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:

A change of heart: new roles for cilia in cardiac development and disease

Abstract

Although cardiac abnormalities have been observed in a growing class of human disorders caused by defective primary cilia, the function of cilia in the heart remains an underexplored area. The primary function of cilia in the heart was long thought to be restricted to left−right axis patterning during embryogenesis. However, new findings have revealed broad roles for cilia in congenital heart disease, valvulogenesis, myocardial fibrosis and regeneration, and mechanosensation. In this Review, we describe advances in our understanding of the mechanisms by which cilia function contributes to cardiac left−right axis development and discuss the latest findings that highlight a broader role for cilia in cardiac development. Specifically, we examine the growing line of evidence connecting cilia function to the pathogenesis of congenital heart disease. Furthermore, we also highlight research from the past 10 years demonstrating the role of cilia function in common cardiac valve disorders, including mitral valve prolapse and aortic valve disease, and describe findings that implicate cardiac cilia in mechanosensation potentially linking haemodynamic and contractile forces with genetic regulation of cardiac development and function. Finally, given the presence of cilia on cardiac fibroblasts, we also explore the potential role of cilia in fibrotic growth and summarize the evidence implicating cardiac cilia in heart regeneration.

Key points

  • Cilia are antenna-like organelles that extend from most eukaryotic cells to obtain and interpret information from the extracellular environment; impaired ciliary signalling has been linked with congenital heart disease.

  • Intraciliary calcium signalling in the embryonic left–right organizer initiates vertebrate left–right patterning; abnormal left–right asymmetry is associated with major congenital heart disease, especially in heterotaxy syndrome.

  • Impaired cilia function and signalling are associated with heterotaxy, congenital heart disease, mitral valve prolapse and numerous other cilia-related disorders with cardiac abnormalities.

  • Primary cilia have a role in heart development beyond establishing left–right asymmetry.

  • Endocardial primary cilia might translate mechanical signals to gene expression during heart valve development.

  • Fibroblast cilia have been implicated in the regulation of cardiac regeneration.

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: Diversity of ciliary structure.
Fig. 2: Signalling pathways involved in heart development and organs involved in ciliopathies.
Fig. 3: Model for intraciliary calcium signalling in left–right patterning.
Fig. 4: Cilia defects and left–right patterning impairment leading to heterotaxy and congenital heart disease.
Fig. 5: Cilia-related genes linked to human congenital heart disease.
Fig. 6: Spatiotemporal distribution of primary cilia during valve development.
Fig. 7: Primary cilia as endocardial mechanosensors.

Similar content being viewed by others

References

  1. Satir, P. & Christensen, S. T. Overview of structure and function of mammalian cilia. Annu. Rev. Physiol. 69, 377–400 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. Leeuwenhoek, V. A. Concerning little animals. Philos. Trans. R. Soc. 12, 821–831 (1677).

    Google Scholar 

  3. Zimmermann, K. W. Beiträge zur kenntniss einiger drüsen und epithelien [German]. Arch. Mikrosk. Anat. 52, 552–706 (1898).

    Article  Google Scholar 

  4. Ibanez-Tallon, I., Heintz, N. & Omran, H. To beat or not to beat: roles of cilia in development and disease. Hum. Mol. Genet. 12, R27–35 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Eggenschwiler, J. T. & Anderson, K. V. Cilia and developmental signaling. Annu. Rev. Cell Dev. Biol. 23, 345–373 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Waters, A. M. & Beales, P. L. Ciliopathies: an expanding disease spectrum. Pediatr. Nephrol. 26, 1039–1056 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Hildebrandt, F., Benzing, T. & Katsanis, N. Ciliopathies. N. Engl. J. Med. 364, 1533–1543 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kathem, S. H., Mohieldin, A. M. & Nauli, S. M. The roles of primary cilia in polycystic kidney disease. AIMS Mol. Sci. 1, 27–46 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Mitchison, H. M. & Valente, E. M. Motile and non-motile cilia in human pathology: from function to phenotypes. J. Pathol. 241, 294–309 (2017).

    Article  PubMed  Google Scholar 

  10. Yuan, S., Zaidi, S. & Brueckner, M. Congenital heart disease: emerging themes linking genetics and development. Curr. Opin. Genet. Dev. 23, 352–359 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fakhro, K. A. et al. Rare copy number variations in congenital heart disease patients identify unique genes in left-right patterning. Proc. Natl Acad. Sci. USA 108, 2915–2920 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zaidi, S. et al. De novo mutations in histone-modifying genes in congenital heart disease. Nature 498, 220–223 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gabriel, G. C. & Lo, C. W. Left-right patterning in congenital heart disease beyond heterotaxy. Am. J. Med. Genet. C Semin. Med. Genet. 184, 90–96 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Gabriel, G. C., Young, C. B. & Lo, C. W. Role of cilia in the pathogenesis of congenital heart disease. Semin. Cell Dev. Biol. 110, 2–10 (2021).

    Article  CAS  PubMed  Google Scholar 

  15. Jin, S. C. et al. Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands. Nat. Genet. 49, 1593–1601 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Klena, N. et al. in Etiology and Morphogenesis of Congenital Heart Disease: From Gene Function and Cellular Interaction to Morphology (eds Nakanishi, T. et al.) 67–79 (Springer, 2016).

  17. Klena, N. T., Gibbs, B. C. & Lo, C. W. Cilia and ciliopathies in congenital heart disease. Cold Spring Harb. Perspect. Biol. 9, a028266 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Li, Y. et al. Global genetic analysis in mice unveils central role for cilia in congenital heart disease. Nature 521, 520–524 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kozminski, K. G., Forscher, P. & Rosenbaum, J. L. Three flagellar motilities in Chlamydomonas unrelated to flagellar beating. Video supplement. Cell Motil. Cytoskeleton 39, 347–348 (1998).

    CAS  PubMed  Google Scholar 

  20. Rosenbaum, J. L. & Witman, G. B. Intraflagellar transport. Nat. Rev. Mol. Cell Biol. 3, 813–82 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Lin, J. & Nicastro, D. Asymmetric distribution and spatial switching of dynein activity generates ciliary motility. Science 360, eaar1968 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. McEwen, D. P., Jenkins, P. M. & Martens, J. R. Olfactory cilia: our direct neuronal connection to the external world. Curr. Top. Dev. Biol. 85, 333–370 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Flock, A. & Duvall, A. J. 3rd The ultrastructure of the kinocilium of the sensory cells in the inner ear and lateral line organs. J. Cell Biol. 25, 1–8 (1965).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Boo, K. S. & Richards, A. G. Fine structure of scolopidia in Johnston’s organ of female Aedes aegypti compared with that of the male. J. Insect Physiol. 21, 1129–1139 (1975).

