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:

Cardiac fibroblasts and mechanosensation in heart development, health and disease

Abstract

The term ‘mechanosensation’ describes the capacity of cells to translate mechanical stimuli into the coordinated regulation of intracellular signals, cellular function, gene expression and epigenetic programming. This capacity is related not only to the sensitivity of the cells to tissue motion, but also to the decryption of tissue geometric arrangement and mechanical properties. The cardiac stroma, composed of fibroblasts, has been historically considered a mechanically passive component of the heart. However, the latest research suggests that the mechanical functions of these cells are an active and necessary component of the developmental biology programme of the heart that is involved in myocardial growth and homeostasis, and a crucial determinant of cardiac repair and disease. In this Review, we discuss the general concept of cell mechanosensation and force generation as potent regulators in heart development and pathology, and describe the integration of mechanical and biohumoral pathways predisposing the heart to fibrosis and failure. Next, we address the use of 3D culture systems to integrate tissue mechanics to mimic cardiac remodelling. Finally, we highlight the potential of mechanotherapeutic strategies, including pharmacological treatment and device-mediated left ventricular unloading, to reverse remodelling in the failing heart.

Key points

  • The sensing of mechanical tissue properties is a process related to cell differentiation, maturation and pathology in multicellular organs such as the heart.

  • Remodelling of the cardiac extracellular matrix, which occurs as a consequence of a pathological stimulus, induces changes in the mechanical properties of the myocardium.

  • Variations in the mechanical properties of the myocardium are related to the activation of pro-fibrotic cells (so-called myofibroblasts).

  • Mechanical cues can potentiate pro-fibrotic humoral signalling.

  • The identification of molecular pathways involved in mechanosensation of myofibroblasts facilitates the identification of therapeutic targets that can reverse mechanically induced pathological activation.

  • The possibility that interfering with mechanical cues in vivo might result in cardiac regeneration opens new therapeutic avenues in cardioprotection.

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

Access options

Buy this article

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

Fig. 1: The mechanical framework of myocardial cell maturation.
Fig. 2: Cellular force transmission and mechanisms of mechanotransduction.
Fig. 3: Effects of ECM remodelling owing to ageing or compensatory mechanisms on local variations in stiffness.
Fig. 4: Mechanical ‘memory’ underlies chronic pathological cellular phenotypes.
Fig. 5: Integration of diagnostic and experimental pipelines for the evaluation of cardiac mechanotherapeutic approaches.

Similar content being viewed by others

References

  1. Pinto, A. R. et al. Revisiting cardiac cellular composition. Circ. Res. 118, 400–409 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Litvinukova, M. et al. Cells of the adult human heart. Nature 588, 466–472 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Herum, K. M., Choppe, J., Kumar, A., Engler, A. J. & McCulloch, A. D. Mechanical regulation of cardiac fibroblast profibrotic phenotypes. Mol. Biol. Cell 28, 1871–1882 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Berry, M. F. et al. Mesenchymal stem cell injection after myocardial infarction improves myocardial compliance. Am. J. Physiol. Heart Circ. Physiol. 290, H2196–H2203 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Van Linthout, S., Miteva, K. & Tschope, C. Crosstalk between fibroblasts and inflammatory cells. Cardiovasc. Res. 102, 258–269 (2014).

    Article  PubMed  Google Scholar 

  8. Steffens, S. et al. Stimulating pro-reparative immune responses to prevent adverse cardiac remodelling: consensus document from the joint 2019 meeting of the ESC Working Groups of Cellular Biology of the Heart and Myocardial Function. Cardiovasc. Res. 116, 1850–1862 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. van Putten, S., Shafieyan, Y. & Hinz, B. Mechanical control of cardiac myofibroblasts. J. Mol. Cell. Cardiol. 93, 133–142 (2016).

    Article  PubMed  Google Scholar 

  10. Yu, J. et al. Topological arrangement of cardiac fibroblasts regulates cellular plasticity. Circ. Res. 123, 73–85 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bracco Gartner, T. C. L. et al. Advanced in vitro modeling to study the paradox of mechanically induced cardiac fibrosis. Tissue Eng. Part. C. Methods 27, 100–114 (2021).

    Article  PubMed  Google Scholar 

  12. Majkut, S., Dingal, P. C. & Discher, D. E. Stress sensitivity and mechanotransduction during heart development. Curr. Biol. 24, R495–R501 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Andres-Delgado, L. & Mercader, N. Interplay between cardiac function and heart development. Biochim. Biophys. Acta 1863, 1707–1716 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Happe, C. L. & Engler, A. J. Mechanical forces reshape differentiation cues that guide cardiomyogenesis. Circ. Res. 118, 296–310 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Courchaine, K., Rykiel, G. & Rugonyi, S. Influence of blood flow on cardiac development. Prog. Biophys. Mol. Biol. 137, 95–110 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Tallquist, M. D. Developmental pathways of cardiac fibroblasts. Cold Spring Harb. Perspect. Biol. 12, a037184 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Majkut, S. et al. Heart-specific stiffening in early embryos parallels matrix and myosin expression to optimize beating. Curr. Biol. 23, 2434–2439 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chiou, K. K. et al. Mechanical signaling coordinates the embryonic heartbeat. Proc. Natl Acad. Sci. USA 113, 8939–8944 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Matrone, G., Tucker, C. S. & Denvir, M. A. Cardiomyocyte proliferation in zebrafish and mammals: lessons for human disease. Cell. Mol. Life Sci. 74, 1367–1378 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Eulalio, A. et al. Functional screening identifies miRNAs inducing cardiac regeneration. Nature 492, 376–381 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Gabisonia, K. et al. MicroRNA therapy stimulates uncontrolled cardiac repair after myocardial infarction in pigs. Nature 569, 418–422 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kennedy-Lydon, T. & Rosenthal, N. Cardiac regeneration: all work and no repair? Sci. Transl Med. 9, eaad9019 (2017).

    Article  PubMed  Google Scholar 

  23. Garcia-Gonzalez, C. & Morrison, J. I. Cardiac regeneration in non-mammalian vertebrates. Exp. Cell Res. 321, 58–63 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Poss, K. D., Wilson, L. G. & Keating, M. T. Heart regeneration in zebrafish. Science 298, 2188–2190 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Sanz-Morejon, A. & Mercader, N. Recent insights into zebrafish cardiac regeneration. Curr. Opin. Genet. Dev. 64, 37–43 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Yu, J. K. et al. Cardiac regeneration following cryoinjury in the adult zebrafish targets a maturation-specific biomechanical remodeling program. Sci. Rep. 8, 15661 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Sanchez-Iranzo, H. et al. Transient fibrosis resolves via fibroblast inactivation in the regenerating zebrafish heart. Proc. Natl Acad. Sci. USA 115, 4188–4193 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ito, K. et al. Differential reparative phenotypes between zebrafish and medaka after cardiac injury. Dev. Dyn. 243, 1106–1115 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Porrello, E. R. et al. Transient regenerative potential of the neonatal mouse heart. Science 331, 1078–1080 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chen, W. C. et al. Decellularized zebrafish cardiac extracellular matrix induces mammalian heart regeneration. Sci. Adv. 2, e1600844 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Wang, Z. et al. Decellularized neonatal cardiac extracellular matrix prevents widespread ventricular remodeling in adult mammals after myocardial infarction. Acta Biomater. 87, 140–151 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Missinato, M. A., Tobita, K., Romano, N., Carroll, J. A. & Tsang, M. Extracellular component hyaluronic acid and its receptor Hmmr are required for epicardial EMT during heart regeneration. Cardiovasc. Res. 107, 487–498 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wang, J., Karra, R., Dickson, A. L. & Poss, K. D. Fibronectin is deposited by injury-activated epicardial cells and is necessary for zebrafish heart regeneration. Dev. Biol. 382, 427–435 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Kuhn, B. et al. Periostin induces proliferation of differentiated cardiomyocytes and promotes cardiac repair. Nat. Med. 13, 962–969 (2007).

