012).

    Google Scholar 

  125. Stano, A., Scott, E. A., Dane, K. Y., Swartz, M. A. & Hubbell, J. A. Tunable T cell immunity towards a protein antigen using polymersomes versus solid-core nanoparticles. Biomaterials 34, 4339–4346 (2013).

    CAS  PubMed  Google Scholar 

  126. Nembrini, C. et al. Nanoparticle conjugation of antigen enhances cytotoxic T cell responses in pulmonary vaccination. Proc. Natl Acad. Sci. USA 108, E989–E997 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. McCarthy, D. P. et al. An antigen-encapsulating nanoparticle platform for TH1/17 immune tolerance therapy. Nanomedicine 13, 191–200 (2017).

    CAS  PubMed  Google Scholar 

  128. Spiller, K. L. et al. Sequential delivery of immunomodulatory cytokines to facilitate the M1-to-M2 transition of macrophages and enhance vascularization of bone scaffolds. Biomaterials 37, 194–207 (2015).

    CAS  PubMed  Google Scholar 

  129. Boehler, R. M. et al. Lentivirus delivery of IL-10 to promote and sustain macrophage polarization towards an anti-inflammatory phenotype. Biotechnol. Bioeng. 111, 1210–1221 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Sridharan, R. et al. Biomaterial based modulation of macrophage polarization: a review and suggested design principles. Mater. Today 18, 313–325 (2015).

    CAS  Google Scholar 

  131. Ratanavaraporn, J., Furuya, H. & Tabata, Y. Local suppression of pro-inflammatory cytokines and the effects in BMP-2-induced bone regeneration. Biomaterials 33, 304–316 (2012).

    CAS  PubMed  Google Scholar 

  132. Chen, W. C. et al. Controlled dual delivery of fibroblast growth factor-2 and Interleukin-10 by heparin-based coacervate synergistically enhances ischemic heart repair. Biomaterials 72, 138–151 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Geckil, H., Xu, F., Zhang, X., Moon, S. & Demirci, U. Engineering hydrogels as extracellular matrix mimics. Nanomedicine 5, 469–484 (2010).

    CAS  PubMed  Google Scholar 

  134. Kim, D.-H., Provenzano, P. P., Smith, C. L. & Levchenko, A. Matrix nanotopography as a regulator of cell function. J. Cell Biol. 197, 351–360 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Wade, R. J., Bassin, E. J., Gramlich, W. M. & Burdick, J. A. Nanofibrous hydrogels with spatially patterned biochemical signals to control cell behavior. Adv. Mater. 27, 1356–1362 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Kim, I. L., Khetan, S., Baker, B. M., Chen, C. S. & Burdick, J. A. Fibrous hyaluronic acid hydrogels that direct MSC chondrogenesis through mechanical and adhesive cues. Biomaterials 34, 5571–5580 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Billiet, T., Vandenhaute, M., Schelfhout, J., Van Vlierberghe, S. & Dubruel, P. A review of trends and limitations in hydrogel-rapid prototyping for tissue engineering. Biomaterials 33, 6020–6041 (2012).

    CAS  PubMed  Google Scholar 

  138. Bullough, P. G., Munuera, L., Murphy, J. & Weinstein, A. M. The strength of the menisci of the knee as it relates to their fine structure. J. Bone Joint Surg. Br. 52, 564–567 (1970).

    CAS  PubMed  Google Scholar 

  139. Proctor, C. S., Schmidt, M. B., Whipple, R. R., Kelly, M. A. & Mow, V. C. Material properties of the normal medial bovine meniscus. J. Orthop. Res. 7, 771–782 (1989).

    CAS  PubMed  Google Scholar 

  140. LeRoux, M. A. & Setton, L. A. Experimental and biphasic FEM determinations of the material properties and hydraulic permeability of the meniscus in tension. J. Biomech. Eng. 124, 315–321 (2002).

    PubMed  Google Scholar 

  141. Mauck, R. L. et al. Engineering on the straight and narrow: the mechanics of nanofibrous assemblies for fiber-reinforced tissue regeneration. Tissue Eng. Part B Rev. 15, 171–193 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Lim, J. Y. & Donahue, H. J. Cell sensing and response to micro-and nanostructured surfaces produced by chemical and topographic patterning. Tissue Eng. 13, 1879–1891 (2007).

    CAS  PubMed  Google Scholar 

  143. Gilchrist, C. L., Ruch, D. S., Little, D. & Guilak, F. Micro-scale and meso-scale architectural cues cooperate and compete to direct aligned tissue formation. Biomaterials 35, 10015–10024 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Baker, B. M. & Mauck, R. L. The effect of nanofiber alignment on the maturation of engineered meniscus constructs. Biomaterials 28, 1967–1977 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Orr, S. B. et al. Aligned multilayered electrospun scaffolds for rotator cuff tendon tissue engineering. Acta Biomater. 24, 117–126 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Ionescu, L. C. & Mauck, R. L. Porosity and cell preseeding influence electrospun scaffold maturation and meniscus integration in vitro. Tissue Eng. Part A 19, 538–547 (2013).