    Article  CAS  PubMed  Google Scholar 

  25. Corbiere-Tichane, G. Fine structure of the sensory apparatus of the mandible of the Speophyes lucidulus larva. (Cavernicolous coleoptera of the subfamily Bathysciinae) [French]. Z. Zellforsch. Mikrosk. Anat. 112, 129–138 (1971).

    CAS  PubMed  Google Scholar 

  26. Gibbons, B. H., Gibbons, I. R. & Baccetti, B. Structure and motility of the 9 + 0 flagellum of eel spermatozoa. J. Submicrosc. Cytol. 15, 15–20 (1983).

    CAS  PubMed  Google Scholar 

  27. Feistel, K. & Blum, M. Three types of cilia including a novel 9+4 axoneme on the notochordal plate of the rabbit embryo. Dev. Dyn. 235, 3348–3358 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Kramer-Zucker, A. G. et al. Cilia-driven fluid flow in the zebrafish pronephros, brain and Kupffer’s vesicle is required for normal organogenesis. Development 132, 1907–1921 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Schweickert, A. et al. Cilia-driven leftward flow determines laterality in Xenopus. Curr. Biol. 17, 60–66 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Nakamura, T. & Hamada, H. Left-right patterning: conserved and divergent mechanisms. Development 139, 3257–3262 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Nonaka, S. et al. Randomization of left-right asymmetry due to loss of nodal cilia generating leftward flow of extraembryonic fluid in mice lacking KIF3B motor protein. Cell 95, 829–837 (1998).

    Article  CAS  PubMed  Google Scholar 

  32. McGrath, J., Somlo, S., Makova, S., Tian, X. & Brueckner, M. Two populations of node monocilia initiate left-right asymmetry in the mouse. Cell 114, 61–73 (2003).

    Article  CAS  PubMed  Google Scholar 

  33. Caspary, T., Larkins, C. E. & Anderson, K. V. The graded response to Sonic Hedgehog depends on cilia architecture. Dev. Cell 12, 767–778 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Gluenz, E. et al. Beyond 9+0: noncanonical axoneme structures characterize sensory cilia from protists to humans. FASEB J. 24, 3117–3121 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Goetz, J. G. et al. Endothelial cilia mediate low flow sensing during zebrafish vascular development. Cell Rep. 6, 799–808 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Sun, S., Fisher, R. L., Bowser, S. S., Pentecost, B. T. & Sui, H. Three-dimensional architecture of epithelial primary cilia. Proc. Natl Acad. Sci. USA 116, 9370–9379 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kiesel, P. et al. The molecular structure of mammalian primary cilia revealed by cryo-electron tomography. Nat. Struct. Mol. Biol. 27, 1115–1124 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Singla, V. & Reiter, J. F. The primary cilium as the cell’s antenna: signaling at a sensory organelle. Science 313, 629–633 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Anvarian, Z., Mykytyn, K., Mukhopadhyay, S., Pedersen, L. B. & Christensen, S. T. Cellular signalling by primary cilia in development, organ function and disease. Nat. Rev. Nephrol. 15, 199–219 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Wheway, G., Nazlamova, L. & Hancock, J. T. Signaling through the primary cilium. Front. Cell Dev. Biol. 6, 8 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Huangfu, D. & Anderson, K. V. Cilia and Hedgehog responsiveness in the mouse. Proc. Natl Acad. Sci. USA 102, 11325–11330 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Corbit, K. C. et al. Vertebrate Smoothened functions at the primary cilium. Nature 437, 1018–1021 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Lienkamp, S., Ganner, A. & Walz, G. Inversin, Wnt signaling and primary cilia. Differentiation 83, S49–55 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Oh, E. C. & Katsanis, N. Context-dependent regulation of Wnt signaling through the primary cilium. J. Am. Soc. Nephrol. 24, 10–18 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Clement, C. A. et al. TGF-beta signaling is associated with endocytosis at the pocket region of the primary cilium. Cell Rep. 3, 1806–1814 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Ezratty, E. J. et al. A role for the primary cilium in Notch signaling and epidermal differentiation during skin development. Cell 145, 1129–1141 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Liu, Z. et al. Primary cilia regulate hematopoietic stem and progenitor cell specification through Notch signaling in zebrafish. Nat. Commun. 10, 1839 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Rohatgi, R., Milenkovic, L. & Scott, M. P. Patched1 regulates hedgehog signaling at the primary cilium. Science 317, 372–376 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Haycraft, C. J. et al. Gli2 and Gli3 localize to cilia and require the intraflagellar transport protein polaris for processing and function. PLoS Genet. 1, e53 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Nauli, S. M. et al. Polycystins 1 and 2 mediate mechanosensation in the primary cilium of kidney cells. Nat. Genet. 33, 129–137 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Delling, M., DeCaen, P. G., Doerner, J. F., Febvay, S. & Clapham, D. E. Primary cilia are specialized calcium signalling organelles. Nature 504, 311–314 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. DeCaen, P. G., Delling, M., Vien, T. N. & Clapham, D. E. Direct recording and molecular identification of the calcium channel of primary cilia. Nature 504, 315–318 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yuan, S., Zhao, L., Brueckner, M. & Sun, Z. Intraciliary calcium oscillations initiate vertebrate left-right asymmetry. Curr. Biol. 25, 556–567 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mizuno, K. et al. Role of Ca2+ transients at the node of the mouse embryo in breaking of left-right symmetry. Sci. Adv. 6, eaba1195 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Su, S. et al. Genetically encoded calcium indicator illuminates calcium dynamics in primary cilia. Nat. Methods 10, 1105–1107 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Shiba, D. et al. Localization of Inv in a distinctive intraciliary compartment requires the C-terminal ninein-homolog-containing region. J. Cell Sci. 122, 44–54 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Bennett, H. W. et al. Novel fibrillar structure in the inversin compartment of primary cilia revealed by 3D single-molecule superresolution microscopy. Mol. Biol. Cell 31, 619–639 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Liu, P. et al. Chlamydomonas PKD2 organizes mastigonemes, hair-like glycoprotein polymers on cilia. J. Cell Biol. 219, e202001122 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Afzelius, B. A. A human syndrome caused by immotile cilia. Science 193, 317–319 (1976).