    Article  PubMed  Google Scholar 

  35. Garcia-Puig, A. et al. Proteomics analysis of extracellular matrix remodeling during zebrafish heart regeneration. Mol. Cell Proteom. 18, 1745–1755 (2019).

    Article  CAS  Google Scholar 

  36. Notari, M. et al. The local microenvironment limits the regenerative potential of the mouse neonatal heart. Sci. Adv. 4, eaao5553 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yahalom-Ronen, Y., Rajchman, D., Sarig, R., Geiger, B. & Tzahor, E. Reduced matrix rigidity promotes neonatal cardiomyocyte dedifferentiation, proliferation and clonal expansion. eLife 4, e07455 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Wang, X. et al. Microenvironment stiffness requires decellularized cardiac extracellular matrix to promote heart regeneration in the neonatal mouse heart. Acta Biomater. 113, 380–392 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bassat, E. et al. The extracellular matrix protein agrin promotes heart regeneration in mice. Nature 547, 179–184 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. van der Pol, A. & Bouten, C. V. C. A brief history in cardiac regeneration, and how the extra cellular matrix may turn the tide. Front. Cardiovasc. Med. 8, 682342 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Gaetani, R. et al. When stiffness matters: mechanosensing in heart development and disease. Front. Cell Dev. Biol. 8, 334 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Perestrelo, A. R. et al. Multiscale analysis of extracellular matrix remodeling in the failing heart. Circ. Res. 128, 24–38 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. Civitarese, R. A. et al. The α11 integrin mediates fibroblast-extracellular matrix-cardiomyocyte interactions in health and disease. Am. J. Physiol. Heart Circ. Physiol. 311, H96–H106 (2016).

    Article  PubMed  Google Scholar 

  44. Gullberg, D. et al. Analysis of alpha 1 beta 1, alpha 2 beta 1 and alpha 3 beta 1 integrins in cell–collagen interactions: identification of conformation dependent alpha 1 beta 1 binding sites in collagen type I. EMBO J. 11, 3865–3873 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Balasubramanian, S. et al. β3 Integrin in cardiac fibroblast is critical for extracellular matrix accumulation during pressure overload hypertrophy in mouse. PLoS ONE 7, e45076 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Schiller, H. B. & Fassler, R. Mechanosensitivity and compositional dynamics of cell-matrix adhesions. EMBO Rep. 14, 509–519 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sit, B., Gutmann, D. & Iskratsch, T. Costameres, dense plaques and podosomes: the cell matrix adhesions in cardiovascular mechanosensing. J. Muscle Res. Cell Motil. 40, 197–209 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen, Y., Lee, H., Tong, H., Schwartz, M. & Zhu, C. Force regulated conformational change of integrin αVβ3. Matrix Biol. 60-61, 70–85 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Shemesh, T., Geiger, B., Bershadsky, A. D. & Kozlov, M. M. Focal adhesions as mechanosensors: a physical mechanism. Proc. Natl Acad. Sci. USA 102, 12383–12388 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kong, F., Garcia, A. J., Mould, A. P., Humphries, M. J. & Zhu, C. Demonstration of catch bonds between an integrin and its ligand. J. Cell Biol. 185, 1275–1284 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kaushik, G. et al. Vinculin network-mediated cytoskeletal remodeling regulates contractile function in the aging heart. Sci. Transl Med. 7, 292ra299 (2015).

    Article  Google Scholar 

  52. Zhang, J. et al. Targeted inhibition of focal adhesion kinase attenuates cardiac fibrosis and preserves heart function in adverse cardiac remodeling. Sci. Rep. 7, 43146 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Manso, A. M. et al. Loss of mouse cardiomyocyte talin-1 and talin-2 leads to β-1 integrin reduction, costameric instability, and dilated cardiomyopathy. Proc. Natl Acad. Sci. USA 114, E6250–E6259 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Civitarese, R. A., Kapus, A., McCulloch, C. A. & Connelly, K. A. Role of integrins in mediating cardiac fibroblast-cardiomyocyte cross talk: a dynamic relationship in cardiac biology and pathophysiology. Basic. Res. Cardiol. 112, 6 (2017).

    Article  PubMed  Google Scholar 

  55. Hinz, B., Pittet, P., Smith-Clerc, J., Chaponnier, C. & Meister, J. J. Myofibroblast development is characterized by specific cell–cell adherens junctions. Mol. Biol. Cell 15, 4310–4320 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Schroer, A. K. & Merryman, W. D. Mechanobiology of myofibroblast adhesion in fibrotic cardiac disease. J. Cell Sci. 128, 1865–1875 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Rakshit, S., Zhang, Y., Manibog, K., Shafraz, O. & Sivasankar, S. Ideal, catch, and slip bonds in cadherin adhesion. Proc. Natl Acad. Sci. USA 109, 18815–18820 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Yao, M. et al. Force-dependent conformational switch of α-catenin controls vinculin binding. Nat. Commun. 5, 4525 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Buckley, C. D. et al. Cell adhesion. The minimal cadherin-catenin complex binds to actin filaments under force. Science 346, 1254211 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Vermij, S. H., Abriel, H. & van Veen, T. A. Refining the molecular organization of the cardiac intercalated disc. Cardiovasc. Res. 113, 259–275 (2017).

    Article  CAS  PubMed  Google Scholar 

  61. Baddam, S. R. et al. The desmosomal cadherin desmoglein-2 experiences mechanical tension as demonstrated by a FRET-based tension biosensor expressed in living cells. Cells 7, 66 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Samarel, A. M. Costameres, focal adhesions, and cardiomyocyte mechanotransduction. Am. J. Physiol. Heart Circ. Physiol. 289, H2291–H2301 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Galie, P. A., Khalid, N., Carnahan, K. E., Westfall, M. V. & Stegemann, J. P. Substrate stiffness affects sarcomere and costamere structure and electrophysiological function of isolated adult cardiomyocytes. Cardiovasc. Pathol. 22, 219–227 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Fancher, I. S. in Cellular Mechanotransduction Mechanisms in Cardiovascular and Fibrotic Diseases Ch. 2 (ed. Fang, Y.) 47–95 (Academic, 2021).

  65. Alonso-Carbajo, L. et al. Muscling in on TRP channels in vascular smooth muscle cells and cardiomyocytes. Cell Calcium 66, 48–61 (2017).

    Article  CAS  PubMed  Google Scholar 

  66. Jakob, D. et al. Piezo1 and BKCa channels in human atrial fibroblasts: interplay and remodelling in atrial fibrillation. J. Mol. Cell. Cardiol. 158, 49–62 (2021).

