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.

A right-handed signalling pathway drives heart looping in vertebrates

Matters Arising to this article was published on 23 September 2020

Abstract

Most animals show external bilateral symmetry, which hinders the observation of multiple internal left–right (L/R) asymmetries that are fundamental to organ packaging and function1,2. In vertebrates, left identity is mediated by the left-specific Nodal–Pitx2 axis that is repressed on the right-hand side by the epithelial–mesenchymal transition (EMT) inducer Snail1 (refs 3, 4). Despite some existing evidence3,5, it remains unclear whether an equivalent instructive pathway provides right-hand-specific information to the embryo. Here we show that, in zebrafish, BMP mediates the L/R asymmetric activation of another EMT inducer, Prrx1a, in the lateral plate mesoderm with higher levels on the right. Prrx1a drives L/R differential cell movements towards the midline, leading to a leftward displacement of the cardiac posterior pole through an actomyosin-dependent mechanism. Downregulation of Prrx1a prevents heart looping and leads to mesocardia. Two parallel and mutually repressed pathways, respectively driven by Nodal and BMP on the left and right lateral plate mesoderm, converge on the asymmetric activation of the transcription factors Pitx2 and Prrx1, which integrate left and right information to govern heart morphogenesis. This mechanism is conserved in the chicken embryo, and in the mouse SNAIL1 acts in a similar manner to Prrx1a in zebrafish and PRRX1 in the chick. Thus, a differential L/R EMT produces asymmetric cell movements and forces, more prominent from the right, that drive heart laterality in vertebrates.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: prrx1a L/R asymmetric expression in zebrafish.
Figure 2: Asymmetric L/R Prrx1a expression drives heart looping in zebrafish.
Figure 3: PRRX1 also drives heart looping in the chick embryo.
Figure 4: Two parallel and mutually exclusive pathways integrate left and right information.
Figure 5: A differential L/R EMT drives heart positioning in vertebrates.

Similar content being viewed by others

References

  1. Raya, A. & Izpisúa Belmonte, J. C. Left–right asymmetry in the vertebrate embryo: from early information to higher-level integration. Nat. Rev. Genet. 7, 283–293 (2006)

    CAS  PubMed  Google Scholar 

  2. López-Gracia, M. L. & Ros, M. A. Left–right asymmetry in vertebrate development. Adv. Anat. Embryol. Cell Biol. 188, 1–121 (2007)

    PubMed  Google Scholar 

  3. Patel, K., Isaac, A. & Cooke, J. Nodal signalling and the roles of the transcription factors SnR and Pitx2 in vertebrate left–right asymmetry. Curr. Biol. 9, 609–612 (1999)

    CAS  PubMed  Google Scholar 

  4. Murray, S. A. & Gridley, T. Snail family genes are required for left–right asymmetry determination, but not neural crest formation, in mice. Proc. Natl Acad. Sci. USA 103, 10300–10304 (2006)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schlueter, J. & Brand, T. A right-sided pathway involving FGF8/Snai1 controls asymmetric development of the proepicardium in the chick embryo. Proc. Natl Acad. Sci. USA 106, 7485–7490 (2009)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 164, 2581–2591 (2014)

    Google Scholar 

  7. Ramsdell, A. F. Left–right asymmetry and congenital cardiac defects: getting to the heart of the matter in vertebrate left–right axis determination. Dev. Biol. 288, 1–20 (2005)

    CAS  PubMed  Google Scholar 

  8. Nieto, M. A., Huang, R. Y., Jackson, R. A. & Thiery, J. P. EMT: 2016. Cell 166, 21–45 (2016)

    CAS  PubMed  Google Scholar 

  9. Acloque, H., Adams, M. S., Fishwick, K., Bronner-Fraser, M. & Nieto, M. A. Epithelial–mesenchymal transitions: the importance of changing cell state in development and disease. J. Clin. Invest. 119, 1438–1449 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Ocaña, O. H. et al. Metastatic colonization requires the repression of the epithelial–mesenchymal transition inducer Prrx1. Cancer Cell 22, 709–724 (2012)