    Article  CAS  PubMed  Google Scholar 

  60. Pazour, G. J., San Agustin, J. T., Follit, J. A., Rosenbaum, J. L. & Witman, G. B. Polycystin-2 localizes to kidney cilia and the ciliary level is elevated in orpk mice with polycystic kidney disease. Curr. Biol. 12, R378–380 (2002).

    Article  CAS  PubMed  Google Scholar 

  61. Hirokawa, N., Tanaka, Y., Okada, Y. & Takeda, S. Nodal flow and the generation of left-right asymmetry. Cell 125, 33–45 (2006).

    Article  CAS  PubMed  Google Scholar 

  62. Shinohara, K. et al. Two rotating cilia in the node cavity are sufficient to break left-right symmetry in the mouse embryo. Nat. Commun. 3, 622 (2012).

    Article  PubMed  Google Scholar 

  63. Norris, D. P. Cilia, calcium and the basis of left-right asymmetry. BMC Biol. 10, 102 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hirokawa, N., Tanaka, Y. & Okada, Y. Cilia, KIF3 molecular motor and nodal flow. Curr. Opin. Cell Biol. 24, 31–39 (2012).

    Article  CAS  PubMed  Google Scholar 

  65. Tabin, C. J. & Vogan, K. J. A two-cilia model for vertebrate left-right axis specification. Genes. Dev. 17, 1–6 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Okada, Y. et al. Abnormal nodal flow precedes situs inversus in iv and inv mice. Mol. Cell 4, 459–468 (1999).

    Article  CAS  PubMed  Google Scholar 

  67. Supp, D. M. et al. Targeted deletion of the ATP binding domain of left-right dynein confirms its role in specifying development of left-right asymmetries. Development 126, 5495–5504 (1999).

    Article  CAS  PubMed  Google Scholar 

  68. Nonaka, S., Shiratori, H., Saijoh, Y. & Hamada, H. Determination of left-right patterning of the mouse embryo by artificial nodal flow. Nature 418, 96–99 (2002).

    Article  CAS  PubMed  Google Scholar 

  69. Marszalek, J. R., Ruiz-Lozano, P., Roberts, E., Chien, K. R. & Goldstein, L. S. Situs inversus and embryonic ciliary morphogenesis defects in mouse mutants lacking the KIF3A subunit of kinesin-II. Proc. Natl Acad. Sci. USA 96, 5043–5048 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Tisler, M., Thumberger, T., Schneider, I., Schweickert, A. & Blum, M. Leftward flow determines laterality in conjoined twins. Curr. Biol. 27, 543–548 (2017).

    Article  CAS  PubMed  Google Scholar 

  71. Hashimoto, M. et al. Planar polarization of node cells determines the rotational axis of node cilia. Nat. Cell Biol. 12, 170–176 (2010).

    Article  CAS  PubMed  Google Scholar 

  72. Sampaio, P. et al. Left-right organizer flow dynamics: how much cilia activity reliably yields laterality? Dev. Cell 29, 716–728 (2014).

    Article  CAS  PubMed  Google Scholar 

  73. Vick, P. et al. Flow on the right side of the gastrocoel roof plate is dispensable for symmetry breakage in the frog Xenopus laevis. Dev. Biol. 331, 281–291 (2009).

    Article  CAS  PubMed  Google Scholar 

  74. Yoshiba, S. et al. Cilia at the node of mouse embryos sense fluid flow for left-right determination via Pkd2. Science 338, 226–231 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Tanaka, Y., Okada, Y. & Hirokawa, N. FGF-induced vesicular release of Sonic hedgehog and retinoic acid in leftward nodal flow is critical for left-right determination. Nature 435, 172–177 (2005).

    Article  CAS  PubMed  Google Scholar 

  76. Wang, J. et al. C. elegans ciliated sensory neurons release extracellular vesicles that function in animal communication. Curr. Biol. 24, 519–525 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wood, C. R. & Rosenbaum, J. L. Ciliary ectosomes: transmissions from the cell’s antenna. Trends Cell Biol. 25, 276–285 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Nager, A. R. et al. An actin network dispatches ciliary GPCRs into extracellular vesicles to modulate signaling. Cell 168, 252–263.e14 (2017).

    Article  CAS  PubMed  Google Scholar 

  79. Phua, S. C. et al. Dynamic remodeling of membrane composition drives cell cycle through primary cilia excision. Cell 168, 264–279.e15 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wood, C. R., Huang, K., Diener, D. R. & Rosenbaum, J. L. The cilium secretes bioactive ectosomes. Curr. Biol. 23, 906–911 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Takao, D. et al. Asymmetric distribution of dynamic calcium signals in the node of mouse embryo during left-right axis formation. Dev. Biol. 376, 23–30 (2013).

    Article  CAS  PubMed  Google Scholar 

  82. Pennekamp, P. et al. The ion channel polycystin-2 is required for left-right axis determination in mice. Curr. Biol. 12, 938–943 (2002).

    Article  CAS  PubMed  Google Scholar 

  83. Meno, C. et al. Mouse Lefty2 and zebrafish antivin are feedback inhibitors of nodal signaling during vertebrate gastrulation. Mol. Cell 4, 287–298 (1999).

    Article  CAS  PubMed  Google Scholar 

  84. Saijoh, Y. et al. Two nodal-responsive enhancers control left-right asymmetric expression of Nodal. Dev. Dyn. 232, 1031–1036 (2005).

    Article  CAS  PubMed  Google Scholar 

  85. Levin, M., Johnson, R. L., Stern, C. D., Kuehn, M. & Tabin, C. A molecular pathway determining left-right asymmetry in chick embryogenesis. Cell 82, 803–814 (1995).

    Article  CAS  PubMed  Google Scholar 

  86. Shiratori, H., Yashiro, K., Shen, M. M. & Hamada, H. Conserved regulation and role of Pitx2 in situs-specific morphogenesis of visceral organs. Development 133, 3015–3025 (2006).

    Article  CAS  PubMed  Google Scholar 

  87. Little, R. B. & Norris, D. P. Right, left and cilia: how asymmetry is established. Semin. Cell Dev. Biol. 110, 11–18 (2021).

    Article  CAS  PubMed  Google Scholar 

  88. Delling, M. et al. Primary cilia are not calcium-responsive mechanosensors. Nature 531, 656–660 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Behringer, R. G. M., Vintersten Nagy, K. & Nagy, A. Manipulating the Mouse Embryo: A Laboratory Manual. 4th edn, 814 (Cold Spring Harbor Laboratory Press, 2014).