    Article  CAS  PubMed  Google Scholar 

  67. Adapala, R. K. et al. TRPV4 channels mediate cardiac fibroblast differentiation by integrating mechanical and soluble signals. J. Mol. Cell. Cardiol. 54, 45–52 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Harada, M. et al. Transient receptor potential canonical-3 channel-dependent fibroblast regulation in atrial fibrillation. Circulation 126, 2051–2064 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Du, J. et al. TRPM7-mediated Ca2+ signals confer fibrogenesis in human atrial fibrillation. Circ. Res. 106, 992–1003 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Davis, J., Burr, A. R., Davis, G. F., Birnbaumer, L. & Molkentin, J. D. A TRPC6-dependent pathway for myofibroblast transdifferentiation and wound healing in vivo. Dev. Cell 23, 705–715 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Dupont, S. & Wickstrom, S. A. Mechanical regulation of chromatin and transcription. Nat. Rev. Genet. 23, 624–643 (2022).

    Article  CAS  PubMed  Google Scholar 

  72. Roper, J. C. et al. The major β-catenin/E-cadherin junctional binding site is a primary molecular mechano-transductor of differentiation in vivo. eLife 7, e33381 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Zhao, X. H. et al. Force activates smooth muscle α-actin promoter activity through the Rho signaling pathway. J. Cell Sci. 120, 1801–1809 (2007).

    Article  CAS  PubMed  Google Scholar 

  74. Ho, C. Y., Jaalouk, D. E., Vartiainen, M. K. & Lammerding, J. Lamin A/C and emerin regulate MKL1-SRF activity by modulating actin dynamics. Nature 497, 507–511 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Arsenovic, P. T. et al. Nesprin-2G, a component of the nuclear LINC complex, is subject to myosin-dependent tension. Biophys. J. 110, 34–43 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Elosegui-Artola, A. et al. Force triggers YAP nuclear entry by regulating transport across nuclear pores. Cell 171, 1397–1410.e14 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Nava, M. M. et al. Heterochromatin-driven nuclear softening protects the genome against mechanical stress-induced damage. Cell 181, 800–817 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Tajik, A. et al. Transcription upregulation via force-induced direct stretching of chromatin. Nat. Mater. 15, 1287–1296 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Sun, J., Chen, J., Mohagheghian, E. & Wang, N. Force-induced gene up-regulation does not follow the weak power law but depends on H3K9 demethylation. Sci. Adv. 6, eaay9095 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  84. Schuller, A. P. et al. The cellular environment shapes the nuclear pore complex architecture. Nature 598, 667–671 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Zimmerli, C. E. et al. Nuclear pores dilate and constrict in cellulo. Science 374, eabd9776 (2021).

    Article  CAS  PubMed  Google Scholar 

  86. Andreu, I. et al. Mechanical force application to the nucleus regulates nucleocytoplasmic transport. Nat. Cell Biol. 24, 896–905 (2022).

    Article  CAS  PubMed  Google Scholar 

  87. Piccolo, S., Dupont, S. & Cordenonsi, M. The biology of YAP/TAZ: hippo signaling and beyond. Physiol. Rev. 94, 1287–1312 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Zhang, K. et al. Mechanical signals regulate and activate SNAIL1 protein to control the fibrogenic response of cancer-associated fibroblasts. J. Cell. Sci. 129, 1989–2002 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Infante, E. et al. The mechanical stability of proteins regulates their translocation rate into the cell nucleus. Nat. Phys. 15, 973–981 (2019).

    Article  CAS  Google Scholar 

  91. Ugolini, G. S. et al. On-chip assessment of human primary cardiac fibroblasts proliferative responses to uniaxial cyclic mechanical strain. Biotechnol. Bioeng. 113, 859–869 (2016).

    Article  CAS  PubMed  Google Scholar 

  92. Niu, L. et al. Matrix stiffness controls cardiac fibroblast activation through regulating YAP via AT1 R. J. Cell. Physiol. 235, 8345–8357 (2020).

    Article  CAS  PubMed  Google Scholar 

  93. Heallen, T. et al. Hippo signaling impedes adult heart regeneration. Development 140, 4683–4690 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Xin, M. et al. Hippo pathway effector Yap promotes cardiac regeneration. Proc. Natl Acad. Sci. USA 110, 13839–13844 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Flinn, M. A., Link, B. A. & O’Meara, C. C. Upstream regulation of the Hippo-Yap pathway in cardiomyocyte regeneration. Semin. Cell Dev. Biol. 100, 11–19 (2020).

    Article  PubMed  Google Scholar 

  96. Weichhart, T., Hengstschlager, M. & Linke, M. Regulation of innate immune cell function by mTOR. Nat. Rev. Immunol. 15, 599–614 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Castillo, E. A., Lane, K. V. & Pruitt, B. L. Micromechanobiology: focusing on the cardiac cell-substrate interface. Annu. Rev. Biomed. Eng. 22, 257–284 (2020).

    Article  CAS  PubMed  Google Scholar 

  98. Ieda, M. et al. Cardiac fibroblasts regulate myocardial proliferation through β1 integrin signaling. Dev. Cell 16, 233–244 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Wu, C. C., Jeratsch, S., Graumann, J. & Stainier, D. Y. R. Modulation of mammalian cardiomyocyte cytokinesis by the extracellular matrix. Circ. Res. 127, 896–907 (2020).

    Article  CAS  PubMed  Google Scholar 

  100. Heallen, T. et al. Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science 332, 458–461 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Halder, G. & Johnson, R. L. Hippo signaling: growth control and beyond. Development 138, 9–22 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Del Re, D. P. et al. Yes-associated protein isoform 1 (Yap1) promotes cardiomyocyte survival and growth to protect against myocardial ischemic injury. J. Biol. Chem. 288, 3977–3988 (2013).

    Article  PubMed  Google Scholar 

  103. Hou, N. et al. Activation of Yap1/Taz signaling in ischemic heart disease and dilated cardiomyopathy. Exp. Mol. Pathol. 103, 267–275 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Monroe, T. O. et al. YAP partially reprograms chromatin accessibility to directly induce adult cardiogenesis in vivo. Dev. Cell 48, 765–779.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Xiao, Y. et al. Hippo pathway deletion in adult resting cardiac fibroblasts initiates a cell state transition with spontaneous and self-sustaining fibrosis. Genes Dev. 33, 1491–1505 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Mosqueira, D. et al. Hippo pathway effectors control cardiac progenitor cell fate by acting as dynamic sensors of substrate mechanics and nanostructure. ACS Nano 8, 2033–2047 (2014).

    Article  CAS  PubMed  Google Scholar 

  107. Morikawa, Y. et al. Actin cytoskeletal remodeling with protrusion formation is essential for heart regeneration in Hippo-deficient mice. Sci. Signal. 8, ra41 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Morikawa, Y., Heallen, T., Leach, J., Xiao, Y. & Martin, J. F. Dystrophin-glycoprotein complex sequesters Yap to inhibit cardiomyocyte proliferation. Nature 547, 227–231 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Aharonov, A. et al. ERBB2 drives YAP activation and EMT-like processes during cardiac regeneration. Nat. Cell Biol. 22, 1346–1356 (2020).