    CAS  PubMed  Google Scholar 

  147. Nam, J., Huang, Y., Agarwal, S. & Lannutti, J. Improved cellular infiltration in electrospun fiber via engineered porosity. Tissue Eng. 13, 2249–2257 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Simonet, M., Schneider, O. D., Neuenschwander, P. & Stark, W. J. Ultraporous 3D polymer meshes by low-temperature electrospinning: use of ice crystals as a removable void template. Polym. Eng. Sci. 47, 2020–2026 (2007).

    CAS  Google Scholar 

  149. Lee, B. L.-P. et al. Synovial stem cells and their responses to the porosity of microfibrous scaffold. Acta Biomater. 9, 7264–7275 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Phipps, M. C., Clem, W. C., Grunda, J. M., Clines, G. A. & Bellis, S. L. Increasing the pore sizes of bone-mimetic electrospun scaffolds comprised of polycaprolactone, collagen I and hydroxyapatite to enhance cell infiltration. Biomaterials 33, 524–534 (2012).

    CAS  PubMed  Google Scholar 

  151. Baker, B. M. et al. Sacrificial nanofibrous composites provide instruction without impediment and enable functional tissue formation. Proc. Natl Acad. Sci. USA 109, 14176–14181 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Cai, S., Xu, H., Jiang, Q. & Yang, Y. Novel 3D electrospun scaffolds with fibers oriented randomly and evenly in three dimensions to closely mimic the unique architectures of extracellular matrices in soft tissues: fabrication and mechanism study. Langmuir 29, 2311–2318 (2013).

    CAS  PubMed  Google Scholar 

  153. Moutos, F. T., Freed, L. E. & Guilak, F. A biomimetic three-dimensional woven composite scaffold for functional tissue engineering of cartilage. Nat. Mater. 6, 162–167 (2007).

    CAS  PubMed  Google Scholar 

  154. Lee, J., Jang, J., Oh, H., Jeong, Y. H. & Cho, D.-W. Fabrication of a three-dimensional nanofibrous scaffold with lattice pores using direct-write electrospinning. Mater. Lett. 93, 397–400 (2013).

    CAS  Google Scholar 

  155. Daly, A. C. et al. 3D bioprinting for cartilage and osteochondral tissue engineering. Adv. Healthc. Mater. 6, 1700298 (2017).

    Google Scholar 

  156. Miller, J. S. et al. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nat. Mater. 11, 768–774 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Freeman, F. E. & Kelly, D. J. Tuning alginate bioink stiffness and composition for controlled growth factor delivery and to spatially direct MSC fate within bioprinted tissues. Sci. Rep. 7, 17042 (2017).

    PubMed  PubMed Central  Google Scholar 

  158. Skardal, A. et al. A hydrogel bioink toolkit for mimicking native tissue biochemical and mechanical properties in bioprinted tissue constructs. Acta Biomater. 25, 24–34 (2015).

    CAS  PubMed  Google Scholar 

  159. Albritton, J. L. & Miller, J. S. 3D bioprinting: improving in vitro models of metastasis with heterogeneous tumor microenvironments. Dis. Model. Mech. 10, 3–14 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Miller, J. S. et al. Bioactive hydrogels made from step-growth derived PEG–peptide macromers. Biomaterials 31, 3736–3743 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Ehrbar, M. et al. Elucidating the role of matrix stiffness in 3D cell migration and remodeling. Biophys. J. 100, 284–293 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Bott, K. et al. The effect of matrix characteristics on fibroblast proliferation in 3D gels. Biomaterials 31, 8454–8464 (2010).

    CAS  PubMed  Google Scholar 

  163. Wade, R. J., Bassin, E. J., Rodell, C. B. & Burdick, J. A. Protease-degradable electrospun fibrous hydrogels. Nat. Commun. 6, 6639 (2015).

    CAS  PubMed  Google Scholar 

  164. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Lee, S. H., Miller, J. S., Moon, J. J. & West, J. L. Proteolytically degradable hydrogels with a fluorogenic substrate for studies of cellular proteolytic activity and migration. Biotechnol. Prog. 21, 1736–1741 (2005).

    CAS  PubMed  Google Scholar 

  166. Feng, Q., Zhu, M., Wei, K. & Bian, L. Cell-mediated degradation regulates human mesenchymal stem cell chondrogenesis and hypertrophy in MMP-sensitive hyaluronic acid hydrogels. PLOS ONE 9, e99587 (2014).

    PubMed  PubMed Central  Google Scholar 

  167. Salinas, C. N. & Anseth, K. S. The enhancement of chondrogenic differentiation of human mesenchymal stem cells by enzymatically regulated RGD functionalities. Biomaterials 29, 2370–2377 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Chaudhuri, O. et al. Substrate stress relaxation regulates cell spreading. Nat. Commun. 6, 6364 (2015).