  90. Chen, T. W. et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499, 295–300 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Ohkura, M. et al. Genetically encoded green fluorescent Ca2+ indicators with improved detectability for neuronal Ca2+ signals. PLoS ONE 7, e51286 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Zhao, Y. et al. An expanded palette of genetically encoded Ca2+ indicators. Science 333, 1888–1891 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Kim, J. H. et al. High cleavage efficiency of a 2A peptide derived from porcine teschovirus-1 in human cell lines, zebrafish and mice. PLoS ONE 6, e18556 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Cho, J. H. et al. The GCaMP-R family of genetically encoded ratiometric calcium indicators. ACS Chem. Biol. 12, 1066–1074 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Schweickert, A. et al. The nodal inhibitor Coco is a critical target of leftward flow in Xenopus. Curr. Biol. 20, 738–743 (2010).

    Article  CAS  PubMed  Google Scholar 

  96. Marques, S. et al. The activity of the Nodal antagonist Cerl-2 in the mouse node is required for correct L/R body axis. Genes Dev. 18, 2342–2347 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Kawasumi, A. et al. Left-right asymmetry in the level of active Nodal protein produced in the node is translated into left-right asymmetry in the lateral plate of mouse embryos. Dev. Biol. 353, 321–330 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Nakamura, T. et al. Fluid flow and interlinked feedback loops establish left-right asymmetric decay of Cerl2 mRNA. Nat. Commun. 3, 1322 (2012).

    Article  PubMed  Google Scholar 

  99. Minegishi, K. et al. Fluid flow-induced left-right asymmetric decay of Dand5 mRNA in the mouse embryo requires a Bicc1-Ccr4 RNA degradation complex. Nat. Commun. 12, 4071 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Hamada, H. Molecular and cellular basis of left-right asymmetry in vertebrates. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 96, 273–296 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Francescatto, L., Rothschild, S. C., Myers, A. L. & Tombes, R. M. The activation of membrane targeted CaMK-II in the zebrafish Kupffer’s vesicle is required for left-right asymmetry. Development 137, 2753–2762 (2010).

    Article  CAS  PubMed  Google Scholar 

  102. Mochizuki, T. et al. Cloning of inv, a gene that controls left/right asymmetry and kidney development. Nature 395, 177–181 (1998).

    Article  CAS  PubMed  Google Scholar 

  103. Burns, T. A. et al. A novel mouse model for cilia-associated cardiovascular anomalies with a high penetrance of total anomalous pulmonary venous return. Anat. Rec. 302, 136–145 (2019).

    Article  CAS  Google Scholar 

  104. Kennedy, M. P. et al. Congenital heart disease and other heterotaxic defects in a large cohort of patients with primary ciliary dyskinesia. Circulation 115, 2814–2821 (2007).

    Article  PubMed  Google Scholar 

  105. Lin, A. E. et al. Laterality defects in the national birth defects prevention study (1998-2007): birth prevalence and descriptive epidemiology. Am. J. Med. Genet. A 164A, 2581–2591 (2014).

    Article  PubMed  Google Scholar 

  106. Nakhleh, N. et al. High prevalence of respiratory ciliary dysfunction in congenital heart disease patients with heterotaxy. Circulation 125, 2232–2242 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  107. Burkhalter, M. D. et al. Imbalanced mitochondrial function provokes heterotaxy via aberrant ciliogenesis. J. Clin. Invest. 129, 2841–2855 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Robson, A. et al. Histone H2B monoubiquitination regulates heart development via epigenetic control of cilia motility. Proc. Natl Acad. Sci. USA 116, 14049–14054 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Desgrange, A., Le Garrec, J. F. & Meilhac, S. M. Left-right asymmetry in heart development and disease: forming the right loop. Development 145, dev162776 (2018).

    Article  PubMed  Google Scholar 

  110. Ray, P. et al. Intrinsic cellular chirality regulates left-right symmetry breaking during cardiac looping. Proc. Natl Acad. Sci. USA 115, E11568–E11577 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Jacobs, J. P. et al. The nomenclature, definition and classification of cardiac structures in the setting of heterotaxy. Cardiol. Young 17 (Suppl. 2), 1–28 (2007).

    PubMed  Google Scholar 

  112. Icardo, J. M. & Sanchez de Vega, M. J. Spectrum of heart malformations in mice with situs solitus, situs inversus, and associated visceral heterotaxy. Circulation 84, 2547–2558 (1991).

    Article  CAS  PubMed  Google Scholar 

  113. Field, S. et al. Pkd1l1 establishes left-right asymmetry and physically interacts with Pkd2. Development 138, 1131–1142 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Shylo, N. A., Emmanouil, E., Ramrattan, D. & Weatherbee, S. D. Loss of ciliary transition zone protein TMEM107 leads to heterotaxy in mice. Dev. Biol. 460, 187–199 (2020).

    Article  CAS  PubMed  Google Scholar 

  115. Slough, J., Cooney, L. & Brueckner, M. Monocilia in the embryonic mouse heart suggest a direct role for cilia in cardiac morphogenesis. Dev. Dyn. 237, 2304–2314 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Burnicka-Turek, O. et al. Cilia gene mutations cause atrioventricular septal defects by multiple mechanisms. Hum. Mol. Genet. 25, 3011–3028 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Frescura, C., Ho, S. Y., Giordano, M. & Thiene, G. Isomerism of the atrial appendages: morphology and terminology. Cardiovasc. Pathol. 47, 107205 (2020).

    Article  PubMed  Google Scholar 

  118. Briggs, L. E. et al. Wnt/beta-catenin and sonic hedgehog pathways interact in the regulation of the development of the dorsal mesenchymal protrusion. Dev. Dyn. 245, 103–113 (2016).

    Article  CAS  PubMed  Google Scholar 

  119. Hoffmann, A. D., Peterson, M. A., Friedland-Little, J. M., Anderson, S. A. & Moskowitz, I. P. Sonic hedgehog is required in pulmonary endoderm for atrial septation. Development 136, 1761–1770 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Digilio, M. C. et al. Atrioventricular canal defect and genetic syndromes: the unifying role of sonic hedgehog. Clin. Genet. 95, 268–276 (2019).

