Volume 53 Issue 1
Mar.  2023
Turn off MathJax
Article Contents
Zhang H, Zhao G Q, Feng J T, Lin M. Cellular mechanobiology: Mediated by force-sensitive adhesion receptors. Advances in Mechanics, 2023, 53(1): 48-153 doi: 10.6052/1000-0992-22-029
Citation: Zhang H, Zhao G Q, Feng J T, Lin M. Cellular mechanobiology: Mediated by force-sensitive adhesion receptors. Advances in Mechanics, 2023, 53(1): 48-153 doi: 10.6052/1000-0992-22-029

Cellular mechanobiology: Mediated by force-sensitive adhesion receptors

doi: 10.6052/1000-0992-22-029
More Information
  • Corresponding author: minlin@xjtu.edu.cn
  • Received Date: 2022-06-10
  • Accepted Date: 2022-08-15
  • Available Online: 2022-08-16
  • Publish Date: 2023-03-25
  • As the interface between cells and their external environment for materials and energy exchange, the cell membrane is an important structure that regulates cellular activities. Representative transmembrane force-sensitive receptors, such as integrins and cadherins, are found to play key roles in mediating cellular interactions with the ECM or adjacent cells. These interactions will then transduce mechanical stimuli into biochemical signals, which in turn activate a series of intracellular signaling cascade, and ultimately affect cell growth, differentiation, proliferation, migration and apoptosis etc. The investigation of cellular mechanobiology regulated by force-sensitive adhesion receptors has thus become the key to explore the mechanobiological mechanisms of cellular actively in response to complex mechanical microenvironments. This provides valuable theoretical and experimental basis for further understanding of the changes in cell functions under physiological and pathological conditions, as well as for revealing the mechanism of disease development. This review summarizes the cutting-edge progresses in cellular mechanobiology regulated force-sensitive adhesion receptors. This review begins by introducing the structure and function of force-sensitive receptors at the adhesion interface, and followed by elaborating systematic mathematical models of how cells sense and respond to mechanical signals mediated by these receptors. It also outlines the processes of mechanical signal transduction through force-sensitive receptors, and the mechanobiological mechanism of adhesion-mediated changes in cell functions. In addition, the techniques for constructing of in vitro mechanical microenvironment that mimic cell-ECM (via integrin ligation) and cell-cell (via cadherin ligation) interactions are described. Finally, we identify the future directions of mechanobiology in terms of force-sensitive receptors regulated cell functions.

     

  • loading
  • [1]
    程波, 徐峰. 2021. 考虑细胞外基质黏弹性行为的细胞黏附力学模型. 应用数学和力学, 42: 1074-1080 (Cheng B, Xu F. 2021. A molecular clutch model of cellular adhesion on viscoelastic substrate. Applied Mathematics and Mechanics, 42: 1074-1080 (in Chinese)).

    Cheng B, Xu F. A molecular clutch model of cellular adhesion on viscoelastic substrate Applied Mathematics and Mechanics, 2021, 42: 1074-1080 (in Chinese)
    [2]
    姜宗来. 2017. 从生物力学到力学生物学的进展. 力学进展, 47: 309-332 (Jiang Z L. 2017. Advance from biomechanics to mechanobiology. Advances in Mechanics, 47: 309-332 (in Chinese)). doi: 10.6052/1000-0992-16-023

    (Z l JIANG. 2017. Advance from biomechanics to mechanobiology. Advances in Mechanics, 47: 309-332 (in Chinese)). doi: 10.6052/1000-0992-16-023
    [3]
    姜宗来. 2021. 发展生物力学 造福人类健康——“十四五”我国生物力学研究发展战略思考. 医用生物力学, 36: 671-675

    Jiang Z l. 2021. Develop biomechanics, benefit human health: pondering on the development strategy of biomechanical researches during the 14th five-year plan in china . Journal of Medical Biomechanics, 36: 671-675 (in Chinese)
    [4]
    李洋, 洪莉. 2019. 整合素与细胞骨架生物学关系研究进展. 医学综述. 25: 44-48

    Li Y, Hong L. 2019. Research progress of biological relationship between integrin and cytoskeleton. Medical Recapitulate, 25: 44-48 (in Chinese)
    [5]
    刘璐, 张云霞, 董欣, 刘虎, 曾强成. 2014. 力学环境下成纤维细胞生物学响应研究. 德州学院学报, 30: 23-29

    Liu L, Zhang Y X, Dong X, Liu H, Zeng Q C. 2014. Research of biological response of fibroblast under the mechanical environment. Journal of Dezhou University, 30: 23-29 (in Chinese)
    [6]
    吕东媛, 周吕文, 龙勉. 2017. 干细胞的生物力学研究. 力学进展, 47: 534-585

    Lü D Y, Zhou L W, Long M. 2017. Biomechanics of stem cells. Advances in Mechanics, 47: 534-585 (in Chinese)
    [7]
    齐颖新. 2022. 细胞核与应力信号转导. 医用生物力学, 37: 385-388

    Qi Y X. 2022. Nucleus in mechanotransduction. Journal of Medical Biomechanics, 37: 385-388 (in Chinese)
    [8]
    施兴华, 张路姚, 李博, 冯西桥. 2018. 肿瘤及其微环境的力学问题. 力学进展, 48: 360-409

    Shi X H, Zhang L Y, Li B, Feng X Q. 2018. The mechanical problems in tumor and tumor microenvironment. Advances in Mechanics, 48: 360-409 (in Chinese)
    [9]
    徐峰, 张晓慧, 鲍雪娇, 赵国旭, 刘付生等. 2018. 基于先进生物材料的心肌细胞力–电微环境体外构建. 力学进展, 48: 320-359