    PubMed  Google Scholar 

  11. Kelly, R. G., Buckingham, M. E. & Moorman, A. F. Heart fields and cardiac morphogenesis. Cold Spring Harb. Perspect. Med. 4, a015750 (2014)

    PubMed  PubMed Central  Google Scholar 

  12. Hami, D., Grimes, A. C., Tsai, H. J. & Kirby, M. L. Zebrafish cardiac development requires a conserved secondary heart field. Development 138, 2389–2398 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Tessadori, F. et al. Identification and functional characterization of cardiac pacemaker cells in zebrafish. PLoS ONE 7, e47644 (2012)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ahn, D.-G., Kourakis, M. J., Rohde, L. A., Silver, L. M., & Ho, R. K. T-box gene tbx5 is essential for formation of the pectoral limb bud. Nature 417, 754–758 (2002)

    ADS  CAS  PubMed  Google Scholar 

  15. Xie, L. et al. Tbx5–hedgehog molecular networks are essential in the second heart field for atrial septation. Dev. Cell 23, 280–291 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Bakkers, J., Verhoeven, M. C. & Abdelilah-Seyfried, S. Shaping the zebrafish heart: from left–right axis specification to epithelial tissue morphogenesis. Dev. Biol. 330, 213–220 (2009)

    CAS  PubMed  Google Scholar 

  17. Veerkamp, J. et al. Unilateral dampening of Bmp activity by nodal generates cardiac left–right asymmetry. Dev. Cell 24, 660–667 (2013)

    CAS  PubMed  Google Scholar 

  18. Noël, E. S. et al. A Nodal-independent and tissue-intrinsic mechanism controls heart-looping chirality. Nat. Commun. 4, 2754 (2013)

    ADS  PubMed  Google Scholar 

  19. Taber, L. A., Voronov, D. A. & Ramasubramanian, A. The role of mechanical forces in the torsional component of cardiac looping. Ann. NY Acad. Sci. 1188, 103–110 (2010)

    ADS  PubMed  Google Scholar 

  20. Domínguez, J. N., Meilhac, S. M., Bland, Y. S., Buckingham, M. E. & Brown, N. A. Asymmetric fate of the posterior part of the second heart field results in unexpected left/right contributions to both poles of the heart. Circ. Res. 111, 1323–1335 (2012)

    PubMed  Google Scholar 

  21. Bayraktar, M. & Männer, J. Cardiac looping may be driven by compressive loads resulting from unequal growth of the heart and pericardial cavity. Observations on a physical simulation model. Front. Physiol. 5, 112 (2014)

    PubMed  PubMed Central  Google Scholar 

  22. Gurung, R. et al. Actin polymerization is stimulated by actin cross-linking protein palladin. Biochem. J. 473, 383–396 (2016)

    CAS  PubMed  Google Scholar 

  23. Najm, P. & El-Sibai, M. Palladin regulation of the actin structures needed for cancer invasion. Cell Adh. Migr. 8, 29–35 (2014)

    PubMed  Google Scholar 

  24. Behrndt, M. et al. Forces driving epithelial spreading in zebrafish gastrulation. Science 338, 257–260 (2012)

    ADS  CAS  PubMed  Google Scholar 

  25. Naganathan, S. R., Middelkoop, T. C., Fürthauer, S. & Grill, S. W. Actomyosin-driven left–right asymmetry: from molecular torques to chiral self organization. Curr. Opin. Cell Biol. 38, 24–30 (2016)

    CAS  PubMed  Google Scholar 

  26. Itasaki, N., Nakamura, H., Sumida, H. & Yasuda, M. Actin bundles on the right side in the caudal part of the heart tube play a role in dextro-looping in the embryonic chick heart. Anat. Embryol. (Berl). 183, 29–39 (1991)