    Article  CAS  PubMed  Google Scholar 

  121. Liu, S. et al. DNAH11 variants and its association with congenital heart disease and heterotaxy syndrome. Sci. Rep. 9, 6683 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Frommer, A. et al. Immunofluorescence analysis and diagnosis of primary ciliary dyskinesia with radial spoke defects. Am. J. Respir. Cell Mol. Biol. 53, 563–573 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Pugnaloni, F., Versacci, P., Marino, B. & Digilio, M. C. Atrioventricular canal defect is the classic congenital heart disease in Bardet-Biedl syndrome. Ann. Hum. Genet. 85, 101–102 (2021).

    Article  PubMed  Google Scholar 

  124. Scott, C. A. et al. Nuclear/cytoplasmic transport defects in BBS6 underlie congenital heart disease through perturbation of a chromatin remodeling protein. PLoS Genet. 13, e1006936 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  125. Olson, A. J., Krentz, A. D., Finta, K. M., Okorie, U. C. & Haws, R. M. Thoraco-abdominal abnormalities in Bardet-Biedl syndrome: situs inversus and heterotaxy. J. Pediatr. 204, 31–37 (2019).

    Article  PubMed  Google Scholar 

  126. Gabriel, G. C., Pazour, G. J. & Lo, C. W. Congenital heart defects and ciliopathies associated with renal phenotypes. Front. Pediatr. 6, 175 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  127. Damerla, R. R. et al. Novel Jbts17 mutant mouse model of Joubert syndrome with cilia transition zone defects and cerebellar and other ciliopathy related anomalies. Hum. Mol. Genet. 24, 3994–4005 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Huang, L. et al. TMEM237 is mutated in individuals with a Joubert syndrome related disorder and expands the role of the TMEM family at the ciliary transition zone. Am. J. Hum. Genet. 89, 713–730 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. French, V. M. et al. NPHP4 variants are associated with pleiotropic heart malformations. Circ. Res. 110, 1564–1574 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. van Bon, B. W. et al. Transposition of the great vessels in a patient with a 2.9 Mb interstitial deletion of 9q31.1 encompassing the inversin gene: clinical report and review. Am. J. Med. Genet. Part. A 146A, 1225–1229 (2008).

    Article  PubMed  Google Scholar 

  131. Watkins, W. S. et al. De novo and recessive forms of congenital heart disease have distinct genetic and phenotypic landscapes. Nat. Commun. 10, 4722 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  132. Knowles, M. R., Zariwala, M. & Leigh, M. Primary ciliary dyskinesia. Clin. Chest Med. 37, 449–461 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  133. Li, A. H. et al. Genetic architecture of laterality defects revealed by whole exome sequencing. Eur. J. Hum. Genet. 27, 563–573 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. van Dam, T. J. et al. The SYSCILIA gold standard (SCGSv1) of known ciliary components and its applications within a systems biology consortium. Cilia 2, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  135. van Dam, T. J. P. et al. CiliaCarta: an integrated and validated compendium of ciliary genes. PLoS ONE 14, e0216705 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  136. Tory, K. et al. Mutations of NPHP2 and NPHP3 in infantile nephronophthisis. Kidney Int. 75, 839–847 (2009).

    Article  CAS  PubMed  Google Scholar 

  137. Baujat, G. & Le Merrer, M. Ellis-van Creveld syndrome. Orphanet J. Rare Dis. 2, 27 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  138. Karp, N., Grosse-Wortmann, L. & Bowdin, S. Severe aortic stenosis, bicuspid aortic valve and atrial septal defect in a child with Joubert syndrome and related disorders (JSRD) — a case report and review of congenital heart defects reported in the human ciliopathies. Eur. J. Med. Genet. 55, 605–610 (2012).

    Article  PubMed  Google Scholar 

  139. Deveault, C. et al. BBS genotype-phenotype assessment of a multiethnic patient cohort calls for a revision of the disease definition. Hum. Mutat. 32, 610–619 (2011).

    Article  CAS  PubMed  Google Scholar 

  140. Lumiaho, A. et al. Mitral valve prolapse and mitral regurgitation are common in patients with polycystic kidney disease type 1. Am. J. Kidney Dis. 38, 1208–1216 (2001).

    Article  CAS  PubMed  Google Scholar 

  141. Toomer, K. A. et al. A role for primary cilia in aortic valve development and disease. Dev. Dyn. 246, 625–634 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Fulmer, D. et al. Defects in the exocyst-cilia machinery cause bicuspid aortic valve disease and aortic stenosis. Circulation 140, 1331–1341 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Koefoed, K., Veland, I. R., Pedersen, L. B., Larsen, L. A. & Christensen, S. T. Cilia and coordination of signaling networks during heart development. Organogenesis 10, 108–125 (2014).

    Article  PubMed  Google Scholar 

  144. Sund, K. L., Roelker, S., Ramachandran, V., Durbin, L. & Benson, D. W. Analysis of Ellis van Creveld syndrome gene products: implications for cardiovascular development and disease. Hum. Mol. Genet. 18, 1813–1824 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Pierpont, M. E., Markwald, R. R. & Lin, A. E. Genetic aspects of atrioventricular septal defects. Am. J. Med. Genet. 97, 289–296 (2000).

    Article  CAS  PubMed  Google Scholar 

  146. Maslen, C. L. et al. CRELD1 mutations contribute to the occurrence of cardiac atrioventricular septal defects in Down syndrome. Am. J. Med. Genet. A 140, 2501–2505 (2006).

    Article  PubMed  Google Scholar 

  147. Ahuja, N., Ostwald, P., Bark, D. & Garrity, D. Biomechanical cues direct valvulogenesis. J. Cardiovasc. Dev. Dis. 7, 18 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  148. Van der Heiden, K. et al. Monocilia on chicken embryonic endocardium in low shear stress areas. Dev. Dyn. 235, 19–28 (2006).

    Article  PubMed  Google Scholar 

  149. Willaredt, M. A., Gorgas, K., Gardner, H. A. & Tucker, K. L. Multiple essential roles for primary cilia in heart development. Cilia 1, 23 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  150. Samsa, L. A. et al. Cardiac contraction activates endocardial Notch signaling to modulate chamber maturation in zebrafish. Development 142, 4080–4091 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Li, X. et al. Primary cilia mediate Klf2-dependant Notch activation in regenerating heart. Protein Cell 11, 433–445 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Iomini, C., Tejada, K., Mo, W., Vaananen, H. & Piperno, G. Primary cilia of human endothelial cells disassemble under laminar shear stress. J. Cell Biol. 164, 811–817 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Rydholm, S. et al. Mechanical properties of primary cilia regulate the response to fluid flow. Am. J. Physiol. Ren. Physiol. 298, F1096–1102 (2010).