    Xu F, Zheng X H, Bao X J, Zhao G X, Liu F S, et al. 2018. Engineering mechanical-electrical cell microenvironment in myocardium using advanced biomaterials. Advances in Mechanics, 48: 320-359 (in Chinese)
    [10]
    Adhikari S, Moran J, Weddle C, Hinczewski M. 2018. Unraveling the mechanism of the cadherin-catenin-actin catch bond. PLoS Computational Biology, 14: e1006399. doi: 10.1371/journal.pcbi.1006399
    [11]
    Alimperti S, Andreadis S T. 2015. CDH2 and CDH11 act as regulators of stem cell fate decisions. Stem Cell Research, 14: 270-282. doi: 10.1016/j.scr.2015.02.002
    [12]
    Alimperti S, You H, George T, Agarwal S K, Andreadis S T. 2014. Cadherin-11 regulates both mesenchymal stem cell differentiation into smooth muscle cells and the development of contractile function in vivo. Journal of Cell Science, 127: 2627-38.
    [13]
    Alisafaei F, Jokhun D S, Shivashankar G V, Shenoy V B. 2019. Regulation of nuclear architecture, mechanics, and nucleocytoplasmic shuttling of epigenetic factors by cell geometric constraints. Proceedings of the national academy of sciences of the united states of america, 116: 13200-13209. doi: 10.1073/pnas.1902035116
    [14]
    Angulo-Urarte A, van der Wal T, Huveneers S. 2020. Cell-cell junctions as sensors and transducers of mechanical forces. Biochimica et Biophysica Acta:Biomembranes, 1862: 183316.
    [15]
    Aragona M, Panciera T, Manfrin A, Giulitti S, Michielin F, et al. 2013. A mechanical checkpoint controls multicellular growth through yap/taz regulation by actin-processing factors. Cell, 154: 1047-1059. doi: 10.1016/j.cell.2013.07.042
    [16]
    Aragona M, Sifrim A, Malfait M, Song Y, Van Herck J, et al. 2020. Mechanisms of stretch-mediated skin expansion at single-cell resolution. Nature, 584: 268-273. doi: 10.1038/s41586-020-2555-7
    [17]
    Aratyn-Schaus Y, Gardel M L. 2010. Transient frictional slip between integrin and the ECM in focal adhesions under myosin II tension. Current Biology, 20: 1145-53. doi: 10.1016/j.cub.2010.05.049
    [18]
    Ashurst W T, Hoover W G. 1976. Microscopic fracture studies in the two-dimensional triangular lattice. Physical Review B, 14: 1465-1473. doi: 10.1103/PhysRevB.14.1465
    [19]
    Austen K, Ringer P, Mehlich A, Chrostek-Grashoff A, Kluger C, et al. 2015. Extracellular rigidity sensing by talin isoform-specific mechanical linkages. Nature Cell Biology, 17: 1597-1606. doi: 10.1038/ncb3268
    [20]
    Bachir A I, Horwitz A R, Nelson W J, Bianchini J M. 2017. Actin-based adhesion modules mediate cell interactions with the extracellular matrix and neighboring cells. Cold Spring Harbor perspectives in biology, 9: a023234.
    [21]
    Bajpai A, Tong J, Qian W, Peng Y, Chen W. 2019. The interplay between cell-cell and cell-matrix forces regulates cell migration dynamics. Biophysical Journal, 117: 1795-1804. doi: 10.1016/j.bpj.2019.10.015
    [22]
    Balestrini J L, Chaudhry S, Sarrazy V, Koehler A, Hinz B. 2012. The mechanical memory of lung myofibroblasts. Integrative Biology, 4: 410-421. doi: 10.1039/c2ib00149g
    [23]
    Bangasser B L, Odde D J. 2013. Master equation-based analysis of a motor-clutch model for cell traction force. Cellular and Molecular Bioengineering, 6: 449-459. doi: 10.1007/s12195-013-0296-5
    [24]
    Bangasser B L, Shamsan G A, Chan C E, Opoku K N, Tüzel E, et al. 2017. Shifting the optimal stiffness for cell migration. Nature Communications, 8: 15313. doi: 10.1038/ncomms15313
    [25]
    Bangasser B L, Rosenfeld S S, Odde D J. 2013. Determinants of maximal force transmission in a motor-clutch model of cell traction in a compliant microenvironment. Biophysical Journal, 105: 581-592. doi: 10.1016/j.bpj.2013.06.027
    [26]
    Bao M, Xie J, Huck Wilhelm T S. 2018. Recent advances in engineering the stem cell microniche in 3D. Advanced Science, 5: 1800448. doi: 10.1002/advs.201800448
    [27]
    Barbazan J, Matic Vignjevic D. 2019. Cancer associated fibroblasts: is the force the path to the dark side? Current Opinion in Cell Biology, 56: 71-79.
    [28]
    Barcelona-Estaje E, Dalby M J, Cantini M, Salmeron-Sanchez M. 2021. You talking to me? Cadherin and integrin crosstalk in biomaterial design. Advanced Healthcare Materials, 10: 2002048. doi: 10.1002/adhm.202002048
    [29]
    Barthes J, Özçelik H, Hindié M, Ndreu-Halili A, Hasan A, Vrana N E. 2014. Cell microenvironment engineering and monitoring for tissue engineering and regenerative medicine: the recent advances. BioMed Research International, 2014: 921905.
    [30]
    Bauer J, Emon M A B, Staudacher J J, Thomas A L, Zessner-Spitzenberg J, et al. 2020. Increased stiffness of the tumor microenvironment in colon cancer stimulates cancer associated fibroblast-mediated prometastatic activin A signaling. Scientific Reports, 10: 50-50. doi: 10.1038/s41598-019-55687-6
    [31]
    Bazellières E, Conte V, Elosegui-Artola A, Serra-Picamal X, Bintanel-Morcillo M, et al. 2015. Control of cell-cell forces and collective cell dynamics by the intercellular adhesome. Nature Cell Biology, 17: 409-420. doi: 10.1038/ncb3135
    [32]
    Belardi B, Son S, Felce J H, Dustin M L, Fletcher D A. 2020. Cell–cell interfaces as specialized compartments directing cell function. Nature Reviews Molecular Cell Biology, 21: 750-764. doi: 10.1038/s41580-020-00298-7
    [33]
    Bell G I, Dembo M, Bongrand P. 1984. Cell adhesion. Competition between nonspecific repulsion and specific bonding. Biophysical Journal, 45: 1051-64. doi: 10.1016/S0006-3495(84)84252-6
    [34]
    Bell G I. 1978. Models for the specific adhesion of cells to cells. Science, 200: 618-27. doi: 10.1126/science.347575
    [35]
    Bennett M, Cantini M, Reboud J, Cooper J M, Roca-Cusachs P, Salmeron-Sanchez M. 2018. Molecular clutch drives cell response to surface viscosity. Proceedings of the national academy of sciences of the united states of america, 115: 1192-1197. doi: 10.1073/pnas.1710653115
    [36]
    Bertocchi C, Wang Y, Ravasio A, Hara Y, Wu Y, et al. 2017. Nanoscale architecture of cadherin-based cell adhesions. Nature Cell Biology, 19: 28-37. doi: 10.1038/ncb3456
    [37]
    Besser A, Safran S A. 2006. Force-induced adsorption and anisotropic growth of focal adhesions. Biophysical Journal, 90: 3469-84. doi: 10.1529/biophysj.105.074377
    [38]
    Besser A, Schwarz U S. 2007. Coupling biochemistry and mechanics in cell adhesion: a model for inhomogeneous stress fiber contraction. New Journal of Physics, 9: 425-425. doi: 10.1088/1367-2630/9/11/425
    [39]
    Bian L, Guvendiren M, Mauck R L, Burdick J A. 2013. Hydrogels that mimic developmentally relevant matrix and N-cadherin interactions enhance MSC chondrogenesis. Proceedings of the national academy of sciences of the united states of america, 110: 10117-10122. doi: 10.1073/pnas.1214100110
    [40]
    Biggs M J P, Fernandez M, Thomas D, Cooper R, Palma M, et al. 2017. The functional response of mesenchymal stem cells to electron-beam patterned elastomeric surfaces presenting micrometer to nanoscale heterogeneous rigidity. Advanced Materials, 29: 1702119. doi: 10.1002/adma.201702119
    [41]
    Blaschuk O W. 2015. N-cadherin antagonists as oncology therapeutics. Philosophical Transactions of the Royal Society B:Biological Sciences, 370: 20140039. doi: 10.1098/rstb.2014.0039
    [42]
    Boggon T J, Murray J, Chappuis-Flament S, Wong E, Gumbiner B M, Shapiro L. 2002. C-cadherin ectodomain structure and implications for cell adhesion mechanisms. Science, 296: 1308-13. doi: 10.1126/science.1071559
    [43]
    Borghi N, Sorokina M, Shcherbakova O G, Weis W I, Pruitt B L, et al. 2012. E-cadherin is under constitutive actomyosin-generated tension that is increased at cell-cell contacts upon externally applied stretch. Proceedings of the national academy of sciences of the united states of america, 109: 12568-73. doi: 10.1073/pnas.1204390109
    [44]
    Boscher C, Zheng Y Z, Lakshminarayan R, Johannes L, Dennis J W, et al. 2012. Galectin-3 protein regulates mobility of N-cadherin and GM1 ganglioside at cell-cell junctions of mammary carcinoma cells. Journal of Biological Chemistry, 287: 32940-52. doi: 10.1074/jbc.M112.353334
    [45]
    Brasch J, Harrison O J, Honig B, Shapiro L. 2012. Thinking outside the cell: how cadherins drive adhesion. Trends in Cell Biology, 22: 299-310. doi: 10.1016/j.tcb.2012.03.004
    [46]
    Breckenridge M T, Desai R A, Yang M T, Fu J, Chen C S. 2014. Substrates with engineered step changes in rigidity induce traction force polarity and durotaxis. Cellular and Molecular Bioengineering, 7: 26-34. doi: 10.1007/s12195-013-0307-6
    [47]
    Bretscher M S. 2008. On the shape of migrating cells — a `front-to-back' model. Journal of Cell Science, 121: 2625-2628. doi: 10.1242/jcs.031120
    [48]
    Buckley C D, Tan J, Anderson K L, Hanein D, Volkmann N, et al. 2014. Cell adhesion. The minimal cadherin-catenin complex binds to actin filaments under force. Science, 346: 1254211. doi: 10.1126/science.1254211
    [49]
    Camper L, Hellman U, Lundgren-Akerlund E. 1998. Isolation, cloning, and sequence analysis of the integrin subunit alpha10, a beta1-associated collagen binding integrin expressed on chondrocytes. Journal of Biological Chemistry, 273: 20383. doi: 10.1074/jbc.273.32.20383
    [50]
    Canel M, Serrels A, Frame M C, Brunton V G. 2013. E-cadherin-integrin crosstalk in cancer invasion and metastasis. Journal of Cell Science, 126: 393-401. doi: 10.1242/jcs.100115
    [51]
    Cao X, Lin Y, Driscoll T P, Franco-Barraza J, Cukierman E, et al. 2015. A chemomechanical model of matrix and nuclear rigidity regulation of focal adhesion size. Biophysical Journal, 109: 1807-17. doi: 10.1016/j.bpj.2015.08.048
    [52]
    Caputo K E, Hammer D A. 2005. Effect of microvillus deformability on leukocyte adhesion explored using adhesive dynamics simulations. Biophysical Journal, 89: 187-200. doi: 10.1529/biophysj.104.054171
    [53]
    Case L B, Waterman C M. 2015. Integration of actin dynamics and cell adhesion by a three-dimensional, mechanosensitive molecular clutch. Nature Cell Biology, 17: 955-963. doi: 10.1038/ncb3191
    [54]
    Caswell P T, Norman J C. 2006. Integrin trafficking and the control of cell migration. Traffic, 7: 14-21. doi: 10.1111/j.1600-0854.2005.00362.x
    [55]
    Cavalcanti-Adam E A, Micoulet A, Blummel J, Auernheimer J, Kessler H, Spatz J P. 2006. Lateral spacing of integrin ligands influences cell spreading and focal adhesion assembly. European Journal of Cell Biology, 85: 219-24. doi: 10.1016/j.ejcb.2005.09.011
    [56]
    Cavallaro U, Christofori G. 2004. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nature Reviews Cancer, 4: 118-32. doi: 10.1038/nrc1276
    [57]
    Chan C E, Odde D J. 2008. Traction dynamics of filopodia on compliant substrates. Science, 322: 1687-1691. doi: 10.1126/science.1163595
    [58]
    Chan W-L, Silberstein J, Hai C-M. 2000. Mechanical strain memory in airway smooth muscle. American Journal of Physiology-Cell Physiology, 278: C895-C904. doi: 10.1152/ajpcell.2000.278.5.C895
    [59]
    Chandra A, Butler M T, Bear J E, Haugh J M. 2022. Modeling cell protrusion predicts how myosin II and actin turnover affect adhesion-based signaling. Biophysical Journal, 121: 102-118. doi: 10.1016/j.bpj.2021.11.2889
    [60]
    Chang B, Svoboda K K H, Liu X. 2019. Cell polarization: from epithelial cells to odontoblasts. European Journal of Cell Biology, 98: 1-11. doi: 10.1016/j.ejcb.2018.11.003
    [61]
    Changede R, Sheetz M. 2017. Integrin and cadherin clusters: A robust way to organize adhesions for cell mechanics. Bioessays, 39: 1-12. doi: 10.1002/bies.201670013