    CAS  PubMed  Google Scholar 

  27. Morales, A. V. et al. Snail genes at the crossroads of symmetric and asymmetric processes in the developing mesoderm. EMBO Rep. 8, 104–109 (2007)

    CAS  PubMed  Google Scholar 

  28. Rana, M. S. et al. Tbx1 coordinates addition of posterior second heart field progenitor cells to the arterial and venous poles of the heart. Circ. Res. 115, 790–799 (2014)

    CAS  PubMed  Google Scholar 

  29. Bressan, M., Liu, G. & Mikawa, T. Early mesodermal cues assign avian cardiac pacemaker fate potential in a tertiary heart field. Science 340, 744–748 (2013)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Pagán-Westphal, S. M. & Tabin, C. J. The transfer of left–right positional information during chick embryogenesis. Cell 93, 25–35 (1998)

    PubMed  Google Scholar 

  31. Chang, H., Zwijsen, A., Vogel, H., Huylebroeck, D. & Matzuk, M. M. Smad5 is essential for left–right asymmetry in mice. Dev. Biol. 219, 71–78 (2000)

    CAS  PubMed  Google Scholar 

  32. Barrallo-Gimeno, A. & Nieto, M. A. Evolutionary history of the Snail/Scratch superfamily. Trends Genet. 25, 248–252 (2009)

    CAS  PubMed  Google Scholar 

  33. Martin, J. F., Bradley, A. & Olson, E. N. The paired-like homeo box gene MHox is required for early events of skeletogenesis in multiple lineages. Genes Dev. 9, 1237–1249 (1995)

    CAS  PubMed  Google Scholar 

  34. Bergwerff, M. et al. Loss of function of the Prx1 and Prx2 homeobox genes alters architecture of the great elastic arteries and ductus arteriosus. Virchows Arch. 436, 12–19 (2000)

    CAS  PubMed  Google Scholar 

  35. Locascio, A., Manzanares, M., Blanco, M. J. & Nieto, M. A. Modularity and reshuffling of Snail and Slug expression during vertebrate evolution. Proc. Natl Acad. Sci. USA 99, 16841–16846 (2002)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Spéder, P., Adám, G. & Noselli, S. Type ID unconventional myosin controls left–right asymmetry in Drosophila. Nature 440, 803–807 (2006)

    ADS  PubMed  Google Scholar 

  37. Gros, J., Feistel, K., Viebahn, C., Blum, M. & Tabin, C. J. Cell movements at Hensen’s node establish left/right asymmetric gene expression in the chick. Science 324, 941–944 (2009)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Maître, J. L. et al. Adhesion functions in cell sorting by mechanically coupling the cortices of adhering cells. Science 338, 253–256 (2012)

    ADS  PubMed  Google Scholar 

  39. Kimmel, C. B., Ballard, W. W., Kimmel, S. R., Ullmann, B. & Schilling, T. F. Stages of embryonic development of the zebrafish. Dev. Dyn. 203, 253–310 (1995)

    CAS  PubMed  Google Scholar 

  40. Hamburger, V. & Hamilton, H. L. A series of normal stages in the development of the chick embryo. J. Morphol. 88, 49–92 (1951)

    CAS  PubMed  Google Scholar 

  41. Rowe, R. G. et al. Mesenchymal cells reactivate Snail1 expression to drive three-dimensional invasion programs. J. Cell Biol. 184, 399–408 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Ruzankina, Y. et al. Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1, 113–126 (2007)

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010)

    CAS  PubMed  Google Scholar 

  44. Carver, E. A., Jiang, R., Lan, Y., Oram, K. F. & Gridley, T. The mouse snail gene encodes a key regulator of the epithelial–mesenchymal transition. Mol. Cell. Biol. 21, 8184–8188 (2001)