    Article  CAS  PubMed  Google Scholar 

  110. Frangogiannis, N. G. Cardiac fibrosis. Cardiovasc. Res. 117, 1450–1488 (2020).

    Article  PubMed Central  Google Scholar 

  111. Zile, M. R. et al. Myocardial stiffness in patients with heart failure and a preserved ejection fraction: contributions of collagen and titin. Circulation 131, 1247–1259 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Thomas, D. P., Cotter, T. A., Li, X., McCormick, R. J. & Gosselin, L. E. Exercise training attenuates aging-associated increases in collagen and collagen crosslinking of the left but not the right ventricle in the rat. Eur. J. Appl. Physiol. 85, 164–169 (2001).

    Article  CAS  PubMed  Google Scholar 

  113. Asif, M. et al. An advanced glycation endproduct cross-link breaker can reverse age-related increases in myocardial stiffness. Proc. Natl Acad. Sci. USA 97, 2809–2813 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Hutchinson, K. R., Lord, C. K., West, T. A. & Stewart, J. A. Jr Cardiac fibroblast-dependent extracellular matrix accumulation is associated with diastolic stiffness in type 2 diabetes. PLoS ONE 8, e72080 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Angelini, A., Trial, J., Ortiz-Urbina, J. & Cieslik, K. A. Mechanosensing dysregulation in the fibroblast: a hallmark of the aging heart. Ageing Res. Rev. 63, 101150 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Villemain, O. et al. Myocardial stiffness evaluation using noninvasive shear wave imaging in healthy and hypertrophic cardiomyopathic adults. JACC Cardiovasc. Imaging 12, 1135–1145 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Engler, A. J. et al. Embryonic cardiomyocytes beat best on a matrix with heart-like elasticity: scar-like rigidity inhibits beating. J. Cell Sci. 121, 3794–3802 (2008).

    Article  CAS  PubMed  Google Scholar 

  118. Pandey, P. et al. Cardiomyocytes sense matrix rigidity through a combination of muscle and non-muscle myosin contractions. Dev. Cell 44, 326–336.e3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Nishimura, M. et al. A dual role for integrin-linked kinase and β1-integrin in modulating cardiac aging. Aging Cell 13, 431–440 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Nawata, J. et al. Differential expression of α1, α3 and α5 integrin subunits in acute and chronic stages of myocardial infarction in rats. Cardiovasc. Res. 43, 371–381 (1999).

    Article  CAS  PubMed  Google Scholar 

  121. Hein, S., Kostin, S., Heling, A., Maeno, Y. & Schaper, J. The role of the cytoskeleton in heart failure. Cardiovasc. Res. 45, 273–278 (2000).

    Article  CAS  PubMed  Google Scholar 

  122. Heling, A. et al. Increased expression of cytoskeletal, linkage, and extracellular proteins in failing human myocardium. Circ. Res. 86, 846–853 (2000).

    Article  CAS  PubMed  Google Scholar 

  123. Sessions, A. O. & Engler, A. J. Mechanical regulation of cardiac aging in model systems. Circ. Res. 118, 1553–1562 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Neff, L. S. & Bradshaw, A. D. Cross your heart? Collagen cross-links in cardiac health and disease. Cell Signal. 79, 109889 (2021).

    Article  CAS  PubMed  Google Scholar 

  125. Al-U’datt, D., Allen, B. G. & Nattel, S. Role of the lysyl oxidase enzyme family in cardiac function and disease. Cardiovasc. Res. 115, 1820–1837 (2019).

    PubMed  Google Scholar 

  126. Gonzalez-Santamaria, J. et al. Matrix cross-linking lysyl oxidases are induced in response to myocardial infarction and promote cardiac dysfunction. Cardiovasc. Res. 109, 67–78 (2016).

    Article  CAS  PubMed  Google Scholar 

  127. Grilo, G. A. et al. Age- and sex-dependent differences in extracellular matrix metabolism associate with cardiac functional and structural changes. J. Mol. Cell. Cardiol. 139, 62–74 (2020).

    Article  CAS  PubMed  Google Scholar 

  128. Yang, J. et al. Targeting LOXL2 for cardiac interstitial fibrosis and heart failure treatment. Nat. Commun. 7, 13710 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Bhana, B. et al. Influence of substrate stiffness on the phenotype of heart cells. Biotechnol. Bioeng. 105, 1148–1160 (2010).

    CAS  PubMed  Google Scholar 

  130. Forte, G. et al. Substrate stiffness modulates gene expression and phenotype in neonatal cardiomyocytes in vitro. Tissue Eng. Part A 18, 1837–1848 (2012).

    Article  CAS  PubMed  Google Scholar 

  131. McCain, M. L., Yuan, H., Pasqualini, F. S., Campbell, P. H. & Parker, K. K. Matrix elasticity regulates the optimal cardiac myocyte shape for contractility. Am. J. Physiol. Heart Circ. Physiol. 306, H1525–H1539 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Jacot, J. G., McCulloch, A. D. & Omens, J. H. Substrate stiffness affects the functional maturation of neonatal rat ventricular myocytes. Biophys. J. 95, 3479–3487 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Ribeiro, A. J. et al. Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness. Proc. Natl Acad. Sci. USA 112, 12705–12710 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Katz, A. M. & Rolett, E. L. Heart failure: when form fails to follow function. Eur. Heart J. 37, 449–454 (2016).

    Article  PubMed  Google Scholar 

  135. Fomovsky, G. M., Rouillard, A. D. & Holmes, J. W. Regional mechanics determine collagen fiber structure in healing myocardial infarcts. J. Mol. Cell. Cardiol. 52, 1083–1090 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Garoffolo, G. et al. Reduction of cardiac fibrosis by interference with YAP-dependent transactivation. Circ. Res. 131, 239–257 (2022).

    Article  CAS  PubMed  Google Scholar 

  137. Khalil, H. et al. Fibroblast-specific TGF-β–Smad2/3 signaling underlies cardiac fibrosis. J. Clin. Invest. 127, 3770–3783 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  138. Dang, S. et al. Blockade of β-adrenergic signaling suppresses inflammasome and alleviates cardiac fibrosis. Ann. Transl Med. 8, 127 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Miteva, K. et al. Human endomyocardial biopsy specimen-derived stromal cells modulate angiotensin II-induced cardiac remodeling. Stem Cell Transl Med. 5, 1707–1718 (2016).

    Article  CAS  Google Scholar 

  140. Tschope, C. et al. Modulation of the acute defence reaction by eplerenone prevents cardiac disease progression in viral myocarditis. ESC Heart Fail. 7, 2838–2852 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  141. Xia, Y. et al. Endogenous thrombospondin 1 protects the pressure-overloaded myocardium by modulating fibroblast phenotype and matrix metabolism. Hypertension 58, 902–911 (2011).

    Article  CAS  PubMed  Google Scholar 

  142. Lorenzen, J. M. et al. Osteopontin is indispensible for AP1-mediated angiotensin II-related miR-21 transcription during cardiac fibrosis. Eur. Heart J. 36, 2184–2196 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Van Linthout, S. & Tschope, C. The quest for antiinflammatory and immunomodulatory strategies in heart failure. Clin. Pharmacol. Ther. 106, 1198–1208 (2019).