    [62]
    Changede R, Xu X, Margadant F, Sheetz M P. 2015. Nascent integrin adhesions form on all matrix rigidities after integrin activation. Developmental Cell, 35: 614-621. doi: 10.1016/j.devcel.2015.11.001
    [63]
    Charras G, Yap A S. 2018. Tensile forces and mechanotransduction at cell-cell junctions. Current Biology, 28: R445-R457. doi: 10.1016/j.cub.2018.02.003
    [64]
    Chaudhuri O, Cooper-White J, Janmey P A, Mooney D J, Shenoy V B. 2020. Effects of extracellular matrix viscoelasticity on cellular behaviour. Nature, 584: 535-546. doi: 10.1038/s41586-020-2612-2
    [65]
    Chaudhuri O, Gu L, Darnell M, Klumpers D, Bencherif S A, et al. 2015a. Substrate stress relaxation regulates cell spreading. Nature Communications, 6: 6364. doi: 10.1038/ncomms7364
    [66]
    Chaudhuri O, Gu L, Klumpers D, Darnell M, Bencherif S A, et al. 2015b. Hydrogels with tunable stress relaxation regulate stem cell fate and activity. Nature Materials, 15: 326-334.
    [67]
    Chaudhuri P K, Low B C, Lim C T. 2018. Mechanobiology of tumor growth. Chemical Reviews, 118: 6499-6515. doi: 10.1021/acs.chemrev.8b00042
    [68]
    Chen B, Ji B, Gao H. 2015. Modeling active mechanosensing in cell-matrix interactions. Annual Review of Biophysics, 44: 1-32. doi: 10.1146/annurev-biophys-051013-023102
    [69]
    Chen C S. 2008. Mechanotransduction - a field pulling together? Journal of Cell Science, 121: 3285-92.
    [70]
    Chen D, Hu S, Liu J, Li S. 2019. E-cadherin regulates biological behaviors of neural stem cells and promotes motor function recovery following spinal cord injury. Experimental and Therapeutic Medicine, 17: 2061-2070.
    [71]
    Chen J, Newhall J, Xie Z R, Leckband D, Wu Y. 2016. A computational model for kinetic studies of cadherin binding and clustering. Biophysical Journal, 111: 1507-1518. doi: 10.1016/j.bpj.2016.08.038
    [72]
    Chen J, Xie Z R, Wu Y. 2014. Computational modeling of the interplay between cadherin-mediated cell adhesion and Wnt signaling pathway. PLoS One, 9: e100702. doi: 10.1371/journal.pone.0100702
    [73]
    Chen T, Yuan D, Wei B, Jiang J, Kang J, et al. 2010. E-cadherin-mediated cell-cell contact is critical for induced pluripotent stem cell generation. Stem Cells, 28: 1315-25. doi: 10.1002/stem.456
    [74]
    Chen Y, Brasch J, Harrison O J, Bidone T C. 2021. Computational model of E-cadherin clustering under force. Biophysical Journal, 120: 4944-4954. doi: 10.1016/j.bpj.2021.10.018
    [75]
    Cheng B, Lin M, Huang G, Li Y, Ji B, et al. 2017. Cellular mechanosensing of the biophysical microenvironment: A review of mathematical models of biophysical regulation of cell responses. Physics of Life Reviews, 22-23: 88-119. doi: 10.1016/j.plrev.2017.06.016
    [76]
    Cheng B, Lin M, Li Y, Huang G, Yang H, et al. 2016. An integrated stochastic model of matrix-stiffness-dependent filopodial dynamics. Biophysical Journal, 111: 2051-2061. doi: 10.1016/j.bpj.2016.09.026
    [77]
    Cheng B, Wan W T, Huang G, Li Y, Genin G M, et al. 2020. Nanoscale integrin cluster dynamics controls cellular mechanosensing via FAKY397 phosphorylation. Science Advances, 6: eaax1909. doi: 10.1126/sciadv.aax1909
    [78]
    Choi C K, Xu Y J, Wang B, Zhu M, Zhang L, Bian L. 2015. Substrate coupling strength of integrin-binding ligands modulates adhesion, spreading, and differentiation of human mesenchymal stem cells. Nano Letters, 15: 6592-600. doi: 10.1021/acs.nanolett.5b02323
    [79]
    Choquet D, Felsenfeld D P, Sheetz M P. 1997. Extracellular matrix rigidity causes strengthening of integrin–cytoskeleton linkages. Cell, 88: 39-48. doi: 10.1016/S0092-8674(00)81856-5
    [80]
    Chrzanowska-Wodnicka M, Burridge K. 1996. Rho-stimulated contractility drives the formation of stress fibers and focal adhesions. Journal of Cell Biology, 133: 1403-15. doi: 10.1083/jcb.133.6.1403
    [81]
    Cirit M, Krajcovic M, Choi C K, Welf E S, Horwitz A F, Haugh J M. 2010. Stochastic model of integrin-mediated signaling and adhesion dynamics at the leading edges of migrating cells. PLoS Computational Biology, 6: e1000688. doi: 10.1371/journal.pcbi.1000688
    [82]
    Clark K, Langeslag M, Figdor C G, van Leeuwen F N. 2007. Myosin II and mechanotransduction: a balancing act. Trends in Cell Biology, 17: 178-86. doi: 10.1016/j.tcb.2007.02.002
    [83]
    Clevers H, Nusse R. 2012. Wnt/β-catenin signaling and disease. Cell, 149: 1192-205. doi: 10.1016/j.cell.2012.05.012
    [84]
    Cohen D J, Nelson W J. 2018. Secret handshakes: cell–cell interactions and cellular mimics. Current Opinion in Cell Biology, 50: 14-19. doi: 10.1016/j.ceb.2018.01.001
    [85]
    Cohen M, Klein E, Geiger B, Addadi L. 2003. Organization and adhesive properties of the hyaluronan pericellular coat of chondrocytes and epithelial cells. Biophysical Journal, 85: 1996-2005. doi: 10.1016/S0006-3495(03)74627-X
    [86]
    Collins C, Denisin A K, Pruitt B L, Nelson W J. 2017. Changes in E-cadherin rigidity sensing regulate cell adhesion. Proceedings of the National Academy of Sciences, 114: E5835.
    [87]
    Constantina B, Cohen D M, Bankston L A, Bobkov A A, Cadwell G W, et al. 2004. Structural basis for vinculin activation at sites of cell adhesion. Nature, 430: 583. doi: 10.1038/nature02610
    [88]
    Copos C A, Walcott S, Del Álamo J C, Bastounis E, Mogilner A, Guy R D. 2017. Mechanosensitive adhesion explains stepping motility in amoeboid cells. Biophysical Journal, 112: 2672-2682. doi: 10.1016/j.bpj.2017.04.033
    [89]
    Corallino S, Malabarba M G, Zobel M, Di Fiore P P, Scita G. 2015. Epithelial-to-mesenchymal plasticity harnesses endocytic circuitries. Frontiers in Oncology, 5: 45.
    [90]
    Cosgrove B D, Mui K L, Driscoll T P, Caliari S R, Mehta K D, et al. 2016. N-cadherin adhesive interactions modulate matrix mechanosensing and fate commitment of mesenchymal stem cells. Nature Materials, 15: 1297-1306. doi: 10.1038/nmat4725
    [91]
    Craig E M, Stricker J, Gardel M, Mogilner A. 2015. Model for adhesion clutch explains biphasic relationship between actin flow and traction at the cell leading edge. Physical Biology, 12: 035002. doi: 10.1088/1478-3975/12/3/035002
    [92]
    Dalby M J, Gadegaard N, Oreffo R O. 2014. Harnessing nanotopography and integrin-matrix interactions to influence stem cell fate. Nature Materials, 13: 558-69. doi: 10.1038/nmat3980
    [93]
    D'Arcangelo E, Wu N C, Cadavid J L, McGuigan A P. 2020. The life cycle of cancer-associated fibroblasts within the tumour stroma and its importance in disease outcome. British Journal of Cancer, 122: 931-942. doi: 10.1038/s41416-019-0705-1
    [94]
    Das R K, Gocheva V, Hammink R, Zouani O F, Rowan A E. 2015. Stress-stiffening-mediated Stem-Cell Commitment Switch in Soft Responsive Hydrogels. Nature Materials, 15: 318.
    [95]
    de Beco S, Amblard F, Coscoy S. 2012. New insights into the regulation of e-cadherin distribution by endocytosis. International Review of Cell and Molecular Biology, 295: 63-108.
    [96]
    De R, Zemel A, Safran S A. 2010. Theoretical concepts and models of cellular mechanosensing. Methods in Cell Biology, 98: 143-175.
    [97]
    de Rooij J. 2014. Cadherin adhesion controlled by cortical actin dynamics. Nature Cell Biology, 16: 508-510. doi: 10.1038/ncb2980
    [98]
    Dembo M, Torney D C, Saxman K, Hammer D. 1988. The reaction-limited kinetics of membrane-to-surface adhesion and detachment. Proceedings of the Royal Society B:Biological Sciences, 234: 55-83.
    [99]
    Deng Z, Yan W, Dai X, Chen M, Qu Q, et al. 2021. N-cadherin regulates the odontogenic differentiation of dental pulp stem cells via β-catenin activity. Frontiers in Cell and Developmental Biology, 9: 661116. doi: 10.3389/fcell.2021.661116
    [100]
    Dingal P C D P, Bradshaw A M, Cho S, Raab M, Buxboim A, et al. 2015. Fractal heterogeneity in minimal matrix models of scars modulates stiff-niche stem-cell responses via nuclear exit of a mechanorepressor. Nature Materials, 14: 951-960. doi: 10.1038/nmat4350
    [101]
    Douezan S, Dumond J, Brochard-Wyart F. 2012. Wetting transitions of cellular aggregates induced by substrate rigidity. Soft Matter, 8: 4578-4583. doi: 10.1039/c2sm07418d
    [102]
    Du V, Luciani N, Richard S, Mary G, Gay C, et al. 2017. A 3D magnetic tissue stretcher for remote mechanical control of embryonic stem cell differentiation. Nature Communications, 8: 400. doi: 10.1038/s41467-017-00543-2
    [103]
    Dumbauld D W, Lee T T, Singh A, Scrimgeour J, Gersbach C A, et al. 2013. How vinculin regulates force transmission. Proceedings of the national academy of sciences of the united states of america, 110: 9788-93. doi: 10.1073/pnas.1216209110
    [104]
    Dumortier Julien G, Le Verge-Serandour M, Tortorelli Anna F, Mielke A, de Plater L, et al. 2019. Hydraulic fracturing and active coarsening position the lumen of the mouse blastocyst. Science, 365: 465-468. doi: 10.1126/science.aaw7709
    [105]
    Elbediwy A, Vincent-Mistiaen Z I, Spencer-Dene B, Stone R K, Boeing S, et al. 2016. Integrin signalling regulates YAP and TAZ to control skin homeostasis. Development, 143: 1674-87.
    [106]
    Elosegui-Artola A, Andreu I, Beedle A E M, Lezamiz A, Uroz M, et al. 2017. Force triggers yap nuclear entry by regulating transport across nuclear pores. Cell, 171: 1397-1410 e14.
    [107]
    Elosegui-Artola A, Bazellières E, Allen M D, Andreu I, Oria R, S et al. 2014. Rigidity sensing and adaptation through regulation of integrin types. Nature Materials, 13: 631-637. doi: 10.1038/nmat3960
    [108]
    Elosegui-Artola A, Oria R, Chen Y, Kosmalska A, Pérez-González C, et al. 2016. Mechanical regulation of a molecular clutch defines force transmission and transduction in response to matrix rigidity. Nature Cell Biology, 18: 540. doi: 10.1038/ncb3336
    [109]
    Elosegui-Artola A, Trepat X, Roca-Cusachs P. 2018. Control of mechanotransduction by molecular clutch dynamics. Trends in Cell Biology, 28: 356-367. doi: 10.1016/j.tcb.2018.01.008
    [110]
    Engler A J, Sen S, Sweeney H L, Discher D E. 2006. Matrix Elasticity Directs Stem Cell Lineage Specification. Cell, 126: 677-689. doi: 10.1016/j.cell.2006.06.044
    [111]
    Engler A, Bacakova L, Newman C, Hategan A, Griffin M, Discher D. 2004. Substrate compliance versus ligand density in cell on gel responses. Biophysical Journal, 86: 617-628. doi: 10.1016/S0006-3495(04)74140-5
    [112]
    Espina J A, Marchant C L, Barriga E H. 2021. Durotaxis: the mechanical control of directed cell migration. FEBS Journal, 289: 2736-2754.
    [113]
    Even-Ram S, Artym V, Yamada K M. 2006. Matrix control of stem cell fate. Cell, 126: 645-7. doi: 10.1016/j.cell.2006.08.008
    [114]
    Fang J, Hsueh Y Y, Soto J, Sun W, Wang J, et al. 2020. Engineering biomaterials with micro/nanotechnologies for cell reprogramming. ACS Nano, 14: 1296-1318. doi: 10.1021/acsnano.9b04837
    [115]
    Feng Q, Gao H, Wen H, Huang H, Li Q, et al. 2020. Engineering the cellular mechanical microenvironment to regulate stem cell chondrogenesis: Insights from a microgel model. Acta Biomater, 113: 393-406. doi: 10.1016/j.actbio.2020.06.046
    [116]
    Fillingham I, Gingras A, Papagrigoriou E, Patel B, Emsley J, et al. 2005. A vinculin binding domain from the talin rod unfolds to form a complex with the vinculin head. Structure, 13: 65-74. doi: 10.1016/j.str.2004.11.006
    [117]
    Fiore V F, Krajnc M, Quiroz F G, Levorse J, Pasolli H A, et al. 2020. Mechanics of a multilayer epithelium instruct tumour architecture and function. Nature, 585: 433-439. doi: 10.1038/s41586-020-2695-9