    Article  CAS  Google Scholar 

  154. Ritter, A., Louwen, F. & Yuan, J. Deficient primary cilia in obese adipose-derived mesenchymal stem cells: obesity, a secondary ciliopathy? Obes. Rev. 19, 1317–1328 (2018).

    Article  CAS  PubMed  Google Scholar 

  155. Groenendijk, B. C., Hierck, B. P., Gittenberger-De Groot, A. C. & Poelmann, R. E. Development-related changes in the expression of shear stress responsive genes KLF-2, ET-1, and NOS-3 in the developing cardiovascular system of chicken embryos. Dev. Dyn. 230, 57–68 (2004).

    Article  CAS  PubMed  Google Scholar 

  156. Egorova, A. D. et al. Lack of primary cilia primes shear-induced endothelial-to-mesenchymal transition. Circ. Res. 108, 1093–1101 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Ten Dijke, P., Egorova, A. D., Goumans, M. J., Poelmann, R. E. & Hierck, B. P. TGF-beta signaling in endothelial-to-mesenchymal transition: the role of shear stress and primary cilia. Sci. Signal. 5, pt2 (2012).

    PubMed  Google Scholar 

  158. Clement, C. A. et al. The primary cilium coordinates early cardiogenesis and hedgehog signaling in cardiomyocyte differentiation. J. Cell Sci. 122, 3070–3082 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Wu, G. et al. Cardiac defects and renal failure in mice with targeted mutations in Pkd2. Nat. Genet. 24, 75–78 (2000).

    Article  CAS  PubMed  Google Scholar 

  160. Paavola, J. et al. Polycystin-2 mutations lead to impaired calcium cycling in the heart and predispose to dilated cardiomyopathy. J. Mol. Cell Cardiol. 58, 199–208 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Peralta, M. et al. Intraflagellar transport complex B proteins regulate the Hippo effector Yap1 during cardiogenesis. Cell Rep. 32, 107932 (2020).

    Article  CAS  PubMed  Google Scholar 

  162. Toomer, K. A. et al. Primary cilia defects causing mitral valve prolapse. Sci. Transl. Med. 11, eaax0290 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Fulmer, D. et al. Desert hedgehog-primary cilia cross talk shapes mitral valve tissue by organizing smooth muscle actin. Dev. Biol. 463, 26–38 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Goddard, L. M. et al. Hemodynamic forces sculpt developing heart valves through a KLF2-WNT9B paracrine signaling axis. Dev. Cell 43, 274–289.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Lehman, J. M. et al. The Oak Ridge polycystic kidney mouse: modeling ciliopathies of mice and men. Dev. Dyn. 237, 1960–1971 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  166. Lambrechts, D. & Carmeliet, P. Sculpting heart valves with NFATc and VEGF. Cell 118, 532–534 (2004).

    Article  CAS  PubMed  Google Scholar 

  167. Wu, B., Baldwin, H. S. & Zhou, B. Nfatc1 directs the endocardial progenitor cells to make heart valve primordium. Trends Cardiovasc. Med. 23, 294–300 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  168. Wu, B. et al. Nfatc1 coordinates valve endocardial cell lineage development required for heart valve formation. Circ. Res. 109, 183–192 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Luu, V. Z., Chowdhury, B., Al-Omran, M., Hess, D. A. & Verma, S. Role of endothelial primary cilia as fluid mechanosensors on vascular health. Atherosclerosis 275, 196–204 (2018).

    Article  CAS  PubMed  Google Scholar 

  170. Van der Heiden, K., Egorova, A. D., Poelmann, R. E., Wentzel, J. J. & Hierck, B. P. Role for primary cilia as flow detectors in the cardiovascular system. Int. Rev. Cell Mol. Biol. 290, 87–119 (2011).

    Article  PubMed  Google Scholar 

  171. Praetorius, H. A. & Spring, K. R. The renal cell primary cilium functions as a flow sensor. Curr. Opin. Nephrol. Hypertens. 12, 517–520 (2003).

    Article  PubMed  Google Scholar 

  172. Praetorius, H. A. & Spring, K. R. Bending the MDCK cell primary cilium increases intracellular calcium. J. Membr. Biol. 184, 71–79 (2001).

    Article  CAS  PubMed  Google Scholar 

  173. Praetorius, H. A. & Spring, K. R. Removal of the MDCK cell primary cilium abolishes flow sensing. J. Membr. Biol. 191, 69–76 (2003).

    Article  CAS  PubMed  Google Scholar 

  174. Nauli, S. M. et al. Endothelial cilia are fluid shear sensors that regulate calcium signaling and nitric oxide production through polycystin-1. Circulation 117, 1161–1171 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. AbouAlaiwi, W. A. et al. Ciliary polycystin-2 is a mechanosensitive calcium channel involved in nitric oxide signaling cascades. Circ. Res. 104, 860–869 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Groenendijk, B. C. et al. Changes in shear stress-related gene expression after experimentally altered venous return in the chicken embryo. Circ. Res. 96, 1291–1298 (2005).

    Article  CAS  PubMed  Google Scholar 

  177. Hierck, B. P. et al. Primary cilia sensitize endothelial cells for fluid shear stress. Dev. Dyn. 237, 725–735 (2008).

    Article  CAS  PubMed  Google Scholar 

  178. Ferreira, R. R., Fukui, H., Chow, R., Vilfan, A. & Vermot, J. The cilium as a force sensor-myth versus reality. J. Cell Sci. 132, jcs213496 (2019).

    Article  Google Scholar 

  179. Poelmann, R. E., Van der Heiden, K., Gittenberger-de Groot, A. & Hierck, B. P. Deciphering the endothelial shear stress sensor. Circulation 117, 1124–1126 (2008).

    Article  PubMed  Google Scholar 

  180. Pala, R., Jamal, M., Alshammari, Q. & Nauli, S. M. The roles of primary cilia in cardiovascular diseases. Cells 7, 233 (2018).

    Article  CAS  PubMed Central  Google Scholar 

  181. Heckel, E. et al. Oscillatory flow modulates mechanosensitive klf2a expression through trpv4 and trpp2 during heart valve development. Curr. Biol. 25, 1354–1361 (2015).

    Article  CAS  PubMed  Google Scholar 

  182. Li, H., Chang, C., Li, X. & Zhang, R. The roles and activation of endocardial Notch signaling in heart regeneration. Cell Regen. 10, 3 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  183. Galvez-Santisteban, M. et al. Hemodynamic-mediated endocardial signaling controls in vivo myocardial reprogramming. eLife 8, e44816 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Rash, J. E., Shay, J. W. & Biesele, J. J. Cilia in cardiac differentiation. J. Ultrastruct. Res. 29, 470–484 (1969).