    Article  PubMed  Google Scholar 

  144. Wu, Y. et al. S100a8/a9 released by CD11b+Gr1+ neutrophils activates cardiac fibroblasts to initiate angiotensin II-Induced cardiac inflammation and injury. Hypertension 63, 1241–1250 (2014).

    Article  CAS  PubMed  Google Scholar 

  145. Lindner, D. et al. Cardiac fibroblasts support cardiac inflammation in heart failure. Basic. Res. Cardiol. 109, 428 (2014).

    Article  PubMed  Google Scholar 

  146. Pappritz, K. et al. Cardiac (myo)fibroblasts modulate the migration of monocyte subsets. Sci. Rep. 8, 5575 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  147. Matz, I., Pappritz, K., Springer, J. & Van Linthout, S. Left ventricle- and skeletal muscle-derived fibroblasts exhibit a differential inflammatory and metabolic responsiveness to interleukin-6. Front. Immunol. 13, 947267 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Sandanger, O. et al. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury. Cardiovasc. Res. 99, 164–174 (2013).

    Article  CAS  PubMed  Google Scholar 

  149. Mia, M. et al. Loss of Yap/Taz in cardiac fibroblasts attenuates adverse remodeling and improves cardiac function. Cardiovasc. Res. 118, 1785–1804 (2022).

    Article  CAS  PubMed  Google Scholar 

  150. Wong, V. W. et al. Mechanical force prolongs acute inflammation via T-cell-dependent pathways during scar formation. FASEB J. 25, 4498–4510 (2011).

    Article  CAS  PubMed  Google Scholar 

  151. Li, C. et al. Mineralocorticoid receptor deficiency in T cells attenuates pressure overload-induced cardiac hypertrophy and dysfunction through modulating T-cell activation. Hypertension 70, 137–147 (2017).

    Article  CAS  PubMed  Google Scholar 

  152. Sun, X. J. et al. Deletion of interleukin 1 receptor-associated kinase 1 (Irak1) improves glucose tolerance primarily by increasing insulin sensitivity in skeletal muscle. J. Biol. Chem. 292, 12339–12350 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Huang, H. W., Fang, X. X., Wang, X. Q., Peng, Y. P. & Qiu, Y. H. Regulation of differentiation and function of helper T cells by lymphocyte-derived catecholamines via α1- and β2-adrenoceptors. Neuroimmunomodulation 22, 138–151 (2015).

    Article  CAS  PubMed  Google Scholar 

  154. Woodall, M. C., Woodall, B. P., Gao, E., Yuan, A. & Koch, W. J. Cardiac fibroblast GRK2 deletion enhances contractility and remodeling following ischemia/reperfusion injury. Circ. Res. 119, 1116–1127 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Travers, J. G. et al. Pharmacological and activated fibroblast targeting of Gβγ-GRK2 after myocardial ischemia attenuates heart failure progression. J. Am. Coll. Cardiol. 70, 958–971 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Sorrentino, G. et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat. Cell Biol. 16, 357–366 (2014).

    Article  CAS  PubMed  Google Scholar 

  157. Iaccarino, G. et al. Elevated myocardial and lymphocyte GRK2 expression and activity in human heart failure. Eur. Heart J. 26, 1752–1758 (2005).

    Article  CAS  PubMed  Google Scholar 

  158. Mia, M. M. et al. YAP/TAZ deficiency reprograms macrophage phenotype and improves infarct healing and cardiac function after myocardial infarction. PLoS Biol. 18, e3000941 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Wang, D. et al. YAP promotes the activation of NLRP3 inflammasome via blocking K27-linked polyubiquitination of NLRP3. Nat. Commun. 12, 2674 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Wu, Y. et al. Helicobacter pylori-induced YAP1 nuclear translocation promotes gastric carcinogenesis by enhancing IL-1β expression. Cancer Med. 8, 3965–3980 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Ramjee, V. et al. Epicardial YAP/TAZ orchestrate an immunosuppressive response following myocardial infarction. J. Clin. Invest. 127, 899–911 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  162. Tomasek, J. J., Gabbiani, G., Hinz, B., Chaponnier, C. & Brown, R. A. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat. Rev. Mol. Cell Biol. 3, 349–363 (2002).

    Article  CAS  PubMed  Google Scholar 

  163. Negmadjanov, U. et al. TGF-β1-mediated differentiation of fibroblasts is associated with increased mitochondrial content and cellular respiration. PLoS ONE 10, e0123046 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Emelyanova, L. et al. Impact of statins on cellular respiration and de-differentiation of myofibroblasts in human failing hearts. ESC Heart Fail. 6, 1027–1040 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  165. Van Linthout, S. et al. Anti-inflammatory effects of atorvastatin improve left ventricular function in experimental diabetic cardiomyopathy. Diabetologia 50, 1977–1986 (2007).

    Article  PubMed  Google Scholar 

  166. Van Linthout, S. et al. Human apolipoprotein A-I gene transfer reduces the development of experimental diabetic cardiomyopathy. Circulation 117, 1563–1573 (2008).

    Article  PubMed  Google Scholar 

  167. Van Linthout, S. et al. Reduced MMP-2 activity contributes to cardiac fibrosis in experimental diabetic cardiomyopathy. Basic. Res. Cardiol. 103, 319–327 (2008).

    Article  PubMed  Google Scholar 

  168. Spillmann, F. et al. High-density lipoproteins reduce palmitate-induced cardiomyocyte apoptosis in an AMPK-dependent manner. Biochem. Biophys. Res. Commun. 466, 272–277 (2015).

    Article  CAS  PubMed  Google Scholar 

  169. Beauloye, C., Bertrand, L., Horman, S. & Hue, L. AMPK activation, a preventive therapeutic target in the transition from cardiac injury to heart failure. Cardiovasc. Res. 90, 224–233 (2011).

    Article  CAS  PubMed  Google Scholar 

  170. Wang, W. et al. AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nat. Cell Biol. 17, 490–499 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Vinci, M. C., Polvani, G. & Pesce, M. Epigenetic programming and risk: the birthplace of cardiovascular disease? Stem Cell Rev. Rep. 9, 241–253 (2013).

    Article  PubMed  Google Scholar 

  172. Felisbino, M. B. & McKinsey, T. A. Epigenetics in cardiac fibrosis: emphasis on inflammation and fibroblast activation. JACC Basic. Transl Sci. 3, 704–715 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  173. Yang, C., Tibbitt, M. W., Basta, L. & Anseth, K. S. Mechanical memory and dosing influence stem cell fate. Nat. Mater. 13, 645–652 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Stewart-Morgan, K. R., Petryk, N. & Groth, A. Chromatin replication and epigenetic cell memory. Nat. Cell Biol. 22, 361–371 (2020).

    Article  CAS  PubMed  Google Scholar 

  175. Ferrari, S. & Pesce, M. Cell-based mechanosensation, epigenetics, and non-coding RNAs in progression of cardiac fibrosis. Int. J. Mol. Sci. 21, 28 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  176. Santinon, G., Pocaterra, A. & Dupont, S. Control of YAP/TAZ activity by metabolic and nutrient-sensing pathways. Trends Cell Biol. 26, 289–299 (2016).