    PubMed  Google Scholar 

  169. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Brunger, J. M., Zutshi, A., Willard, V. P., Gersbach, C. A. & Guilak, F. CRISPR/Cas9 editing of murine induced pluripotent stem cells for engineering inflammation-resistant tissues. Arthritis Rheumatol. 69, 1111–1121 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Brunger, J. M., Zutshi, A., Willard, V. P., Gersbach, C. A. & Guilak, F. Genome engineering of stem cells for autonomously regulated, closed-loop delivery of biologic drugs. Stem Cell Rep. 8, 1202–1213 (2017).

    CAS  Google Scholar 

  172. Brunger, J. M. et al. Scaffold-mediated lentiviral transduction for functional tissue engineering of cartilage. Proc. Natl Acad. Sci. USA 111, E798–806 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Glass, K. A. et al. Tissue-engineered cartilage with inducible and tunable immunomodulatory properties. Biomaterials 35, 5921–5931 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The work of F.Q., F.G. and R.L.M. is supported by the US National Institutes of Health (AR060719, AR056624, EB008722, AR50245, AR48852, AG46927, AG15768, AR48182, AR067467 and AR057235), the US Department of Veterans’ Affairs (I01 RX000174), the Arthritis Foundation, the Nancy Taylor Foundation for Chronic Diseases and the Collaborative Research Center of the AO Foundation in Davos.

Reviewer information

Nature Reviews Rheumatology thanks D. Grande and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Farshid Guilak or Robert L. Mauck.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Glossary

Stress fibre

Contractile bundles in non-muscle cells composed of actin filaments and non-muscle myosin II; myosin motor activity results in contraction of the actomyosin bundles.

Micromechanics

The mechanical properties of a material assessed at a local level (that is, at the micrometre scale). This approach can identify heterogeneities in materials or tissues that are indicative of the constituent materials and their properties at that location.

Microstructure

The microscopic structure of a material or tissue.

Chemotaxis

Directional cell movement along a soluble biochemical gradient.

Haptotaxis

Directional cell movement along a substrate-bound insoluble gradient.

Collective migration

The process by which a group of cells move together while maintaining cell–cell contact.

Tensile modulus

Young’s modulus of a material evaluated in tension (that is, a measurement of tensile strength, which is the ability of a material to withstand being stretched).

Compressive modulus

Young’s modulus of a material evaluated in compression (that is, a measurement of compressive strength, which is the ability of a material to withstand being compressed).

Shear modulus

Young’s modulus of a material evaluated in shear (that is, a measurement of the shear strength, which is the ability of a material to withstand forces that can cause the internal structure of the material to slide against itself).

Young’s modulus

A mechanical property that defines the relationship between stress (force per unit area) and strain (proportional deformation) of a linearly elastic material during uniaxial deformation (also referred to as the elastic modulus; measured in MPa). Although commonly referred to as tissue stiffness or rigidity, these two terms are actually structural properties (that is, dependent on the size and shape of the tissue) and are not inherent material properties.

Contact guidance

The response of cells to topographic cues; the direction of cell alignment and migration is affected by geometrical patterns such as grooves or fibres.

Pannus

An abnormal layer of fibrovascular tissue, which can occur in rheumatoid arthritis.

Heterotopic ossification

The presence of bone in soft tissue where bone does not normally exist.

Scar tissue

Dense fibrous tissue that replaces original tissue during wound healing; the scar tissue is generally disordered and does not match the original tissue in terms of the biochemical content or mechanical properties.

Adhesion

Abnormal formation of scar tissue after injury that connects normally separated tissues and impedes joint motion.

Micromoulding

A technique for generating microstructures using moulds with micrometre-scale features.

Photolithography

A technique that uses light to transfer geometric patterns from a photomask (an opaque plate that enables light to shine through in a defined pattern) to a light-sensitive chemical on a substrate.

Electrospinning

A technique that produces nanofibres by charging and pulling a polymer solution through a spinneret under a high-voltage electrical field.

Rapid prototyping

A group of techniques for constructing a 3D product using computer-aided design (for example, 3D printing).

Anisotropic

Describes mechanical and physical properties that vary on the basis of the testing direction.

Porogens

Space-filling particles used to create porous materials, which are later removed to generate voids.

Bioinks

Extrudable polymer solutions used in 3D bioprinting that can contain cells and/or other biologics that can solidify after printing.

Stress relaxation

A time-dependent decrease in stress of a material in response to the same level of strain.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qu, F., Guilak, F. & Mauck, R.L. Cell migration: implications for repair and regeneration in joint disease. Nat Rev Rheumatol 15, 167–179 (2019). https://doi.org/10.1038/s41584-018-0151-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41584-018-0151-0

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