    [118]
    Foster C T, Gualdrini F, Treisman R. 2017. Mutual dependence of the MRTF-SRF and YAP-TEAD pathways in cancer-associated fibroblasts is indirect and mediated by cytoskeletal dynamics. Genes and Development, 31: 2361-2375. doi: 10.1101/gad.304501.117
    [119]
    Francesco R, Antonio G, Manlio B, Alfonso B. 2010. From cell-ECM interactions to tissue engineering. Journal of Cellular Physiology, 199: 174-180.
    [120]
    Friedl P, Gilmour D. 2009. Collective cell migration in morphogenesis, regeneration and cancer. Nature Reviews Molecular Cell Biology, 10: 445-457. doi: 10.1038/nrm2720
    [121]
    Friedl P, Mayor R. 2017. Tuning collective cell migration by cell-cell junction regulation. Cold Spring Harbor perspectives in biology, 9: a029199. doi: 10.1101/cshperspect.a029199
    [122]
    Friedland J C, Lee M H, Boettiger D. 2009. Mechanically activated integrin switch controls α5β1 function. Science, 323: 642. doi: 10.1126/science.1168441
    [123]
    Frisch T, Thoumine O. 2002. Predicting the kinetics of cell spreading. Journal of Biomechanics, 35: 1137-1141. doi: 10.1016/S0021-9290(02)00075-1
    [124]
    Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall J F, et al. 2007. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nature Cell Biology, 9: 1392-1400. doi: 10.1038/ncb1658
    [125]
    Garcia M A, Nelson W J, Chavez N. 2018. Cell-cell junctions organize structural and signaling networks. Cold Spring Harbor perspectives in biology, 10.
    [126]
    Gardel M L, Sabass B, Ji L, Danuser G, Schwarz U S, Waterman C M. 2008. Traction stress in focal adhesions correlates biphasically with actin retrograde flow speed. Journal of Cell Biology, 183: 999-1005. doi: 10.1083/jcb.200810060
    [127]
    Gauthier N C, Roca-Cusachs P. 2018. Mechanosensing at integrin-mediated cell-matrix adhesions: from molecular to integrated mechanisms. Current Opinion in Cell Biology, 50: 20-26. doi: 10.1016/j.ceb.2017.12.014
    [128]
    Geiger B, Spatz J P, Bershadsky A D. 2009. Environmental sensing through focal adhesions. Nature Reviews Molecular Cell Biology, 10: 21-33. doi: 10.1038/nrm2593
    [129]
    Ghassemi S, Meacci G, Liu S, Gondarenko A A, Mathur A, et al. 2012. Cells test substrate rigidity by local contractions on submicrometer pillars. Proceedings of the national academy of sciences of the united states of america, 109: 5328-33. doi: 10.1073/pnas.1119886109
    [130]
    Ghibaudo M, Saez A, Trichet L, Xayaphoummine A, Browaeys J, et al. 2008. Traction forces and rigidity sensing regulate cell functions. Soft Matter, 4: 1836-1843. doi: 10.1039/b804103b
    [131]
    Giannone G. 2015. Super-resolution links vinculin localization to function in focal adhesions. Nature Cell Biology, 17: 845-847. doi: 10.1038/ncb3196
    [132]
    Gkretsi V, Stylianopoulos T. 2018. Cell adhesion and matrix stiffness: coordinating cancer cell invasion and metastasis. Frontiers in Oncology, 8: 145-145. doi: 10.3389/fonc.2018.00145
    [133]
    Gong Z, Szczesny S E, Caliari S R, Charrier E E, Chaudhuri O, et al. 2018. Matching material and cellular timescales maximizes cell spreading on viscoelastic substrates. Proceedings of the national academy of sciences of the united states of america, 115: E2686-E2695.
    [134]
    Gooding J M, Yap K L, Ikura M. 2004. The cadherin–catenin complex as a focal point of cell adhesion and signalling: new insights from three‐dimensional structures. Bioessays, 26: 497-511. doi: 10.1002/bies.20033
    [135]
    Gough R E, Goult B T. 2018. The tale of two talins - two isoforms to fine-tune integrin signalling. Febs Letters, 592: 2108-2125. doi: 10.1002/1873-3468.13081
    [136]
    Grolman J M, Weinand P, Mooney D J. 2020. Extracellular matrix plasticity as a driver of cell spreading. Proceedings of the national academy of sciences of the united states of america, 117: 25999-26007. doi: 10.1073/pnas.2008801117
    [137]
    Guan X. 2015. Cancer metastases: challenges and opportunities. Acta Pharmaceutica Sinica B, 5: 402-418. doi: 10.1016/j.apsb.2015.07.005
    [138]
    Guilluy C, Swaminathan V, Garcia-Mata R, Timothy O’Brien E, Superfine R, Burridge K. 2011. The Rho GEFs LARG and GEF-H1 regulate the mechanical response to force on integrins. Nature Cell Biology, 13: 722-727. doi: 10.1038/ncb2254
    [139]
    Gul I S, Hulpiau P, Saeys Y, van Roy F. 2017. Evolution and diversity of cadherins and catenins. Experimental cell research, 358: 3-9. doi: 10.1016/j.yexcr.2017.03.001
    [140]
    Hadden W J, Young J L, Holle A W, McFetridge M L, Kim D Y, et al. 2017. Stem cell migration and mechanotransduction on linear stiffness gradient hydrogels. Proceedings of the national academy of sciences of the united states of america, 114: 5647-5652. doi: 10.1073/pnas.1618239114
    [141]
    Haining A W, Lieberthal T J, Del Río Hernández A. 2016. Talin: a mechanosensitive molecule in health and disease. FASEB Journal, 30: 2073-85. doi: 10.1096/fj.201500080R
    [142]
    Hamidi H, Ivaska J. 2018. Every step of the way: integrins in cancer progression and metastasis. Nature Reviews Cancer, 18: 533-548. doi: 10.1038/s41568-018-0038-z
    [143]
    Han S-B, Kim J-K, Lee G, Kim D-H. 2020. Mechanical properties of materials for stem cell differentiation. Advanced Biosystems, 4: 2000247. doi: 10.1002/adbi.202000247
    [144]
    Harland B, Walcott S, Sun S X. 2011. Adhesion dynamics and durotaxis in migrating cells. Physical Biology, 8: 015011-015011. doi: 10.1088/1478-3975/8/1/015011
    [145]
    Harrison O J, Bahna F, Katsamba P S, Jin X, Brasch J, et al. 2010. Two-step adhesive binding by classical cadherins. Nature Structural and Molecular Biology, 17: 348-57. doi: 10.1038/nsmb.1784
    [146]
    Hoffman B D, Grashoff C, Schwartz M A. 2011. Dynamic molecular processes mediate cellular mechanotransduction. Nature, 475: 316-23. doi: 10.1038/nature10316
    [147]
    Holmes W R, Edelstein-Keshet L. 2012. A comparison of computational models for eukaryotic cell shape and motility. PLoS Computational Biology, 8: e1002793. doi: 10.1371/journal.pcbi.1002793
    [148]
    Hong S, Troyanovsky R B, Troyanovsky S M. 2011. Cadherin exits the junction by switching its adhesive bond. Journal of Cell Biology, 192: 1073-83. doi: 10.1083/jcb.201006113
    [149]
    Hou Y, Yu L, Xie W, Camacho L C, Zhang M, Chu Z, et al. 2020. Surface roughness and substrate stiffness synergize to drive cellular mechanoresponse. Nano Lettersers, 20: 748-757. doi: 10.1021/acs.nanolett.9b04761
    [150]
    Houk A R, Jilkine A, Mejean C O, Boltyanskiy R, Dufresne E R, et al. 2012. Membrane tension maintains cell polarity by confining signals to the leading edge during neutrophil migration. Cell, 148: 175-88. doi: 10.1016/j.cell.2011.10.050
    [151]
    Hu S, Chen T-H, Zhao Y, Wang Z, Lam R H W. 2018. Protein–substrate adhesion in microcontact printing regulates cell behavior. Langmuir, 34: 1750-1759. doi: 10.1021/acs.langmuir.7b02935
    [152]
    Huang C, Akaishi S, Ogawa R. 2012. Mechanosignaling pathways in cutaneous scarring. Archives of Dermatological Research, 304: 589-597. doi: 10.1007/s00403-012-1278-5
    [153]
    Huang G, Li F, Zhao X, Ma Y, Li Y, et al. 2017. Functional and biomimetic materials for engineering of the three-dimensional cell microenvironment. Chemical Reviews, 117: 12764-12850. doi: 10.1021/acs.chemrev.7b00094
    [154]
    Huang J, Peng X, Xiong C, Fang J. 2011. Influence of substrate stiffness on cell-substrate interfacial adhesion and spreading: a mechano-chemical coupling model. Journal of Colloid and Interface Science, 355: 503-8. doi: 10.1016/j.jcis.2010.12.055
    [155]
    Huber A H, Weis W I. 2001. The structure of the β-catenin/e-cadherin complex and the molecular basis of diverse ligand recognition by β-catenin. Cell, 105: 391-402. doi: 10.1016/S0092-8674(01)00330-0
    [156]
    Humphries J D, Pengbo W, Charles S, Benny G, Humphries M J, Christoph B. 2007. Vinculin controls focal adhesion formation by direct interactions with talin and actin. Journal of Cell Biology, 179: 1043-1057. doi: 10.1083/jcb.200703036
    [157]
    Hynes R O. 2002. Integrins: bidirectional, allosteric signaling machines. Cell, 110: 673-687. doi: 10.1016/S0092-8674(02)00971-6
    [158]
    Isomursu A, Park K-Y, Hou J, Cheng B, Shamsan G, et al. 2022. Negative durotaxis: cell movement toward softer environments. Nature Materials.
    [159]
    Jannie K M, Ellerbroek S M, Zhou D W, Sophia C, Crompton D J, et al. 2015. Vinculin-dependent actin bundling regulates cell migration and traction forces. Biophysical Journal, 465: 383-93.
    [160]
    Jansen K A, Atherton P, Ballestrem C. 2017. Mechanotransduction at the cell-matrix interface. Seminars in Cell and Developmental Biology, 71: 75-83. doi: 10.1016/j.semcdb.2017.07.027
    [161]
    Jiang H, Qian J, Lin Y, Ni Y, He L. 2015. Aggregation dynamics of molecular bonds between compliant materials. Soft Matter, 11: 2812-20. doi: 10.1039/C4SM02903H
    [162]
    Jin J-K, Tien P-C, Cheng C-J, Song J H, Huang C, et al. 2015. Talin1 phosphorylation activates β1 integrins: a novel mechanism to promote prostate cancer bone metastasis. Oncogene, 34: 1811-1821. doi: 10.1038/onc.2014.116
    [163]
    Judokusumo E, Tabdanov E, Kumari S, Dustin M L, Kam L C. 2012. Mechanosensing in T lymphocyte activation. Biophysical Journal, 102: L5-7. doi: 10.1016/j.bpj.2011.12.011
    [164]
    Kaunas R, Usami S, Chien S. 2006. Regulation of stretch-induced JNK activation by stress fiber orientation. Cellular Signalling, 18: 1924-31. doi: 10.1016/j.cellsig.2006.02.008
    [165]
    Kechagia J Z, Ivaska J, Roca-Cusachs P. 2019. Integrins as biomechanical sensors of the microenvironment. Nature Reviews Molecular Cell Biology, 20: 457-473. doi: 10.1038/s41580-019-0134-2
    [166]
    Khalil A A, Friedl P. 2010. Determinants of leader cells in collective cell migration. Integrative Biology (Camb), 2: 568-74. doi: 10.1039/c0ib00052c
    [167]
    Killaars A R, Grim J C, Walker C J, Hushka E A, Brown T E, Anseth K S. 2019. Extended exposure to stiff microenvironments leads to persistent chromatin remodeling in human mesenchymal stem cells. Advanced Science, 6: 1801483. doi: 10.1002/advs.201801483
    [168]
    Kim D-H, Khatau S B, Feng Y, Walcott S, Sun S X, et al. 2012. Actin cap associated focal adhesions and their distinct role in cellular mechanosensing. Scientific Reports, 2: 555. doi: 10.1038/srep00555
    [169]
    Kim J-K, Louhghalam A, Lee G, Schafer B W, Wirtz D, Kim D-H. 2017. Nuclear lamin A/C harnesses the perinuclear apical actin cables to protect nuclear morphology. Nature Communications, 8: 2123. doi: 10.1038/s41467-017-02217-5
    [170]
    Kong D, Ji B, Dai L. 2010. Stabilizing to disruptive transition of focal adhesion response to mechanical forces. Journal of Biomechanics, 43: 2524-9. doi: 10.1016/j.jbiomech.2010.05.019
    [171]
    Kourtidis A, Ngok S P, Anastasiadis P Z. 2013. Chapter eighteen - p120 catenin: an essential regulator of cadherin stability, adhesion-induced signaling, and cancer progression. Progress in molecular biology and translational science, 116: 409-432.
    [172]
    Krasik E F, Caputo K E, Hammer D A. 2008. Adhesive dynamics simulation of neutrophil arrest with stochastic activation. Biophysical Journal, 95: 1716-28. doi: 10.1529/biophysj.107.119677
    [173]
    Kumar S, Parekh S H. 2021. Molecular control of the interfacial fibronectin structure on graphene oxide steers cell fate. ACS applied materials & interfaces, 13: 2346-2359.
    [174]