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Sefton, M., Sánchez, S. & Nieto, M. A. Conserved and divergent roles for members of the Snail family of transcription factors in the chick and mouse embryo. Development 125, 3111–3121 (1998)

    CAS  PubMed  Google Scholar 

  46. Lee, E. C. et al. A highly efficient Escherichia coli-based chromosome engineering system adapted for recombinogenic targeting and subcloning of BAC DNA. Genomics 73, 56–65 (2001)

    CAS  PubMed  Google Scholar 

  47. Nasevicius, A. & Ekker, S. C. The zebrafish as a novel system for functional genomics and therapeutic development applications. Curr. Opin. Mol. Ther. 3, 224–228 (2001)

    CAS  PubMed  Google Scholar 

  48. Riedl, J. et al. Lifeact: a versatile marker to visualize F-actin. Nat. Methods 5, 605–607 (2008)

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Lombardo, V. A., Sporbert, A. & Abdelilah-Seyfried, S. Cell tracking using photoconvertible proteins during zebrafish development. J. Vis. Exp. 67, 4350 (2012)

    Google Scholar 

  50. Acloque, H., Ocaña, O. H., Abad, D., Stern, C. D. & Nieto, M. A. Snail2 and Zeb2 repress P-cadherin to define embryonic territories in the chick embryo. Development 144, 649–656 (2017)

    CAS  PubMed  Google Scholar 

  51. Bertocchini, F. & Stern, C. D. The hypoblast of the chick embryo positions the primitive streak by antagonizing nodal signaling. Dev. Cell 3, 735–744 (2002)

    CAS  PubMed  Google Scholar 

  52. Gagnon, J. A. et al. Efficient mutagenesis by Cas9 protein-mediated oligonucleotide insertion and large-scale assessment of single-guide RNAs. PLoS ONE 9, e98186 (2014)

    ADS  PubMed  PubMed Central  Google Scholar 

  53. Hwang, W. Y. et al. Efficient genome editing in zebrafish using a CRISPR–Cas system. Nat. Biotechnol. 31, 227–229 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Chapman, S. C., Collignon, J., Schoenwolf, G. C. & Lumsden, A. Improved method for chick whole-embryo culture using a filter paper carrier. Dev. Dyn. 220, 284–289 (2001)

    CAS  PubMed  Google Scholar 

  55. Nieto, M. A., Patel, K. & Wilkinson, D. G. In situ hybridization analysis of chick embryos in whole mount and tissue sections. Methods Cell Biol. 51, 219–235 (1996)

    CAS  PubMed  Google Scholar 

  56. Acloque, H., Wilkinson, D. G. & Nieto, M. A. In situ hybridization analysis of chick embryos in whole-mount and tissue sections. Methods Cell Biol. 87, 169–185 (2008)

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of M.A.N. laboratory for continuous and helpful discussions, J. M. Mingot (IN) for the purification of Cas9 protein, B. Alsina (Universitat Pompeu Fabra) for the myl12.1-mCherry transgenic line (originally from C. P. Heisenberg’s laboratory22), L. M. Gutiérrez (IN) for the LifeAct construct, J. Expósito (IN) for her technical advice with confocal microscopy, J. F. López Téllez (BIONAND) for the semithin histological sections and S. Ingham (IN) for his help with the graphics. This work was supported by the Spanish Ministry of Economy and Competitiveness (BFU2008-01042; BFU2014-53128-R co-financed by the European Regional Development Fund, ERDF), Generalitat Valenciana (PROMETEOII/2013/002) and European Research Council (ERC AdG 322694) to M.A.N., and BFU2014-52299-P, Instituto de Salud Carlos III-TERCEL network (RD12/0019-0022) and Junta de Andalucía (P11-CTS-07564) to R.M.-C. M.A.N. acknowledges financial support from the AEI, through the ‘Severo Ochoa’ Programme for Centres of Excellence in R&D (SEV-2013-0317) to Instituto de Neurociencias (IN).