    Article  CAS  PubMed  Google Scholar 

  177. Lin, Z. et al. Acetylation of VGLL4 regulates Hippo-YAP signaling and postnatal cardiac growth. Dev. Cell 39, 466–479 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Levy, L. et al. Acetylation of β-catenin by p300 regulates β-catenin-Tcf4 interaction. Mol. Cell Biol. 24, 3404–3414 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Yang, Y., Li, Z., Guo, J. & Xu, Y. Deacetylation of MRTF-A by SIRT1 defies senescence induced down-regulation of collagen type I in fibroblast cells. Biochim. Biophys. Acta Mol. Basis Dis. 1866, 165723 (2020).

    Article  CAS  PubMed  Google Scholar 

  180. Bradshaw, P. C. Acetyl-CoA metabolism and histone acetylation in the regulation of aging and lifespan. Antioxidants 10, 572 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Francois, A., Canella, A., Marcho, L. M. & Stratton, M. S. Protein acetylation in cardiac aging. J. Mol. Cell. Cardiol. 157, 90–97 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Kimball, T. H. & Vondriska, T. M. Metabolism, epigenetics, and causal inference in heart failure. Trends Endocrinol. Metab. 31, 181–191 (2020).

    Article  CAS  PubMed  Google Scholar 

  183. Ferrari, S. & Pesce, M. Stiffness and aging in cardiovascular diseases: the dangerous relationship between force and senescence. Int. J. Mol. Sci. 22, 3404 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Osmanagic-Myers, S., Dechat, T. & Foisner, R. Lamins at the crossroads of mechanosignaling. Genes Dev. 29, 225–237 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Hampoelz, B. & Lecuit, T. Nuclear mechanics in differentiation and development. Curr. Opin. Cell Biol. 23, 668–675 (2011).

    Article  CAS  PubMed  Google Scholar 

  186. Maeshima, K., Tamura, S. & Shimamoto, Y. Chromatin as a nuclear spring. Biophys. Physicobiol. 15, 189–195 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Kupfer, M. E. et al. In situ expansion, differentiation, and electromechanical coupling of human cardiac muscle in a 3D bioprinted, chambered organoid. Circ. Res. 127, 207–224 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Drakhlis, L. et al. Human heart-forming organoids recapitulate early heart and foregut development. Nat. Biotechnol. 39, 737–746 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Tiburcy, M. et al. Defined engineered human myocardium with advanced maturation for applications in heart failure modeling and repair. Circulation 135, 1832–1847 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Ott, H. C. et al. Perfusion-decellularized matrix: using nature’s platform to engineer a bioartificial heart. Nat. Med. 14, 213–221 (2008).

    Article  CAS  PubMed  Google Scholar 

  191. Kanoldt, V., Fischer, L. & Grashoff, C. Unforgettable force – crosstalk and memory of mechanosensitive structures. Biol. Chem. 400, 687–698 (2019).

    Article  CAS  PubMed  Google Scholar 

  192. Norman, M. D. A., Ferreira, S. A., Jowett, G. M., Bozec, L. & Gentleman, E. Measuring the elastic modulus of soft culture surfaces and three-dimensional hydrogels using atomic force microscopy. Nat. Protoc. 16, 2418–2449 (2021).

    Article  CAS  PubMed  Google Scholar 

  193. Sadeghi, A. H. et al. Engineered 3D cardiac fibrotic tissue to study fibrotic remodeling. Adv. Healthc. Mater. 6, 1601434 (2017).

    Article  Google Scholar 

  194. Bracco Gartner, T. C. L. et al. Anti-fibrotic effects of cardiac progenitor cells in a 3D-model of human cardiac fibrosis. Front. Cardiovasc. Med. 6, 52 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  195. Ragazzini, S. et al. Mechanosensor YAP cooperates with TGF-β1 signaling to promote myofibroblast activation and matrix stiffening in a 3D model of human cardiac fibrosis. Acta Biomater. 152, 300–312 (2022).

    Article  CAS  PubMed  Google Scholar 

  196. Wang, H., Haeger, S. M., Kloxin, A. M., Leinwand, L. A. & Anseth, K. S. Redirecting valvular myofibroblasts into dormant fibroblasts through light-mediated reduction in substrate modulus. PLoS ONE 7, e39969 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Milani-Nejad, N. & Janssen, P. M. Small and large animal models in cardiac contraction research: advantages and disadvantages. Pharmacol. Ther. 141, 235–249 (2014).

    Article  CAS  PubMed  Google Scholar 

  198. Abramochkin, D. V., Lozinsky, I. T. & Kamkin, A. Influence of mechanical stress on fibroblast–myocyte interactions in mammalian heart. J. Mol. Cell. Cardiol. 70, 27–36 (2014).

    Article  CAS  PubMed  Google Scholar 

  199. Li, X., Garcia-Elias, A., Benito, B. & Nattel, S. The effects of cardiac stretch on atrial fibroblasts: analysis of the evidence and potential role in atrial fibrillation. Cardiovasc. Res. 118, 440–460 (2022).

    Article  CAS  PubMed  Google Scholar 

  200. Guo, Y. et al. Extracellular matrix of mechanically stretched cardiac fibroblasts improves viability and metabolic activity of ventricular cells. Int. J. Med. Sci. 10, 1837–1845 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Papakrivopoulou, J., Lindahl, G. E., Bishop, J. E. & Laurent, G. J. Differential roles of extracellular signal-regulated kinase 1/2 and p38MAPK in mechanical load-induced procollagen α1(I) gene expression in cardiac fibroblasts. Cardiovasc. Res. 61, 736–744 (2004).

    Article  CAS  PubMed  Google Scholar 

  202. Watson, C. J. et al. Mechanical stretch up-regulates the B-type natriuretic peptide system in human cardiac fibroblasts: a possible defense against transforming growth factor-β mediated fibrosis. Fibrogenes. Tissue Repair 5, 9 (2012).

    Article  CAS  Google Scholar 

  203. Watson, C. J. et al. Extracellular matrix sub-types and mechanical stretch impact human cardiac fibroblast responses to transforming growth factor beta. Connect. Tissue Res. 55, 248–256 (2014).

    Article  CAS  PubMed  Google Scholar 

  204. Li, Y., Asfour, H. & Bursac, N. Age-dependent functional crosstalk between cardiac fibroblasts and cardiomyocytes in a 3D engineered cardiac tissue. Acta Biomater. 55, 120–130 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Kreutzer, F. P. et al. Development and characterization of anti-fibrotic natural compound similars with improved effectivity. Basic. Res. Cardiol. 117, 9 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Perbellini, F. & Thum, T. Living myocardial slices: a novel multicellular model for cardiac translational research. Eur. Heart J. 41, 2405–2408 (2020).

    Article  PubMed  Google Scholar 

  207. Valls-Margarit, M. et al. Engineered macroscale cardiac constructs elicit human myocardial tissue-like functionality. Stem Cell Rep. 13, 207–220 (2019).

    Article  CAS  Google Scholar 

  208. Salvi, M. et al. Automated segmentation of fluorescence microscopy images for 3D cell detection in human-derived cardiospheres. Sci. Rep. 9, 6644 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  209. Neuber, S., Nazari-Shafti, T. Z., Nugraha, B., Falk, V. & Emmert, M. Y. The link between regeneration and extracellular matrix in the heart – can three-dimensional in vitro models uncover it? Eur. Heart J. 42, 2518–2522 (2021).