    Kuninty P R, Bansal R, De Geus S W L, Mardhian D F, Schnittert J, et al. 2019. ITGA5 inhibition in pancreatic stellate cells attenuates desmoplasia and potentiates efficacy of chemotherapy in pancreatic cancer. Science Advances, 5: eaax2770. doi: 10.1126/sciadv.aax2770
    [175]
    Kuo C H, Xian J, Brenton J D, Franze K, Sivaniah E. 2012. Complex stiffness gradient substrates for studying mechanotactic cell migration. Advanced Materials, 24: 6059-64. doi: 10.1002/adma.201202520
    [176]
    Kwa M Q, Herum K M, Brakebusch C. 2019. Cancer-associated fibroblasts: how do they contribute to metastasis? Clinical & Experimental Metastasi, 36: 71-86.
    [177]
    Kwon M Y, Vega S L, Gramlich W M, Kim M, Mauck R L, Burdick J A. 2018. Dose and timing of n-cadherin mimetic peptides regulate msc chondrogenesis within hydrogels. Advanced healthcare materials, 7: e1701199. doi: 10.1002/adhm.201701199
    [178]
    Labernadie A, Kato T, Brugues A, Serra-Picamal X, Derzsi S, et al. 2017. A mechanically active heterotypic E-cadherin/N-cadherin adhesion enables fibroblasts to drive cancer cell invasion. Nature Cell Biology, 19: 224-237. doi: 10.1038/ncb3478
    [179]
    Lachowski D, Cortes E, Robinson B, Rice A, Rombouts K, Del Río Hernández A E. 2018. FAK controls the mechanical activation of YAP, a transcriptional regulator required for durotaxis. FASEB Journal, 32: 1099-1107. doi: 10.1096/fj.201700721R
    [180]
    Ladoux B, Mege R M. 2017. Mechanobiology of collective cell behaviours. Nature Reviews Molecular Cell Biology, 18: 743-757. doi: 10.1038/nrm.2017.98
    [181]
    Ladoux B, Anon E, Lambert M, Rabodzey A, Hersen P, et al. 2010. Strength Dependence of Cadherin-Mediated Adhesions. Biophysical Journal, 98: 534-542. doi: 10.1016/j.bpj.2009.10.044
    [182]
    Ladoux B, Mège R-M, Trepat X. 2016. Front–rear polarization by mechanical cues: from single cells to tissues. Trends in Cell Biology, 26: 420-433. doi: 10.1016/j.tcb.2016.02.002
    [183]
    Ladoux B, Nelson W J, Yan J, Mège R M. 2015. The mechanotransduction machinery at work at adherens junctions. Integrative Biology (Camb), 7: 1109-19. doi: 10.1039/c5ib00070j
    [184]
    le Duc Q, Shi Q, Blonk I, Sonnenberg A, Wang N, et al. 2010. Vinculin potentiates E-cadherin mechanosensing and is recruited to actin-anchored sites within adherens junctions in a myosin II-dependent manner. Journal of Cell Biology, 189: 1107-15. doi: 10.1083/jcb.201001149
    [185]
    Leckband D E, Rooij J D. 2014. Cadherin adhesion and mechanotransduction. Annual Review of Cell and Developmental Biology, 30: 291-315. doi: 10.1146/annurev-cellbio-100913-013212
    [186]
    Leckband D E, Duc Q L, Ning W, Rooij J D. 2011. Mechanotransduction at cadherin-mediated adhesions. Current Opinion in Cell Biology, 23: 523-530. doi: 10.1016/j.ceb.2011.08.003
    [187]
    Lee H-p, Stowers R, Chaudhuri O. 2019. Volume expansion and trpv4 activation regulate stem cell fate in three-dimensional microenvironments. Nature Communications, 10: 529. doi: 10.1038/s41467-019-08465-x
    [188]
    Lee J W, An H, Lee K Y. 2017. Introduction of N-cadherin-binding motif to alginate hydrogels for controlled stem cell differentiation. Colloids and Surfaces, B:Biointerfaces, 155: 229-237. doi: 10.1016/j.colsurfb.2017.04.014
    [189]
    Legate K R, Wickström S A, Fässler R. 2009. Genetic and cell biological analysis of integrin outside-in signaling. Genes & Development, 23: 397-418.
    [190]
    Lei K, Kurum A, Tang L. 2020. Mechanical immunoengineering of t cells for therapeutic applications. Accounts of Chemical Research, 53: 2777-2790. doi: 10.1021/acs.accounts.0c00486
    [191]
    Leiphart R J, Chen D, Peredo A P, Loneker A E, Janmey P A. 2019. Mechanosensing at cellular interfaces. Langmuir, 35: 7509-7519. doi: 10.1021/acs.langmuir.8b02841
    [192]
    Levental K R, Yu H, Kass L, Lakins J N, Egeblad M, et al. 2009. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell, 139: 891-906. doi: 10.1016/j.cell.2009.10.027
    [193]
    Li C X, Talele N P, Boo S, Koehler A, Knee-Walden E, et al. 2016. Microrna-21 preserves the fibrotic mechanical memory of mesenchymal stem cells. Nature Materials, 16: 379.
    [194]
    Li J, Di Russo J, Hua X, Chu Z, Spatz J P, Wei Q. 2019. Surface immobilized e-cadherin mimetic peptide regulates the adhesion and clustering of epithelial cells. Advanced Healthcare Materials, 8: 1801384. doi: 10.1002/adhm.201801384
    [195]
    Li J, Li X, Zhang J, Kawazoe N, Chen G. 2017. Induction of chondrogenic differentiation of human mesenchymal stem cells by biomimetic gold nanoparticles with tunable rgd density. Advanced Healthcare Materials, 6. 28489328
    [196]
    Li L, Bennett S A L, Wang L. 2012. Role of E-cadherin and other cell adhesion molecules in survival and differentiation of human pluripotent stem cells. Cell Adhesion & Migration, 6: 59-70.
    [197]
    Li R, Xu J, Wong D S H, Li J, Zhao P, Bian L. 2017. Self-assembled N-cadherin mimetic peptide hydrogels promote the chondrogenesis of mesenchymal stem cells through inhibition of canonical Wnt/β-catenin signaling. Biomaterials, 145: 33-43. doi: 10.1016/j.biomaterials.2017.08.031
    [198]
    Li S, Wang X, Cao B, Ye K, Li Z, Ding J. 2015. Effects of nanoscale spatial arrangement of arginine-glycine-aspartate peptides on dedifferentiation of chondrocytes. Nano Letters, 15: 7755-65. doi: 10.1021/acs.nanolett.5b04043
    [199]
    Li Y, Kilian K A. 2015. Bridging the gap: from 2d cell culture to 3d microengineered extracellular matrices. Advanced Healthcare Materials, 4: 2780-2796. doi: 10.1002/adhm.201500427
    [200]
    Li Y, Altorelli N L, Bahna F, Honig B, Shapiro L, Palmer A G. 2013. Mechanism of E-cadherin dimerization probed by NMR relaxation dispersion. Proceedings of the National Academy of Sciences of the United States of America, 110: 16462-7. doi: 10.1073/pnas.1314303110
    [201]
    Li Y, Mao A S, Seo B R, Zhao X, Gupta S K, et al. 2020. Compression-induced dedifferentiation of adipocytes promotes tumor progression. Science Advances, 6: eaax5611. doi: 10.1126/sciadv.aax5611
    [202]
    Li Z, Shen D, Hu S, Su T, Huang K, et al. 2018. Pretargeting and bioorthogonal click chemistry-mediated endogenous stem cell homing for heart repair. ACS Nano, 12: 12193-12200. doi: 10.1021/acsnano.8b05892
    [203]
    Lin S Z, Li B, Lan G, Feng X Q. 2017. Activation and synchronization of the oscillatory morphodynamics in multicellular monolayer. Proceedings of the National Academy of Sciences of the United States of America, 114: 8157-8162. doi: 10.1073/pnas.1705492114
    [204]
    Ling K, Doughman R L, Firestone A J, Bunce M W, Anderson R A. 2002. Type Iγ phosphatidylinositol phosphate kinase targets and regulates focal adhesions. Nature, 420: 89-93. doi: 10.1038/nature01082
    [205]
    Ling K, Doughman R L, Iyer V V, Firestone A J, Bairstow S F, et al. 2003. Tyrosine phosphorylation of type Iγ phosphatidylinositol phosphate kinase by Src regulates an integrin–talin switch. Journal of Cell Biology, 163: 1339-1349. doi: 10.1083/jcb.200310067
    [206]
    Liu J, Tan Y, Zhang H, Zhang Y, Xu P, et al. 2012. Soft fibrin gels promote selection and growth of tumorigenic cells. Nature materials, 11: 734-741. doi: 10.1038/nmat3361
    [207]
    Liu L, Zhang S X, Liao W, Farhoodi H P, Wong C W, et al. 2017. Mechanoresponsive stem cells to target cancer metastases through biophysical cues. Science Translational Medicine, 9: eaan2966. doi: 10.1126/scitranslmed.aan2966
    [208]
    Liu Y A, Liang B Y, Guan Y, You J, Zhu L, et al. 2015. Loss of N-cadherin is associated with loss of E-cadherin expression and poor outcomes of liver resection in hepatocellular carcinoma. Journal of Surgical Research, 194: 167-76. doi: 10.1016/j.jss.2014.09.031
    [209]
    Liu Z, Tan J L, Cohen D M, Yang M T, Sniadecki N J, et al. 2010. Mechanical tugging force regulates the size of cell—cell junctions. Proceedings of the National Academy of Sciences of the United States of America, 107: 9944-9949. doi: 10.1073/pnas.0914547107
    [210]
    Lo C-M, Wang H-B, Dembo M, Wang Y-l. 2000. Cell movement is guided by the rigidity of the substrate. Biophysical Journal, 79: 144-152. doi: 10.1016/S0006-3495(00)76279-5
    [211]
    Lou J, Yago T, Klopocki A G, Mehta P, Chen W, et al. 2006. Flow-enhanced adhesion regulated by a selectin interdomain hinge. Journal of Cell Biology, 174: 1107-17. doi: 10.1083/jcb.200606056
    [212]
    Lyons A J, Jones J. 2007. Cell adhesion molecules, the extracellular matrix and oral squamous carcinoma. International Journal of Oral and Maxillofacial Implants, 36: 671-679. doi: 10.1016/j.ijom.2007.04.002
    [213]
    Ma R, Kellner A V, Ma V P, Su H, Deal B R, et al. 2019. DNA probes that store mechanical information reveal transient piconewton forces applied by T cells. Proceedings of the National Academy of Sciences of the United States of America, 116: 16949-16954. doi: 10.1073/pnas.1904034116
    [214]
    Ma Y, Lin M, Huang G, Li Y, Wang S, et al. 2018. 3D spatiotemporal mechanical microenvironment: a hydrogel-based platform for guiding stem cell fate. Advanced Materials, 30: 1705911. doi: 10.1002/adma.201705911
    [215]
    Madl C M, LeSavage B L, Dewi R E, Dinh C B, Stowers R S, et al. 2017. Maintenance of neural progenitor cell stemness in 3d hydrogels requires matrix remodelling. Nature Materials, 16: 1233-1242. doi: 10.1038/nmat5020
    [216]
    Madl C M, LeSavage B L, Dewi R E, Lampe K J, Heilshorn S C. 2019. Matrix remodeling enhances the differentiation capacity of neural progenitor cells in 3D hydrogels. Advanced Materials, 6: 1801716.
    [217]
    Manibog K, Li H, Rakshit S, Sivasankar S. 2014. Resolving the molecular mechanism of cadherin catch bond formation. Nature Communications, 5: 3941. doi: 10.1038/ncomms4941
    [218]
    Manibog K, Sankar K, Kim S A, Zhang Y, Jernigan R L, Sivasankar S. 2016. Molecular determinants of cadherin ideal bond formation: Conformation-dependent unbinding on a multidimensional landscape. Proceedings of the National Academy of Sciences of the United States of America, 113: E5711-20.
    [219]
    Mao C Y, Wang Y G, Zhang X, Zheng X Y, Tang T T, Lu E Y. 2016. Double-edged-sword effect of IL-1β on the osteogenesis of periodontal ligament stem cells via crosstalk between the NF-κB, MAPK and BMP/Smad signaling pathways. Cell Death & Disease, 7: e2296.
    [220]
    Martino F, Perestrelo A R, Vinarsky V, Pagliari S, Forte G. 2018. Cellular mechanotransduction: from tension to function. Frontiers in Physiology, 9: 824. doi: 10.3389/fphys.2018.00824
    [221]
    Maruthamuthu V, Aratyn-Schaus Y and Gardel M L. 2010. Conserved F-actin dynamics and force transmission at cell adhesions. Current Opinion in Cell Biology, 22: 583-8. doi: 10.1016/j.ceb.2010.07.010
    [222]
    Maruthamuthu V, Sabass B, Schwarz U S, Gardel M L, Chien S. 2011. Cell-ECM traction force modulates endogenous tension at cell-cell contacts. Proceedings of the National Academy of Sciences of the United States of America, 108: 4708-4713. doi: 10.1073/pnas.1011123108
    [223]
    Mayor R, Etienne-Manneville S. 2016. The front and rear of collective cell migration. Nature Reviews Molecular Cell Biology, 17: 97-109. doi: 10.1038/nrm.2015.14
    [224]
    Mège R M, Gavard J, Lambert M. 2006. Regulation of cell–cell junctions by the cytoskeleton. Current Opinion in Cell Biology, 18: 541-548. doi: 10.1016/j.ceb.2006.08.004
    [225]
    Miller K D, Nogueira L, Mariotto A B, Rowland J H, Yabroff K R, et al. 2019. Cancer treatment and survivorship statistics. CA:A Cancer Journal for Clinicians, 69: 363-385. doi: 10.3322/caac.21565
    [226]