    Article  CAS  PubMed  Google Scholar 

  185. Myklebust, R., Engedal, H., Saetersdal, T. S. & Ulstein, M. Primary 9 + 0 cilia in the embryonic and the adult human heart. Anat. Embryol. 151, 127–139 (1977).

    Article  CAS  Google Scholar 

  186. Diguet, N., Le Garrec, J. F., Lucchesi, T. & Meilhac, S. M. Imaging and analyzing primary cilia in cardiac cells. Methods Cell Biol. 127, 55–73 (2015).

    Article  CAS  PubMed  Google Scholar 

  187. Villalobos, E. et al. Fibroblast primary cilia are required for cardiac fibrosis. Circulation 139, 2342–2357 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Blom, J. N., Lu, X., Kim, M. Y. & Feng, Q. Primary cilia disassembly promotes mammalian cardiac regeneration [abstr.]. FASEB J. 30 (Suppl. 1), 1207.7 (2016).

    Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  190. Raya, A. et al. Activation of Notch signaling pathway precedes heart regeneration in zebrafish. Proc. Natl Acad. Sci. USA 100 (Suppl. 1), 11889–11895 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Zhao, L. et al. Notch signaling regulates cardiomyocyte proliferation during zebrafish heart regeneration. Proc. Natl Acad. Sci. USA 111, 1403–1408 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Zhao, L., Ben-Yair, R., Burns, C. E. & Burns, C. G. Endocardial notch signaling promotes cardiomyocyte proliferation in the regenerating zebrafish heart through wnt pathway antagonism. Cell Rep. 26, 546–554.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Guichard, C. et al. Axonemal dynein intermediate-chain gene (DNAI1) mutations result in situs inversus and primary ciliary dyskinesia (Kartagener syndrome). Am. J. Hum. Genet. 68, 1030–1035 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Zariwala, M. A. et al. Mutations of DNAI1 in primary ciliary dyskinesia: evidence of founder effect in a common mutation. Am. J. Respir. Crit. Care Med. 174, 858–866 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Liu, H. et al. Next-generation sequencing in a series of 80 fetuses with complex cardiac malformations and/or heterotaxy. Hum. Mutat. 41, 2167–2178 (2020).

    Article  CAS  PubMed  Google Scholar 

  196. Mitchison, H. M. et al. Mutations in axonemal dynein assembly factor DNAAF3 cause primary ciliary dyskinesia. Nat. Genet. 44, 381–389 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Hornef, N. et al. DNAH5 mutations are a common cause of primary ciliary dyskinesia with outer dynein arm defects. Am. J. Respir. Crit. Care Med. 174, 120–126 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Duriez, B. et al. A common variant in combination with a nonsense mutation in a member of the thioredoxin family causes primary ciliary dyskinesia. Proc. Natl Acad. Sci. USA 104, 3336–3341 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Knowles, M. R. et al. Mutations of DNAH11 in patients with primary ciliary dyskinesia with normal ciliary ultrastructure. Thorax 67, 433–441 (2012).

    Article  PubMed  Google Scholar 

  200. Bartoloni, L. et al. Mutations in the DNAH11 (axonemal heavy chain dynein type 11) gene cause one form of situs inversus totalis and most likely primary ciliary dyskinesia. Proc. Natl Acad. Sci. USA 99, 10282–10286 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Omran, H. et al. Ktu/PF13 is required for cytoplasmic pre-assembly of axonemal dyneins. Nature 456, 611–616 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Merveille, A. C. et al. CCDC39 is required for assembly of inner dynein arms and the dynein regulatory complex and for normal ciliary motility in humans and dogs. Nat. Genet. 43, 72–78 (2011).

    Article  CAS  PubMed  Google Scholar 

  203. Becker-Heck, A. et al. The coiled-coil domain containing protein CCDC40 is essential for motile cilia function and left-right axis formation. Nat. Genet. 43, 79–84 (2011).

    Article  CAS  PubMed  Google Scholar 

  204. Mazor, M. et al. Primary ciliary dyskinesia caused by homozygous mutation in DNAL1, encoding dynein light chain 1. Am. J. Hum. Genet. 88, 599–607 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Panizzi, J. R. et al. CCDC103 mutations cause primary ciliary dyskinesia by disrupting assembly of ciliary dynein arms. Nat. Genet. 44, 714–719 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Horani, A. et al. Whole-exome capture and sequencing identifies HEATR2 mutation as a cause of primary ciliary dyskinesia. Am. J. Hum. Genet. 91, 685–693 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Kott, E. et al. Loss-of-function mutations in LRRC6, a gene essential for proper axonemal assembly of inner and outer dynein arms, cause primary ciliary dyskinesia. Am. J. Hum. Genet. 91, 958–964 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Onoufriadis, A. et al. Splice-site mutations in the axonemal outer dynein arm docking complex gene CCDC114 cause primary ciliary dyskinesia. Am. J. Hum. Genet. 92, 88–98 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Zariwala, M. A. et al. ZMYND10 is mutated in primary ciliary dyskinesia and interacts with LRRC6. Am. J. Hum. Genet. 93, 336–345 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Moore, D. J. et al. Mutations in ZMYND10, a gene essential for proper axonemal assembly of inner and outer dynein arms in humans and flies, cause primary ciliary dyskinesia. Am. J. Hum. Genet. 93, 346–356 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Hjeij, R. et al. ARMC4 mutations cause primary ciliary dyskinesia with randomization of left/right body asymmetry. Am. J. Hum. Genet. 93, 357–367 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Tarkar, A. et al. DYX1C1 is required for axonemal dynein assembly and ciliary motility. Nat. Genet. 45, 995–1003 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Austin-Tse, C. et al. Zebrafish ciliopathy screen plus human mutational analysis identifies C21orf59 and CCDC65 defects as causing primary ciliary dyskinesia. Am. J. Hum. Genet. 93, 672–686 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Knowles, M. R. et al. Mutations in SPAG1 cause primary ciliary dyskinesia associated with defective outer and inner dynein arms. Am. J. Hum. Genet. 93, 711–720 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Hjeij, R. et al. CCDC151 mutations cause primary ciliary dyskinesia by disruption of the outer dynein arm docking complex formation. Am. J. Hum. Genet. 95, 257–274 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Paff, T. et al. Mutations in PIH1D3 cause X-linked primary ciliary dyskinesia with outer and inner dynein arm defects. Am. J. Hum. Genet. 100, 160–168 (2017).