Author information

Authors and Affiliations

Authors

Contributions

O.H.O. performed the majority of the experiments, analysed the data and contributed to writing. H.C. performed histological and expression studies. C.M. generated the tbx5a zebrafish transgenic line, P.M. generated the anti-Prrx1 antibody in E.M.T.’s laboratory, and J.G. designed and performed the CRISPR-mediated prrx1a mutagenesis and the prrx1-promoter analysis. R.M.-C. carried out the morphological analysis and helped to interpret the data. M.A.N. conceived the project, interpreted the data and wrote the manuscript.

Corresponding author

Correspondence to M. Angela Nieto.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks M. Bronner 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.

Extended data figures and tables

Extended Data Figure 1 prrx1a downregulation induces heart-looping defects.

a, Downregulation of prrx1a transcripts upon prrx1aMO1 microinjection. Ctrol, control. Data represent mean ± s.e.m.; ***P < 0.001; Student’s t-test; n = 3 biological replicates. b, Microinjection of a prrx1aMO2 morpholino located in the 5′ UTR also impairs normal heart looping at 50 hpf. Heart looping assessed by in situ hybridization with a myl7 probe in embryos microinjected with control or prrx1aMO2 morpholinos. The frequency of the phenotypes is indicated below the images. c, Scheme of the prrx1a locus indicating its exon organization. The coding sequence is depicted in blue and the location of the single-guide RNAs (sgRNA) is indicated. The location of primers used for screening is indicated by the two coloured arrows. d, Representative images of ‘crispants’ obtained by injection of the indicated sgRNA and Cas9. The frequency of the phenotypes is indicated below the images. e, Representative T7 endonuclease assays of four embryos per phenotype indicated in d and two wild-type (WT) embryos used as negative controls. f, T7 endonuclease assay of 8 crispants and one WT embryo after injection of the indicated sgRNA. g, The PCR products obtained from embryos 1, 2 and 5 in f were cloned and sequenced. Representative sequences of those embryos (embryo number indicated on the right), with the mutations indicated in red and the deletions with hyphens. The location of the sgRNA is indicated in green and the corresponding adjacent PAM is highlighted in yellow. Scale bars, 100 μm.

Source data

Extended Data Figure 2 Cardiac phenotype of prrx1a-morphant embryos.

a, Bright-field images of whole-mount and haematoxylin–eosin staining of sagittal sections of control and prrx1a-morphant embryos (n = 5 embryos per condition). b, Snapshots of surface-rendering images of representative control and prrx1a-morphant embryos hybridized with the atrial marker amhc. The graph represents the area of amhc expression (μm2; n = 7 per condition). Data represent mean ± s.e.m.; ***P < 0.001; Student’s t-test. c, Parasagittal semithin sections of control and morphant embryos (48 hpf) obtained at the right (R), middle (M) and left (L) levels. The ventricle (V) is on the right and the sinus venosus (SV) is well-developed in the control. However, the morphant has the ventricle aligned with the atrium (A), and lacks a SV in the cardiac posterior pole (PP). OFT, outflow tract. d, Ventral view of a maximum-intensity projection and z-plane sections of confocal images (right) through the outflow tract showing the expression of the anterior SHF marker latent TGFβ binding protein-3 (ltbp3) in control and prrx1a-morphant zebrafish embryos. The cardiomyocytes are visualized by MF20 immunofluorescence. Scale bars, 50 μm.