    Mitchison T, Kirschner M. 1988. Cytoskeletal dynamics and nerve growth. Neuron, 1: 761-72. doi: 10.1016/0896-6273(88)90124-9
    [227]
    Mitra S K, Schlaepfer D D. 2006. Integrin-regulated FAK–Src signaling in normal and cancer cells. Current Opinion in Cell Biology, 18: 516-523. doi: 10.1016/j.ceb.2006.08.011
    [228]
    Miyazaki K, Oyanagi J, Hoshino D, Togo S, Kumagai H, Miyagi Y. 2019. Cancer cell migration on elongate protrusions of fibroblasts in collagen matrix. Scientific Reports, 9: 292. doi: 10.1038/s41598-018-36646-z
    [229]
    Mongera A, Rowghanian P, Gustafson H J, Shelton E, Kealhofer D A, et al. 2018. A fluid-to-solid jamming transition underlies vertebrate body axis elongation. Nature, 561: 401-405. doi: 10.1038/s41586-018-0479-2
    [230]
    Muhamed I, Wu J, Sehgal P, Kong X, Tajik A, et al. 2016. E-cadherin-mediated force transduction signals regulate global cell mechanics. Journal of Cell Science, 129: 1843-54.
    [231]
    Mui K L, Chen C S, Assoian R K. 2016. The mechanical regulation of integrin-cadherin crosstalk organizes cells, signaling and forces. Journal of Cell Science, 129: 1093-1100.
    [232]
    Najafi M, Farhood B, Mortezaee K. 2019. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. Journal of Cellular Biochemistry, 120: 2782-2790. doi: 10.1002/jcb.27681
    [233]
    Nasrollahi S, Walter C, Loza A J, Schimizzi G V, Longmore G D, Pathak A. 2017. Past matrix stiffness primes epithelial cells and regulates their future collective migration through a mechanical memory. Biomaterials, 146: 146-155. doi: 10.1016/j.biomaterials.2017.09.012
    [234]
    Nemec S, Kilian K A. 2021. Materials control of the epigenetics underlying cell plasticity. Nature Reviews Materials, 6: 69-83. doi: 10.1038/s41578-020-00238-z
    [235]
    Ng M R, Besser A, Danuser G, Brugge J S. 2012. Substrate stiffness regulates cadherin-dependent collective migration through myosin-II contractility. Journal of Cell Biology, 199: 545-563. doi: 10.1083/jcb.201207148
    [236]
    Nia H T, Munn L L, Jain R K. 2020. Physical traits of cancer. Science, 370: eaaz0868. doi: 10.1126/science.aaz0868
    [237]
    Nickaeen M, Novak I L, Pulford S, Rumack A, Brandon J, et al. 2017. A free-boundary model of a motile cell explains turning behavior. PLoS Computational Biology, 13: e1005862. doi: 10.1371/journal.pcbi.1005862
    [238]
    Nicolas A, Safran S A. 2006. Limitation of cell adhesion by the elasticity of the extracellular matrix. Biophysical Journal, 91: 61-73. doi: 10.1529/biophysj.105.077115
    [239]
    Nicolas A, Besser A, Safran S A. 2008. Dynamics of cellular focal adhesions on deformable substrates: consequences for cell force microscopy. Biophysical Journal, 95: 527-39. doi: 10.1529/biophysj.107.127399
    [240]
    Nicolas A, Geiger B, Safran S A. 2004. Cell mechanosensitivity controls the anisotropy of focal adhesions. Proceedings of the National Academy of Sciences of the United States of America, 101: 12520-5. doi: 10.1073/pnas.0403539101
    [241]
    Nolan J, Mahdi A F, Dunne C P, Kiely P A. 2020. Collagen and fibronectin promote an aggressive cancer phenotype in breast cancer cells but drive autonomous gene expression patterns. Gene, 761.
    [242]
    O'Connor R S, Hao X, Shen K, Bashour K, Akimova T, et al. 2012. Substrate rigidity regulates human T cell activation and proliferation. Journal of Immunology, 189: 1330-9. doi: 10.4049/jimmunol.1102757
    [243]
    Onder T T, Gupta P B, Mani S A, Yang J, Lander E S, Weinberg R A. 2008. Loss of e-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Research, 68: 3645. doi: 10.1158/0008-5472.CAN-07-2938
    [244]
    Oria R, Wiegand T, Escribano J, Elosegui-Artola A, Uriarte J J, et al. 2017. Force loading explains spatial sensing of ligands by cells. Nature, 552: 219-224. doi: 10.1038/nature24662
    [245]
    Owen L M, Adhikari A S, Patel M, Grimmer P, Leijnse N, et al. 2017. A cytoskeletal clutch mediates cellular force transmission in a soft, three-dimensional extracellular matrix. Molecular Biology of the Cell, 28: 1959-1974. doi: 10.1091/mbc.e17-02-0102
    [246]
    Pannekoek W, de Rooij J, Gloerich M. 2019. Force transduction by cadherin adhesions in morphogenesis. F1000Research, 8.
    [247]
    Papusheva E, Heisenberg C-P. 2010. Spatial organization of adhesion: force-dependent regulation and function in tissue morphogenesis. EMBO Journal, 29: 2753-2768. doi: 10.1038/emboj.2010.182
    [248]
    Parsons J T, Horwitz A R, Schwartz M A. Cell adhesion: integrating cytoskeletal dynamics and cellular tension. Nature Reviews Molecular Cell Biology, 11: 633-643.
    [249]
    Paszek M J, Boettiger D, Weaver V M, Hammer D A. 2009. Integrin clustering is driven by mechanical resistance from the glycocalyx and the substrate. PLoS Computational Biology, 5: e1000604. doi: 10.1371/journal.pcbi.1000604
    [250]
    Paszek M J, DuFort C C, Rossier O, Bainer R, Mouw J K, et al. 2014. The cancer glycocalyx mechanically primes integrin-mediated growth and survival. Nature, 511: 319-25. doi: 10.1038/nature13535
    [251]
    Peng X, Huang J, Xiong C, Fang J. 2012. Cell adhesion nucleation regulated by substrate stiffness: a Monte Carlo study. Journal of Biomechanics, 45: 116-22. doi: 10.1016/j.jbiomech.2011.09.013
    [252]
    Peng Y, Chen Z, Chen Y, Li S, Jiang Y, et al. 2019. ROCK isoforms differentially modulate cancer cell motility by mechanosensing the substrate stiffness. Acta Biomaterialia, 88: 86-101. doi: 10.1016/j.actbio.2019.02.015
    [253]
    Peng Y, Chen Z, He Y, Li P, Chen Y, et al. 2022. Non-muscle myosin II isoforms orchestrate substrate stiffness sensing to promote cancer cell contractility and migration. Cancer Letters, 524: 245-258. doi: 10.1016/j.canlet.2021.10.030
    [254]
    Perez T D, Tamada M, Sheetz M P, Nelson W J. 2008. Immediate-early signaling induced by E-cadherin engagement and adhesion. Journal of Biomechanics, 283: 5014-22.
    [255]
    Petrie R J, Doyle A D, Yamada K M. 2009. Random versus directionally persistent cell migration. Nature Reviews Molecular Cell Biology, 10: 538-49. doi: 10.1038/nrm2729
    [256]
    Picaut L, Trichet L, Helary C, Ducourthial G, Bonnin M-A, et al. 2021. Core-shell pure collagen threads extruded from highly concentrated solutions promote colonization and differentiation of C3H10T1/2 cells. ACS Biomaterials Science & Engineering, 7: 626-635.
    [257]
    Piersma B, Bank R A, Boersema M. 2015. Signaling in fibrosis: TGF-β, WNT, and YAP/TAZ converge. Frontiers in Medicine (Lausanne), 2: 59.
    [258]
    Plotnikov S V, Waterman C M. 2013. Guiding cell migration by tugging. Current Opinion in Cell Biology, 25: 619-626. doi: 10.1016/j.ceb.2013.06.003
    [259]
    Plotnikov S V, Pasapera A M, Sabass B, Waterman C M. 2012. Force fluctuations within focal adhesions mediate ECM-rigidity sensing to guide directed cell migration. Cell, 151: 1513-27. doi: 10.1016/j.cell.2012.11.034
    [260]
    Pollard T D, Borisy G G. 2003. Cellular motility driven by assembly and disassembly of actin filaments. Cell, 112: 453-465. doi: 10.1016/S0092-8674(03)00120-X
    [261]
    Priest A V, Shafraz O, Sivasankar S. 2017. Biophysical basis of cadherin mediated cell-cell adhesion. Experimental Cell Research, 358: 10-13. doi: 10.1016/j.yexcr.2017.03.015
    [262]
    Priya R, Gomez G A, Budnar S, Verma S, Cox H L, et al. 2015. Feedback regulation through myosin II confers robustness on RhoA signalling at E-cadherin junctions. Nature Cell Biology, 17: 1282-93. doi: 10.1038/ncb3239
    [263]
    Priya R, Yap A S, Gomez G A. 2013. E-cadherin supports steady-state Rho signaling at the epithelial zonula adherens. Differentiation, 86: 133-40. doi: 10.1016/j.diff.2013.01.002
    [264]
    Qi L, Jafari N, Li X, Chen Z, Li L, et al. 2016. Talin2-mediated traction force drives matrix degradation and cell invasion. Journal of Cell Science, 129: 3661-3674. doi: 10.1242/jcs.185959
    [265]
    Qi Y X, Yao Q P, Huang K, Shi Q, Zhang P, et al. 2016. Nuclear envelope proteins modulate proliferation of vascular smooth muscle cells during cyclic stretch application. Proceedings of the National Academy of Sciences of the United States of America, 113: 5293-8. doi: 10.1073/pnas.1604569113
    [266]
    Qi Y-X, Jiang J, Jiang X-H, Wang X-D, Ji S-Y, et al. 2011. PDGF-BB and TGF-β1 on cross-talk between endothelial and smooth muscle cells in vascular remodeling induced by low shear stress. Proceedings of the National Academy of Sciences of the United States of America, 108: 1908-1913. doi: 10.1073/pnas.1019219108
    [267]
    Qian J, Liu H, Lin Y, Chen W, Gao H. 2013. A mechanochemical model of cell reorientation on substrates under cyclic stretch. PLoS One, 8: e65864. doi: 10.1371/journal.pone.0065864
    [268]
    Qian J, Wang J, Gao H. 2008. Lifetime and strength of adhesive molecular bond clusters between elastic media. Langmuir, 24: 1262-70. doi: 10.1021/la702401b
    [269]
    Qian J, Wang J, Lin Y, Gao H. 2009. Lifetime and strength of periodic bond clusters between elastic media under inclined loading. Biophysical Journal, 97: 2438-45. doi: 10.1016/j.bpj.2009.08.027
    [270]
    Qian L, Saltzman W M. 2004. Improving the expansion and neuronal differentiation of mesenchymal stem cells through culture surface modification. Biomaterials, 25: 1331-1337. doi: 10.1016/j.biomaterials.2003.08.013
    [271]
    Qin X, Park B O, Liu J, Chen B, Choesmel-Cadamuro V, et al. 2017. Cell-matrix adhesion and cell-cell adhesion differentially control basal myosin oscillation and Drosophila egg chamber elongation. Nature Communications, 8: 14708. doi: 10.1038/ncomms14708
    [272]
    Qin X, Zhang Y, He Y, Chen K, Zhang Y, et al. 2021. Shear stress triggered circular dorsal ruffles formation to facilitate cancer cell migration. Archives of Biochemistry and Biophysics, 709: 108967. doi: 10.1016/j.abb.2021.108967
    [273]
    Raghupathy R, Anilkumar Anupama A, Polley A, Singh Parvinder P, Yadav M, et al. 2015. Transbilayer lipid interactions mediate nanoclustering of lipid-anchored proteins. Cell, 161: 581-594. doi: 10.1016/j.cell.2015.03.048
    [274]
    Rakshit S, Zhang Y, Manibog K, Shafraz O, Sivasankar S. 2012. Ideal, catch, and slip bonds in cadherin adhesion. Proceedings of the National Academy of Sciences of the United States of America, 109: 18815. doi: 10.1073/pnas.1208349109
    [275]
    Ramaswamy G, Bidez M W, Misch C E. 2015. Chapter 6 – Bone response to mechanical loads//Dental Implant Prosthetics, 2nd edn. Netgherlands: Elsevier.
    [276]
    Ratnikov B I, Partridge AWGinsberg M H. 2005. Integrin activation by talin. Journal of Thrombosis and Haemostasis, 3: 1783-1790. doi: 10.1111/j.1538-7836.2005.01362.x
    [277]
    Redmer T, Diecke S, Grigoryan T, Quiroga-Negreira A, Birchmeier W, Besser D. 2011. E-cadherin is crucial for embryonic stem cell pluripotency and can replace OCT4 during somatic cell reprogramming. EMBO Reports, 12: 720-726. doi: 10.1038/embor.2011.88
    [278]
    Rees D J G, Ades S E, Singer S J, Hynes R O. 1990. Sequence and domain structure of talin. Nature, 347: 685-689. doi: 10.1038/347685a0
    [279]
    Rens E G, Merks R M H. 2020. Cell Shape and Durotaxis Explained from Cell-Extracellular Matrix Forces and Focal Adhesion Dynamics. iScience, 23: 101488. doi: 10.1016/j.isci.2020.101488
    [280]
    Richardson A M, Havel L S, Koyen A E, Konen J M, Shupe J, et al. 2018. Vimentin is required for lung adenocarcinoma metastasis via heterotypic tumor cell-cancer-associated fibroblast interactions during collective invasion. Clinical Cancer Research, 24: 420-432. doi: 10.1158/1078-0432.CCR-17-1776