    Article  CAS  PubMed  Google Scholar 

  217. Duquesnoy, P. et al. Loss-of-function mutations in the human ortholog of Chlamydomonas reinhardtii ODA7 disrupt dynein arm assembly and cause primary ciliary dyskinesia. Am. J. Hum. Genet. 85, 890–896 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Loges, N. T. et al. Deletions and point mutations of LRRC50 cause primary ciliary dyskinesia due to dynein arm defects. Am. J. Hum. Genet. 85, 883–889 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Hartill, V. L. et al. DNAAF1 links heart laterality with the AAA+ ATPase RUVBL1 and ciliary intraflagellar transport. Hum. Mol. Genet. 27, 529–545 (2018).

    Article  CAS  PubMed  Google Scholar 

  220. Perles, Z. et al. A human laterality disorder associated with recessive CCDC11 mutation. J. Med. Genet. 49, 386–390 (2012).

    Article  CAS  PubMed  Google Scholar 

  221. Li, Y. et al. DNAH6 and its interactions with PCD genes in heterotaxy and primary ciliary dyskinesia. PLoS Genet. 12, e1005821 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  222. Wallmeier, J. et al. TTC25 deficiency results in defects of the outer dynein arm docking machinery and primary ciliary dyskinesia with left-right body asymmetry randomization. Am. J. Hum. Genet. 99, 460–469 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Bataille, S. et al. Association of PKD2 (polycystin 2) mutations with left-right laterality defects. Am. J. Kidney Dis. 58, 456–460 (2011).

    Article  CAS  PubMed  Google Scholar 

  224. Vetrini, F. et al. Bi-allelic mutations in PKD1L1 are associated with laterality defects in humans. Am. J. Hum. Genet. 99, 886–893 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Otto, E. A. et al. Mutations in INVS encoding inversin cause nephronophthisis type 2, linking renal cystic disease to the function of primary cilia and left-right axis determination. Nat. Genet. 34, 413–420 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Okada, M. et al. Association of INVS (NPHP2) mutation in an adolescent exhibiting nephronophthisis (NPH) and complete situs inversus. Clin. Nephrol. 69, 135–141 (2008).

    Article  CAS  PubMed  Google Scholar 

  227. Chaki, M. et al. Genotype-phenotype correlation in 440 patients with NPHP-related ciliopathies. Kidney Int. 80, 1239–1245 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Bergmann, C. et al. Loss of nephrocystin-3 function can cause embryonic lethality, Meckel-Gruber-like syndrome, situs inversus, and renal-hepatic-pancreatic dysplasia. Am. J. Hum. Genet. 82, 959–970 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Hoff, S. et al. ANKS6 is a central component of a nephronophthisis module linking NEK8 to INVS and NPHP3. Nat. Genet. 45, 951–956 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Frank, V. et al. Mutations in NEK8 link multiple organ dysplasia with altered Hippo signalling and increased c-MYC expression. Hum. Mol. Genet. 22, 2177–2185 (2013).

    Article  CAS  PubMed  Google Scholar 

  231. Stone, D. L. et al. Mutation of a gene encoding a putative chaperonin causes McKusick–Kaufman syndrome. Nat. Genet. 25, 79–82 (2000).

    Article  CAS  PubMed  Google Scholar 

  232. Kaasinen, E. et al. Recessively inherited right atrial isomerism caused by mutations in growth/differentiation factor 1 (GDF1). Hum. Mol. Genet. 19, 2747–2753 (2010).

    Article  CAS  PubMed  Google Scholar 

  233. Boskovski, M. T. et al. The heterotaxy gene GALNT11 glycosylates Notch to orchestrate cilia type and laterality. Nature 504, 456–459 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Ruiz-Perez, V. L. et al. Mutations in a new gene in Ellis–van Creveld syndrome and Weyers acrodental dysostosis. Nat. Genet. 24, 283–286 (2000).

    Article  CAS  PubMed  Google Scholar 

  235. Durst, R. et al. Mutations in DCHS1 cause mitral valve prolapse. Nature 525, 109–113 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Del Viso, F. et al. Congenital heart disease genetics uncovers context-dependent organization and function of nucleoporins at cilia. Dev. Cell 38, 478–492 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  237. Williams, K., Carson, J. & Lo, C. Genetics of congenital heart disease. Biomolecules 9, 879 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  238. Hamada, H. in Etiology and Morphogenesis of Congenital Heart Disease: From Gene Function and Cellular Interaction to Morphology (eds Nakanishi, T. et al.) 57–65 (Springer, 2016).

Download references

Author information

Authors and Affiliations

Authors

Contributions

All authors researched data for the article, discussed its content, wrote the manuscript, and reviewed and edited it before submission.

Corresponding authors

Correspondence to Martina Brueckner or Shiaulou Yuan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

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

Publisher’s note

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

Glossary

Cilia

Membrane-bound, microtubule-based, antenna-like sensory organelles that project from the surface of most animal cells to coordinate numerous signalling pathways during development and in tissue homeostasis.

Congenital heart disease

(CHD). A disease characterized by a structural abnormality of the heart that is not an acquired condition.

Left–right organizer

(LRO). Evolutionarily conserved embryonic ciliated organ of laterality in which breaking of left–right asymmetry occurs.

Heterotaxy

(HTX). Any arrangement of the organs across the left–right axis differing from situs solitus and situs inversus.

Atrioventricular septal defects

(AVSDs). Conditions characterized by improper atrial and ventricular septa and adjoining valve development.

Transposition of the great arteries

(TGA). A congenital heart defect characterized by a switch in the position of the pulmonary artery and the aorta.

Endocardial cushion

(ECC). A small pocket of cardiac jelly between the endocardial lining and the myocardium in the atrioventricular canal and outflow tract.

Atrioventricular canal

(AVC). The channel connecting the developing atrium and ventricles in embryonic development.

Outflow tract

(OFT). The channel connecting the developing ventricles and dorsal aorta in embryonic development.

Endothelial-to-mesenchymal transition

(EndoMT). The differentiation of endothelial cells to mesenchyme during cardiac cushion development.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Djenoune, L., Berg, K., Brueckner, M. et al. A change of heart: new roles for cilia in cardiac development and disease. Nat Rev Cardiol 19, 211–227 (2022). https://doi.org/10.1038/s41569-021-00635-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41569-021-00635-z

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