    Article  PubMed  Google Scholar 

  210. de Boer, R. A. et al. Towards better definition, quantification and treatment of fibrosis in heart failure. A scientific roadmap by the Committee of Translational Research of the Heart Failure Association (HFA) of the European Society of Cardiology. Eur. J. Heart Fail. 21, 272–285 (2019).

    Article  PubMed  Google Scholar 

  211. Santos, G. L., Hartmann, S., Zimmermann, W. H., Ridley, A. & Lutz, S. Inhibition of Rho-associated kinases suppresses cardiac myofibroblast function in engineered connective and heart muscle tissues. J. Mol. Cell. Cardiol. 134, 13–28 (2019).

    Article  CAS  PubMed  Google Scholar 

  212. Francisco, J. et al. Blockade of fibroblast YAP attenuates cardiac fibrosis and dysfunction through MRTF-A inhibition. JACC Basic. Transl Sci. 5, 931–945 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  213. Nagaraju, C. K. et al. Myofibroblast phenotype and reversibility of fibrosis in patients with end-stage heart failure. J. Am. Coll. Cardiol. 73, 2267–2282 (2019).

    Article  PubMed  Google Scholar 

  214. Tzahor, E. & Poss, K. D. Cardiac regeneration strategies: staying young at heart. Science 356, 1035–1039 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Leach, J. P. et al. Hippo pathway deficiency reverses systolic heart failure after infarction. Nature 550, 260–264 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  216. Wang, J., Liu, S., Heallen, T. & Martin, J. F. The Hippo pathway in the heart: pivotal roles in development, disease, and regeneration. Nat. Rev. Cardiol. 15, 672–684 (2018).

    Article  CAS  PubMed  Google Scholar 

  217. Bouvet, M. et al. Anti-integrin αv therapy improves cardiac fibrosis after myocardial infarction by blunting cardiac PW1+ stromal cells. Sci. Rep. 10, 11404 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  218. Esposito, M. L. et al. Left ventricular unloading before reperfusion promotes functional recovery after acute myocardial infarction. J. Am. Coll. Cardiol. 72, 501–514 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  219. Spillmann, F. et al. Mode-of-action of the PROPELLA concept in fulminant myocarditis. Eur. Heart J. 40, 2164–2169 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  220. Burkhoff, D., Topkara, V. K., Sayer, G. & Uriel, N. Reverse remodeling with left ventricular assist devices. Circ. Res. 128, 1594–1612 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Levin, H. R. et al. Reversal of chronic ventricular dilation in patients with end-stage cardiomyopathy by prolonged mechanical unloading. Circulation 91, 2717–2720 (1995).

    Article  CAS  PubMed  Google Scholar 

  222. Mann, D. L., Barger, P. M. & Burkhoff, D. Myocardial recovery and the failing heart: myth, magic, or molecular target? J. Am. Coll. Cardiol. 60, 2465–2472 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  223. Birks, E. J. et al. Prospective multicenter study of myocardial recovery using left ventricular assist devices (RESTAGE-HF [Remission from Stage D Heart Failure]): medium-term and primary end point results. Circulation 142, 2016–2028 (2020).

    Article  PubMed  Google Scholar 

  224. Diakos, N. A. et al. Myocardial atrophy and chronic mechanical unloading of the failing human heart: implications for cardiac assist device-induced myocardial recovery. J. Am. Coll. Cardiol. 64, 1602–1612 (2014).

    Article  PubMed  Google Scholar 

  225. Terracciano, C. M. et al. Clinical recovery from end-stage heart failure using left-ventricular assist device and pharmacological therapy correlates with increased sarcoplasmic reticulum calcium content but not with regression of cellular hypertrophy. Circulation 109, 2263–2265 (2004).

    Article  CAS  PubMed  Google Scholar 

  226. Diakos, N. A. et al. Evidence of glycolysis up-regulation and pyruvate mitochondrial oxidation mismatch during mechanical unloading of the failing human heart: implications for cardiac reloading and conditioning. JACC Basic Transl Sci. 1, 432–444 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  227. Vatta, M. et al. Molecular remodelling of dystrophin in patients with end-stage cardiomyopathies and reversal in patients on assistance-device therapy. Lancet 359, 936–941 (2002).

    Article  CAS  PubMed  Google Scholar 

  228. Canseco, D. C. et al. Human ventricular unloading induces cardiomyocyte proliferation. J. Am. Coll. Cardiol. 65, 892–900 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  229. Symons, J. D. et al. Effect of continuous-flow left ventricular assist device support on coronary artery endothelial function in ischemic and nonischemic cardiomyopathy. Circ. Heart Fail. 12, e006085 (2019).

    Article  PubMed  Google Scholar 

  230. Castillero, E. et al. Structural and functional cardiac profile after prolonged duration of mechanical unloading: potential implications for myocardial recovery. Am. J. Physiol. Heart Circ. Physiol. 315, H1463–H1476 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Klotz, S. et al. Left ventricular assist device support normalizes left and right ventricular beta-adrenergic pathway properties. J. Am. Coll. Cardiol. 45, 668–676 (2005).

    Article  PubMed  Google Scholar 

  232. Bruckner, B. A. et al. Degree of cardiac fibrosis and hypertrophy at time of implantation predicts myocardial improvement during left ventricular assist device support. J. Heart Lung Transpl. 23, 36–42 (2004).

    Article  Google Scholar 

  233. Segura, A. M., Frazier, O. H., Demirozu, Z. & Buja, L. M. Histopathologic correlates of myocardial improvement in patients supported by a left ventricular assist device. Cardiovasc. Pathol. 20, 139–145 (2011).

    Article  PubMed  Google Scholar 

  234. Pan, S. et al. Incidence and predictors of myocardial recovery on long-term left ventricular assist device support: results from the United Network for Organ Sharing database. J. Heart Lung Transpl. 34, 1624–1629 (2015).

    Article  Google Scholar 

  235. Topkara, V. K. et al. Myocardial recovery in patients receiving contemporary left ventricular assist devices: results from the Interagency Registry for Mechanically Assisted Circulatory Support (INTERMACS). Circ. Heart Fail. 9, e003157 (2016).

    Article  PubMed  Google Scholar 

  236. Wever-Pinzon, O. et al. Cardiac recovery during long-term left ventricular assist device support. J. Am. Coll. Cardiol. 68, 1540–1553 (2016).

    Article  PubMed  Google Scholar 

  237. Margulies, K. B. et al. Mixed messages: transcription patterns in failing and recovering human myocardium. Circ. Res. 96, 592–599 (2005).

    Article  CAS  PubMed  Google Scholar 

  238. Yang, K. C. et al. Deep RNA sequencing reveals dynamic regulation of myocardial noncoding RNAs in failing human heart and remodeling with mechanical circulatory support. Circulation 129, 1009–1021 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Tschope, C. et al. Mechanical unloading by fulminant myocarditis: LV-IMPELLA, ECMELLA, BI-PELLA, and PROPELLA concepts. J. Cardiovasc. Transl Res. 12, 116–123 (2019).