    [281]
    Ridley A J. 2006. Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends in Cell Biology, 16: 522-9. doi: 10.1016/j.tcb.2006.08.006
    [282]
    Roca-Cusachs P, Iskratsch T, Sheetz M. 2012. Finding the weakest link: Exploring integrin-mediated mechanical molecular pathways. Journal of Cell Science, 125: 3025-38.
    [283]
    Romanazzo S, Lin K, Srivastava P, Kilian K A. 2020. Targeting cell plasticity for regeneration: From in vitro to in vivo reprogramming. Advanced Drug Delivery Reviews, 161-162: 124-144. doi: 10.1016/j.addr.2020.08.007
    [284]
    Ronen Z B, Benjamin G. 2010. The switchable integrin adhesome. Journal of Cell Science, 123: 1385-1388. doi: 10.1242/jcs.066183
    [285]
    Roy B, Venkatachalapathy S, Ratna P, Wang Y, Jokhun D S, et al. 2018. Laterally confined growth of cells induces nuclear reprogramming in the absence of exogenous biochemical factors. Proceedings of the National Academy of Sciences of the United States of America, 115: E4741.
    [286]
    Roy B, Yuan L, Lee Y, Bharti A, Mitra A, Shivashankar G V. 2020. Fibroblast rejuvenation by mechanical reprogramming and redifferentiation. Proceedings of the National Academy of Sciences of the United States of America, 117: 10131-10141. doi: 10.1073/pnas.1911497117
    [287]
    Ruggiero F, Comte J, Cabañas C, Garrone R. 1996. Structural requirements for alpha 1 beta 1 and alpha 2 beta 1 integrin mediated cell adhesion to collagen V. Journal of Cell Science, 109 ( Pt 7): 1865-1874.
    [288]
    Saenz-de-Santa-Maria I, Celada L, Chiara M D. 2020. The leader position of mesenchymal cells expressing n-cadherin in the collective migration of epithelial cancer. Cells, 9: 731. doi: 10.3390/cells9030731
    [289]
    Saunders R M, Holt M R, Jennings L, Sutton D H, Barsukov I L, et al. 2006. Role of vinculin in regulating focal adhesion turnover. European Journal of Cell Biology, 85: 487-500. doi: 10.1016/j.ejcb.2006.01.014
    [290]
    Schiller H B, Hermann M R, Polleux J, Vignaud T, Zanivan S, et al. 2013. Beta1- and alphav-class integrins cooperate to regulate myosin II during rigidity sensing of fibronectin-based microenvironments. Nature Cell Biology, 15: 625-36. doi: 10.1038/ncb2747
    [291]
    Schnittert J, Bansal R, Storm G, Prakash J. 2018. Integrins in wound healing, fibrosis and tumor stroma: High potential targets for therapeutics and drug delivery. Advanced Drug Delivery Reviews, 129: 37-53. doi: 10.1016/j.addr.2018.01.020
    [292]
    Schroeder M E, Gonzalez Rodriguez A, Speckl K F, Walker C J, Midekssa F S, et al. 2021. Collagen networks within 3D PEG hydrogels support valvular interstitial cell matrix mineralization. Acta Biomaterialia, 119: 197-210. doi: 10.1016/j.actbio.2020.11.012
    [293]
    Segel M, Neumann B, Hill M F E, Weber I P, Viscomi C, et al. 2019. Niche stiffness underlies the ageing of central nervous system progenitor cells. Nature, 573: 130-134. doi: 10.1038/s41586-019-1484-9
    [294]
    Seong J, Tajik A, Sun J, Guan J-L, Humphries M J, et al. 2013. Distinct biophysical mechanisms of focal adhesion kinase mechanoactivation by different extracellular matrix proteins. Proceedings of the National Academy of Sciences of the United States of America, 110: 19372. doi: 10.1073/pnas.1307405110
    [295]
    Shapiro L, Weis W I. 2009. Structure and biochemistry of cadherins and catenins. Cold Spring Harbor Perspectives in Biology, 1: a003053.
    [296]
    Shapiro L, Fannon A M, Kwong P D, Thompson A, Lehmann M S, et al. 1995. Structural basis of cell-cell adhesion by cadherins. Nature, 374: 327-37. doi: 10.1038/374327a0
    [297]
    Shellard A, Mayor R. 2020. All Roads Lead to Directional Cell Migration. Trends in Cell Biology, 30: 852-868. doi: 10.1016/j.tcb.2020.08.002
    [298]
    Shibata A C E, Fujiwara T K, Chen L, Suzuki K G N, Ishikawa Y, et al. 2012. Archipelago architecture of the focal adhesion: Membrane molecules freely enter and exit from the focal adhesion zone. Cytoskeleton, 69: 380-392. doi: 10.1002/cm.21032
    [299]
    Shih Y R, Tseng K F, Lai H Y, Lin C H, Lee O K. 2011. Matrix stiffness regulation of integrin-mediated mechanotransduction during osteogenic differentiation of human mesenchymal stem cells. Journal of Bone and Mineral Research, 26: 730-8. doi: 10.1002/jbmr.278
    [300]
    Shiu J-Y, Aires L, Lin Z, Vogel V. 2018. Nanopillar force measurements reveal actin-cap-mediated YAP mechanotransduction. Nature Cell Biology, 20: 262-271. doi: 10.1038/s41556-017-0030-y
    [301]
    Shivashankar G V. 2019. Mechanical regulation of genome architecture and cell-fate decisions. Current Opinion in Cell Biology, 56: 115-121. doi: 10.1016/j.ceb.2018.12.001
    [302]
    Silva C S, Pinto R D, Amorim S, Pires R A, Correia-Neves M, et al. 2020. Fibronectin-functionalized fibrous meshes as a substrate to support cultures of thymic epithelial cells. Biomacromolecules, 21: 4771-4780. doi: 10.1021/acs.biomac.0c00933
    [303]
    Sivasankar S. 2013. Tuning the kinetics of cadherin adhesion. Journal of Investigative Dermatology, 133: 2318-2323. doi: 10.1038/jid.2013.229
    [304]
    Slack R J, Macdonald S J F, Roper J A, Jenkins R G, Hatley R J D. 2022. Emerging therapeutic opportunities for integrin inhibitors. Nature Reviews Drug Discovery, 21: 60-78. doi: 10.1038/s41573-021-00284-4
    [305]
    Slováková J, Sikora M, Arslan F N, Caballero-Mancebo S, Krens S F G, et al. 2022. Tension-dependent stabilization of E-cadherin limits cell–cell contact expansion in zebrafish germ-layer progenitor cells. Proceedings of the National Academy of Sciences of the United States of America, 119: e2122030119. doi: 10.1073/pnas.2122030119
    [306]
    Smith M A, Hoffman L M, Beckerle M C. 2014. LIM proteins in actin cytoskeleton mechanoresponse. Trends in Cell Biology, 24: 575-83. doi: 10.1016/j.tcb.2014.04.009
    [307]
    Smith Q, Chan X Y, Carmo A M, Trempel M, Saunders M, Gerecht S. 2017. Compliant substratum guides endothelial commitment from human pluripotent stem cells. Science Advances, 3: e1602883. doi: 10.1126/sciadv.1602883
    [308]
    Smutny M, Cox H L, Leerberg J M, Kovacs E M, Conti M A, et al. 2010. Myosin II isoforms identify distinct functional modules that support integrity of the epithelial zonula adherens. Nature Cell Biology, 12: 696-702. doi: 10.1038/ncb2072
    [309]
    S Sonal, Ganzinger K A, Vogel S K, Mücksch J, Blumhardt P, Schwille P. 2018. Myosin-II activity generates a dynamic steady state with continuous actin turnover in a minimal actin cortex. Journal of Cell Science, 132: JCS219899.
    [310]
    Spoerri P M, Strohmeyer N, Sun Z, Fässler R, Müller D J. 2020. Protease-activated receptor signalling initiates α(5)β(1)-integrin-mediated adhesion in non-haematopoietic cells. Nature Materials, 19: 218-226. doi: 10.1038/s41563-019-0580-4
    [311]
    Srichai M B, Zent R. 2010. Integrin Structure and Function//Cell-Extracellular Matrix Interactions in Cancer. New York: Springer
    [312]
    Stephanou A, Mylona E, Chaplain M, Tracqui P. 2008. A computational model of cell migration coupling the growth of focal adhesions with oscillatory cell protrusions. Journal of Theoretical Biology, 253: 701-16. doi: 10.1016/j.jtbi.2008.04.035
    [313]
    Strohmeyer N, Bharadwaj M, Costell M, Fässler R, Müller D J. 2017. Fibronectin-bound α5β1 integrins sense load and signal to reinforce adhesion in less than a second. Nature Materials, 16: 1262-1270. doi: 10.1038/nmat5023
    [314]
    Stutchbury B, Atherton P, Tsang R, Wang D Y, Ballestrem C. 2017. Distinct focal adhesion protein modules control different aspects of mechanotransduction. Journal of Cell Science, 130: 1612-1624.
    [315]
    Sun M, Spill F, Zaman Muhammad H. 2016. A Computational Model of YAP/TAZ Mechanosensing. Biophysical Journal, 110: 2540-2550. doi: 10.1016/j.bpj.2016.04.040
    [316]
    Sun Z, Costell M, Faessler R. 2019. Integrin activation by talin, kindlin and mechanical forces. Nature Cell Biology, 21: 25-31. doi: 10.1038/s41556-018-0234-9
    [317]
    Sun Z, Guo S S, Fässler R. 2016. Integrin-mediated mechanotransduction. Journal of Cell Biology, 215: 445-456. doi: 10.1083/jcb.201609037
    [318]
    Sun Z, Tseng H Y, Tan S, Senger F, Kurzawa L, et al. 2016. Kank2 activates talin, reduces force transduction across integrins and induces central adhesion formation. Nature Cell Biology, 18: 941-53. doi: 10.1038/ncb3402
    [319]
    Sung Haeng L, Roberto D. 2010. Regulation of actin cytoskeleton dynamics in cells. Molecules & Cells, 29: 311-325.
    [320]
    Sunyer R, Conte V, Escribano J, Elosegui-Artola A, Labernadie A, et al. 2016. Collective cell durotaxis emerges from long-range intercellular force transmission. Science, 353: 1157-1161. doi: 10.1126/science.aaf7119
    [321]
    Tadokoro S, Shattil S J, Eto K, Tai V, Liddington R C, et al. 2003. Talin binding to integrin β tails: a final common step in integrin activation. Science, 302: 103-106. doi: 10.1126/science.1086652
    [322]
    Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 126: 663-76. doi: 10.1016/j.cell.2006.07.024
    [323]
    Takeichi M. 1991. Cadherin cell adhesion receptors as a morphogenetic regulator. Science, 251: 1451-1455. doi: 10.1126/science.2006419
    [324]
    Takeichi M. 2014. Dynamic contacts: rearranging adherens junctions to drive epithelial remodelling. Nature Reviews Molecular Cell Biology, 15: 397-410. doi: 10.1038/nrm3802
    [325]
    Tam R Y, Smith L J, Shoichet M S. 2017. Engineering cellular microenvironments with photo- and enzymatically responsive hydrogels: toward biomimetic 3d cell culture models. Accounts of Chemical Research, 50: 703-713. doi: 10.1021/acs.accounts.6b00543
    [326]
    Tang K, Xin Y, Li K, Chen X, Tan Y. 2021. Cell cytoskeleton and stiffness are mechanical indicators of organotropism in breast cancer. Biology, 10: 259. doi: 10.3390/biology10040259
    [327]
    Tang S, Ma H, Tu H-C, Wang H-R, Lin P-C, Anseth K S. 2018. Adaptable fast relaxing boronate-based hydrogels for probing cell–matrix interactions. Advanced Science, 5: 1800638. doi: 10.1002/advs.201800638
    [328]
    Tang Y, Feinberg T, Keller E T, Li X-Y, Weiss S J. 2016. Snail/slug binding interactions with yap/taz control skeletal stem cell self-renewal and differentiation. Nature Cell Biology, 18: 917-929. doi: 10.1038/ncb3394
    [329]
    Tao J, Li Y, Vig D K, Sun S X. 2017. Cell mechanics: a dialogue. Reports on Progress in Physics, 80: 036601. doi: 10.1088/1361-6633/aa5282
    [330]
    Tashiro K, Monji A, I, Hayashi Y, Matsuda K, Tashiro N, Mitsuyama Y. 1999. An IKLLI-containing peptide derived from the laminin alpha1 chain mediating heparin-binding, cell adhesion, neurite outgrowth and proliferation, represents a binding site for integrin alpha3beta1 and heparan sulphate proteoglycan. Biochemical Journal, 340: 119-126. doi: 10.1042/bj3400119
    [331]
    Terekhova K, Pokutta S, Kee Y S, Li J, Tajkhorshid E, et al. 2019. Binding partner- and force-promoted changes in αE-catenin conformation probed by native cysteine labeling. Scientific Reports, 9: 15375. doi: 10.1038/s41598-019-51816-3
    [332]
    Tomakidi P, Schulz S, Proksch S, Weber W, Steinberg T. 2014. Focal adhesion kinase (FAK) perspectives in mechanobiology: implications for cell behaviour. Cell and Tissue Research, 357: 515-26. doi: 10.1007/s00441-014-1945-2
    [333]
    Totaro A, Castellan M, Battilana G, Zanconato F, Azzolin L, et al. 2017. YAP/TAZ link cell mechanics to notch signalling to control epidermal stem cell fate. Nature Communications, 8: 15206. doi: 10.1038/ncomms15206
    [334]
    Trujillo S, Vega S L, Song K H, San Felix A, Dalby M J, et al. 2020. Engineered full-length fibronectin-hyaluronic acid hydrogels for stem cell engineering. Advanced Healthcare Materials, 9: e2000989. doi: 10.1002/adhm.202000989
    [335]
    van der Flier A, Sonnenberg A. 2001. Function and interactions of integrins. Cell and Tissue Research, 305: 285-98. doi: 10.1007/s004410100417
    [336]
    Vazquez-Hidalgo E, Farris C M, Rowat A C, Katira P. 2022. Chemo-mechanical factors that limit cellular force generation. Frontiers in Physics, 10: 831776. doi: 10.3389/fphy.2022.831776
    [337]
    Velling T, Kusche-Gullberg M, Sejersen T, Gullberg D. 1999. cDNA cloning and chromosomal localization of human alpha(11) integrin. A collagen-binding, I domain-containing, beta(1)-associated integrin alpha-chain present in muscle tissues. Journal of Biomechanics, 274: 25735-25742.
    [338]
    Venhuizen J-H, Zegers M M. 2017. Making heads or tails of it: cell-cell adhesion in cellular and supracellular polarity in collective migration. Cold Spring Harbor Perspectives in Biology, 9: a027854. doi: 10.1101/cshperspect.a027854
    [339]
    Vining K H, Mooney D J. 2017. Mechanical forces direct stem cell behaviour in development and regeneration. Nature Reviews Molecular Cell Biology, 18: 728. doi: 10.1038/nrm.2017.108
    [340]
    Wahl A, Dinet C, Dillard P, Nassereddine A, Puech P H, et al. 2019. Biphasic mechanosensitivity of T cell receptor-mediated spreading of lymphocytes. Proceedings of the National Academy of Sciences of the United States of America, 116: 5908-5913. doi: 10.1073/pnas.1811516116
    [341]
    Walcott S, Kim D H, Wirtz D, Sun S X. 2011. Nucleation and decay initiation are the stiffness-sensitive phases of focal adhesion maturation. Biophysical Journal, 101: 2919-28. doi: 10.1016/j.bpj.2011.11.010
    [342]
    Wan W, Cheng B, Zhang C, Ma Y, Li A, et al. 2019. Synergistic effect of matrix stiffness and inflammatory factors on osteogenic differentiation of msc. Biophysical Journal, 117: 129-142. doi: 10.1016/j.bpj.2019.05.019
    [343]
    Wang J, Gao H. 2008. Clustering instability in adhesive contact between elastic solids via diffusive molecular bonds. Journal of the Mechanics and Physics of Solids, 56: 251-266. doi: 10.1016/j.jmps.2007.05.011
    [344]
    Wang X, Li S, Yan C, Liu P, Ding J. 2015. Fabrication of RGD micro/nanopattern and corresponding study of stem cell differentiation. Nano Letters, 15: 1457-67. doi: 10.1021/nl5049862
    [345]
    Wang Y-K, Chen C S. 2013. Cell adhesion and mechanical stimulation in the regulation of mesenchymal stem cell differentiation. Journal of Cellular and Molecular Medicine, 17: 823-832. doi: 10.1111/jcmm.12061
    [346]
    Wehrle-Haller B. 2007. Analysis of Integrin Dynamics by Fluorescence Recovery After Photobleaching. Methods in Molecular Biology, 370: 173-202. doi: 10.1007/978-1-59745-353-0_13
    [347]
    Welf E S, Johnson H E, Haugh J M. 2013. Bidirectional coupling between integrin-mediated signaling and actomyosin mechanics explains matrix-dependent intermittency of leading-edge motility. Molecular Biology of the Cell, 24: 3945-55. doi: 10.1091/mbc.e13-06-0311
    [348]
    Wells R G. 2008. The role of matrix stiffness in regulating cell behavior. Hepatology, 47: 1394-1400. doi: 10.1002/hep.22193
    [349]
    Winograd-Katz S E, Fässler R, Geiger B, Legate K R. 2014. The integrin adhesome: from genes and proteins to human disease. Nature Reviews Molecular Cell Biology, 15: 273-288. doi: 10.1038/nrm3769
    [350]
    Wolfenson H, Yang B, Sheetz M P. 2019. Steps in Mechanotransduction Pathways that Control Cell Morphology. Annual Review of Physiology, 81: 585-605. doi: 10.1146/annurev-physiol-021317-121245
    [351]
    Wu Y, Honig B, Ben-Shaul A. 2013. Theory and simulations of adhesion receptor dimerization on membrane surfaces. Biophysical Journal, 104: 1221-9. doi: 10.1016/j.bpj.2013.02.009
    [352]
    Wu Y, Jin X, Harrison O, Shapiro L, Honig B H, Ben-Shaul A. 2010. Cooperativity between trans and cis interactions in cadherin-mediated junction formation. Proceedings of the National Academy of Sciences of the United States of America, 107: 17592-7. doi: 10.1073/pnas.1011247107
    [353]
    Wu Y, Vendome J, Shapiro L, Ben-Shaul A, Honig B. 2011. Transforming binding affinities from three dimensions to two with application to cadherin clustering. Nature, 475: 510-3. doi: 10.1038/nature10183
    [354]
    Xia S, Kanchanawong P. 2017. Nanoscale mechanobiology of cell adhesions. Seminars in Cell & Developmental Biology, 71: 53-67.
    [355]
    Xia Y, Pfeifer C R, Cho S, Discher D E, Irianto J. 2018. Nuclear mechanosensing. Emerging Topics in Life Sciences, 2: 713-725. doi: 10.1042/ETLS20180051
    [356]
    Xiao P, Cuff L E, Lawton C D, Demali K A. 2010. Vinculin regulates cell-surface E-cadherin expression by binding to beta-catenin. Journal of Cell Science, 123: 567-577. doi: 10.1242/jcs.056432
    [357]
    Xie Z R, Chen J, Wu Y. 2014. A coarse-grained model for the simulations of biomolecular interactions in cellular environments. Journal of Chemical Physics, 140: 054112. doi: 10.1063/1.4863992
    [358]
    Xu K, Shuai Q, Li X, Zhang Y, Gao C, et al. 2016. Human VE-Cadherin Fusion Protein as an Artificial Extracellular Matrix Enhancing the Proliferation and Differentiation Functions of Endothelial Cell. Biomacromolecules, 17: 756-766. doi: 10.1021/acs.biomac.5b01467
    [359]
    Xu L, Gutbrod S R, Bonifas A P, Su Y, Sulkin M S, et al. 2014. 3D multifunctional integumentary membranes for spatiotemporal cardiac measurements and stimulation across the entire epicardium. Nature Communications, 5: 3329. doi: 10.1038/ncomms4329
    [360]
    Yamauchi M, Barker T H, Gibbons D L, Kurie J M. 2018. The fibrotic tumor stroma. Journal of Clinical Investigation, 128: 16-25. doi: 10.1172/JCI93554
    [361]
    Yang C, DelRio F W, Ma H, Killaars A R, Basta L P, et al. 2016. Spatially patterned matrix elasticity directs stem cell fate. Proceedings of the National Academy of Sciences of the United States of America, 113: E4439-45.
    [362]
    Yang C, Tibbitt M W, Basta L, Anseth K S. 2014. Mechanical memory and dosing influence stem cell fate. Nature Materials, 13: 645-652. doi: 10.1038/nmat3889
    [363]
    Yang Y, Jiang H. 2017. Shape and Dynamics of Adhesive Cells: Mechanical Response of Open Systems. Physical Review Letters, 118: 208102. doi: 10.1103/PhysRevLett.118.208102
    [364]
    Yang Y, Jiang H. 2018. Cellular volume regulation and substrate stiffness modulate the detachment dynamics of adherent cells. Journal of the Mechanics and Physics of Solids, 112: 594-618. doi: 10.1016/j.jmps.2017.10.009
    [365]
    Ye K, Wang X, Cao L, Li S, Li Z, et al. 2015. Matrix stiffness and nanoscale spatial organization of cell-adhesive ligands direct stem cell fate. Nano Letters, 15: 4720-9. doi: 10.1021/acs.nanolett.5b01619
    [366]
    Yeung T, Georges P C, Flanagan L A, Marg B, Ortiz M, et al. 2005. Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Motility and the Cytoskeleton, 60: 24-34. doi: 10.1002/cm.20041
    [367]
    Yi B, Xu Q, Liu W. 2021. An overview of substrate stiffness guided cellular response and its applications in tissue regeneration. Bioactive Materials, 15: 82-102.
    [368]
    Yonemura S, Wada Y T, Nagafuchi A, Shibata M. 2010. alpha-Catenin as a tension transducer that induces adherens junction development. Nature Cell Biology, 12: 533-542. doi: 10.1038/ncb2055
    [369]
    Yu J, Huang J, Jansen J A, Xiong C, Walboomers X F. 2017. Mechanochemical mechanism of integrin clustering modulated by nanoscale ligand spacing and rigidity of extracellular substrates. Journal of the Mechanical Behavior of Biomedical Materials, 72: 29-37. doi: 10.1016/j.jmbbm.2017.04.018
    [370]
    Yue X-S, Murakami Y, Tamai T, Nagaoka M, Cho C-S, et al. 2010. A fusion protein N-cadherin-Fc as an artificial extracellular matrix surface for maintenance of stem cell features. Biomaterials, 31: 5287-5296. doi: 10.1016/j.biomaterials.2010.03.035
    [371]
    Yuka M, Naoko I, Koji N, Nagatoki K, Hiroshi H, Shigenobu Y. 2006. Actomyosin tension is required for correct recruitment of adherens junction components and zonula occludens formation. Experimental Cell Research, 312: 1637-1650. doi: 10.1016/j.yexcr.2006.01.031
    [372]
    Zamir E, Katz M, Posen Y, Erez N, Yamada K M, et al. 2000. Dynamics and segregation of cell-matrix adhesions in cultured fibroblasts. Nature Cell Biology, 2: 191-6. doi: 10.1038/35008607
    [373]
    Zeeshan R, Mutahir Z. 2017. Cancer metastasis - tricks of the trade. Bosnian Journal of Basic Medical Sciences, 17: 172-182.
    [374]
    Zhang C, Zhu H, Ren X, Gao B, Cheng B, et al. 2021. Mechanics-driven nuclear localization of YAP can be reversed by N-cadherin ligation in mesenchymal stem cells. Nature Communications, 12: 6229. doi: 10.1038/s41467-021-26454-x
    [375]
    Zhang G, Ma L, Bai L, Li M, Guo T, et al. 2021. Inflammatory microenvironment-targeted nanotherapies. Journal of Controlled Release, 334: 114-126. doi: 10.1016/j.jconrel.2021.04.018
    [376]
    Zhang S, Teng X, Toyama Y, Saunders T E. 2020. Periodic oscillations of myosin-ii mechanically proofread cell-cell connections to ensure robust formation of the cardiac vessel. Current Biology, 30: 3364-3377.e4. doi: 10.1016/j.cub.2020.06.041
    [377]
    Zhang Y, Mao H, Qian M, Hu F, Cao L, et al. 2016. Surface modification with E-cadherin fusion protein for mesenchymal stem cell culture. Journal of Materials Chemistry B, 4: 4267-4277. doi: 10.1039/C6TB00765A
    [378]
    Zhang Y, Qin Z, Qu Z, Ge M, Yang J. 2020. Cadherin-based biomaterials: Inducing stem cell fate towards tissue construction and therapeutics. Progress in Natural Science:Materials International, 30: 597-608. doi: 10.1016/j.pnsc.2020.09.001
    [379]
    Zheng C, Chen J, Liu S, Jin Y. 2019. Stem cell-based bone and dental regeneration: a view of microenvironmental modulation. International Journal of Oral Science, 11: 23. doi: 10.1038/s41368-019-0060-3
    [380]
    Zhong C, Chrzanowska-Wodnicka M, Brown J, Shaub A, Belkin A M, Burridge K. 1998. Rho-mediated contractility exposes a cryptic site in fibronectin and induces fibronectin matrix assembly. Journal of Cell Biology, 141: 539-51. doi: 10.1083/jcb.141.2.539
    [381]
    Zhou J, Lee P L, Tsai C S, Lee C I, Yang T L, et al. 2012. Force-specific activation of Smad1/5 regulates vascular endothelial cell cycle progression in response to disturbed flow. Proceedings of the National Academy of Sciences of the United States of America, 109: 7770-5. doi: 10.1073/pnas.1205476109
    [382]
    Zhou J, Wang K-C, Wu W, Subramaniam S, Shyy J Y J, et al. 2011. MicroRNA-21 targets peroxisome proliferators-activated receptor-alpha in an autoregulatory loop to modulate flow-induced endothelial inflammation. Proceedings of the National Academy of Sciences of the United States of America, 108: 10355-10360. doi: 10.1073/pnas.1107052108
    [383]
    Zhu M, Lin S, Sun Y, Feng Q, Li G, Bian L. 2016. Hydrogels functionalized with N-cadherin mimetic peptide enhance osteogenesis of hMSCs by emulating the osteogenic niche. Biomaterials, 77: 44-52. doi: 10.1016/j.biomaterials.2015.10.072
    [384]
    Zuidema A, Wang W, Sonnenberg A. 2020. Crosstalk between Cell Adhesion Complexes in Regulation of Mechanotransduction. Bioessays, 42: 2000119. doi: 10.1002/bies.202000119
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(58)  / Tables(2)

    Article Metrics

    Article views (3979) PDF downloads(1237) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return