    Article  PubMed  Google Scholar 

  240. Weinheimer, C. J. et al. Load-dependent changes in left ventricular structure and function in a pathophysiologically relevant murine model of reversible heart failure. Circ. Heart Fail. 11, e004351 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  241. Oriyanhan, W. et al. Determination of optimal duration of mechanical unloading for failing hearts to achieve bridge to recovery in a rat heterotopic heart transplantation model. J. Heart Lung Transpl. 26, 16–23 (2007).

    Article  Google Scholar 

  242. Webber, M., Jackson, S. P., Moon, J. C. & Captur, G. Myocardial fibrosis in heart failure: anti-fibrotic therapies and the role of cardiovascular magnetic resonance in drug trials. Cardiol. Ther. 9, 363–376 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  243. Pezel, T. et al. Imaging interstitial fibrosis, left ventricular remodeling, and function in stage A and B heart failure. JACC Cardiovasc. Imaging 14, 1038–1052 (2021).

    Article  PubMed  Google Scholar 

  244. Khalique, Z. et al. Diffusion tensor cardiovascular magnetic resonance in cardiac amyloidosis. Circ. Cardiovasc. Imaging 13, e009901 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  245. Tschope, C. et al. Cardiac contractility modulation: mechanisms of action in heart failure with reduced ejection fraction and beyond. Eur. J. Heart Fail. 21, 14–22 (2019).

    Article  PubMed  Google Scholar 

  246. Daneshgar, A. et al. The human liver matrisome – proteomic analysis of native and fibrotic human liver extracellular matrices for organ engineering approaches. Biomaterials 257, 120247 (2020).

    Article  CAS  PubMed  Google Scholar 

  247. Moriel, N. et al. NovoSpaRc: flexible spatial reconstruction of single-cell gene expression with optimal transport. Nat. Protoc. 16, 4177–4200 (2021).

    Article  CAS  PubMed  Google Scholar 

  248. Aghajanian, H. et al. Targeting cardiac fibrosis with engineered T cells. Nature 573, 430–433 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Rurik, J. G. et al. CAR T cells produced in vivo to treat cardiac injury. Science 375, 91–96 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Croft, A. P. et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 570, 246–251 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Nguyen, P. D., de Bakker, D. E. M. & Bakkers, J. Cardiac regenerative capacity: an evolutionary afterthought. Cell. Mol. Life Sci. 78, 5107–5122 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Silva, A. C., Pereira, C., Fonseca, A., Pinto-do, O. P. & Nascimento, D. S. Bearing my heart: the role of extracellular matrix on cardiac development, homeostasis, and injury response. Front. Cell Dev. Biol. 8, 621644 (2020).

    Article  PubMed  Google Scholar 

  253. Pesce, M., Messina, E., Chimenti, I. & Beltrami, A. P. Cardiac mechanoperception: a life-long story from early beats to aging and failure. Stem Cell Dev. 26, 77–90 (2017).

    Article  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

M.P. is supported by institutional grants from the Italian Ministry of Health (Ricerca Corrente, Ricerca 5 per mille). G.N.D. is supported by the Deutsche Forschungsgemeinschaft (SFB 1444). G.F. is supported by the European Regional Development Fund – Project ENOCH (CZ.02.1.01/0.0/0.0/16_019/0000868) and Project MAGNET (CZ.02.1.01/0.0/0.0/15_003/0000492). H.G. is supported by the European Regional Development Fund through the Operational Program for Competitiveness Factors (under the projects HealthyAging2020 CENTRO-01-0145-FEDER-000012-N2323, CENTRO-01-0145-FEDER-032179, CENTRO-01-0145-FEDER-032414, POCI-01-0145-FEDER-022122, UIDB/04539/2020 and UIDP/04539/2020). A.R. is supported by the Spanish Ministry of Economy and Competitiveness (RTI2018-095377-B-100), Instituto de Salud Carlos III-ISCIII/FEDER (TerCel RD16/0011/0024), AGAUR (2017-SGR-899) and CERCA Programme Generalitat de Catalunya. P.R.-C. is supported by the Spanish Ministry of Science and Innovation (PID2019-110298GB-I00), the European Commission (H2020-FETPROACT-01-2016-731957), the ICREA Academia prize for excellence in research, Fundació la Marató de TV3 (201936-30-31) and la Caixa Foundation (agreement LCF/PR/HR20/52400004). J.P.G.S. is supported by a European Union H2020 programme grant EVICARE (725229) and BRAV (874827), and the Gravitation Program (Materials Driven Regeneration 024.003.013) by the Netherlands Organization for Scientific Research. C.T. and S.V.L. are supported by the Deutsche Forschungsgemeinschaft (SFB 1470).

Author information

Authors and Affiliations

Authors

Contributions

M.P. and S.V.L. contributed to the discussion of manuscript content. All the authors contributed to writing, reviewing and editing the manuscript before submission.

Corresponding authors

Correspondence to Maurizio Pesce or Sophie Van Linthout.

Ethics declarations

Competing interests

C.T. has received speaker fees and has contributed to congresses organized by Abbott, Abiomed, AstraZeneca, Bayer, Boehringer-Ingelheim, Novartis, Pfizer and Servier. The other authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Cardiology thanks Nikolaos Frangogiannis and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Glossary

Costameres

Submembranous, Z-line-associated structures found in striated muscle that have important roles in force transmission from the sarcomeres to the sarcolemma and the extracellular matrix, maintenance of mechanical integrity of the sarcolemma, and orchestration of mechanically related signalling.

Dense plaques

Also known as dense bodies. Intercellular adhesion complexes that are functional equivalents of the Z-discs in the striated muscle and fulfil a mechanical function that allows the coordinated contraction of smooth muscle cells.

Dystroglycan complex

A large multicomponent complex that is composed of transmembrane, cytoplasmic and extracellular proteins, including dystrophin, sarcoglycans, dystroglycan, dystrobrevins, syntrophins, sarcospan, caveolin 3 and nitric oxide synthase, and has both mechanical stabilizing and signalling roles in mediating interactions between the cytoskeleton, plasma membrane and extracellular matrix.

Euchromatin

A less condensed chromatin than heterochromatin and more accessible to transcription factors.

Heterochromatin

A densely packed chromatin that is inaccessible to transcription factors and has an important role in maintaining the structural and functional integrity of specific chromosomal regions, such as centromeres and telomeres.

Podosomes

Actin-based dynamic protrusions of the plasma membrane that act as sites of attachment to, and degradation of, the extracellular matrix.

Rigidity

The capacity of a material to withstand deformation when subjected to mechanical loading (such as tension).

Strain

Strain (ε) is the deformation of a material or tissue when subjected to stress; stress (σ) is an internal loading on a material caused by an external force.

Stiffness

The extent to which a material resists deformation in response to an applied load, and is the inverse of compliance.

Teleost

Teleost fish are the most species-rich vertebrate clade, roughly making up half of the existing vertebrate species on the planet, and have extensive genetic and phenotypic variation, resulting in their use in biodiversity and genome evolution studies.

Young’s modulus

(E). Also referred to as modulus of elasticity, it is a property of the material that indicates how easily it can stretch and deform, and is defined as the ratio of tensile stress (σ) to tensile strain (ε).

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pesce, M., Duda, G.N., Forte, G. et al. Cardiac fibroblasts and mechanosensation in heart development, health and disease. Nat Rev Cardiol 20, 309–324 (2023). https://doi.org/10.1038/s41569-022-00799-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41569-022-00799-2

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