Source data

Extended Data Figure 3 Prrx1a expression in the early embryo.

a, Maximum-intensity projection of confocal images showing Prrx1 expression in Tg(tbx5a:eGFP) embryos with DAPI staining of cell nuclei (blue). Prrx1 and tbx5a colocalize in the LPM (left). The asymmetric L/R expression of Prrx1 in the right LPM is better seen in the single-channel view (middle) and the transverse sections at the level of the white dotted line (right). tbx5a is absent from the neural crest (NC) and the posterior LPM (PLPM). b, Prrx1 and tbx5 colocalization (yellow) identifies the Prrx1+ cardiac precursor population once the PHT has formed. Number of double-positive cells in the left and right halves of the LPM. Data represent mean ± s.e.m.; **P < 0.01; Student’s t-test; n = 6. c, Heat map of Prrx1-expression levels in the LPM cells identified in b. Colour code is shown on the right. Intensity was measured in individual cells of the selected regions in the left and right LPM and an average intensity value was obtained per side. The plot shows the average value obtained in the right relative to the left LPM. Data represent mean ± s.e.m.; *P < 0.05; Student’s paired t-test; n = 6. d, Dorsal view of zebrafish embryos to visualize the expression of prrx1a and the pan-neural crest marker crestin. Colocalization of prrx1a and crestin identifies the prrx1a+ neural crest cell population. tbx5 is excluded from the neural crest. NT, neural tube. Scale bars, 50 μm.

Source data

Extended Data Figure 4 Prrx1 and Tbx5 co-localization in developing heart tissues.

ac, Ventral views or sagittal sections showing more Prrx1+tbx5a+ cells in the visceral pericardium (P) in contact with the heart tube (HT) on the right side (arrowheads in sections and enlarged images in a and c). Prrx1 is absent from the heart tube. Boxes in c indicate the areas highlighted on the right. d, Sagittal sections of a 48 hpf zebrafish embryo showing Prrx1 expression compared with the cardiomyocyte marker MF20, or the atrial marker S46. Scale bars, 50 μm. A, atrium; E, eye; PV, pericardial vesicle; SV, sinus venosus; V, ventricle.

Extended Data Figure 5 Cell movements during heart development in the zebrafish embryo.

a, Snapshots from time-lapse analysis of Tg(tbx5:eGFP) control and prrx1-morphant embryos at heart-looping stages. b, Randomization of cardiac jogging in prrx1a morphants. Representative images of control (Ctrol MO) and prrx1a-morphant (prrx1aMO1) embryos hybridized with a myosin light chain 7 (myl7) probe to assess heart tube jogging direction. Images are dorsal views with anterior to the top. Scale bars, 100 μm. c, Quantification of heart-jogging direction. A, atrium; PHT, primary heart tube; SV, sinus venosus; V, ventricle.

Source data

Extended Data Figure 6 Photoablation of mesodermal cells in the LPM.

a, b, Photoablation was performed at 26–27 hpf on the left (a) or the right side (b). Ventral views with merged bright-field and z-plane confocal images, before and after unilateral photoablation (Pa) of tbx5:eGFP+ cells. The photoablated areas are located in the most anterior region of the LPM adjacent to the limit of the extra-embryonic tissue. Higher power images of the photoablated areas (right panels) correspond to the boxed areas. c, Analysis of heart location at 50 hpf. Hearts visualized after myl7 in situ hybridization in wild-type and left- or right-photoablated embryos. Mesocardia only develops in embryos photoablated on the right-hand side. d, Cartoon of a ventral view of a zebrafish embryo at 30–32 hpf with a boxed area that corresponds to that in the enlarged areas shown in Fig. 2f, g. e, Snapshots of actomyosin fibres, generated by the merged images extracted from time-lapse confocal microscopy in live control (left) and prrx1a-morphant embryos (right) of the Tg(tbx5a:eGFP) reporter line injected with LifeAct (n = 7 embryos per condition).Brackets indicate the region of the cable-like structure. f, Snapshots from time-lapse videos of Tg(tbx5a:eGFP) embryos treated with blebbistatin or with the vehicle alone. Scale bars, 50 μm.

Extended Data Figure 7 PRRX1 expression in chick embryos.

a, Whole-mount in situ hybridization in chick embryos showing the transient asymmetric expression of SNAIL1 in the LPM. b, Expression analysis by double fluorescence in situ hybridization of PRRX1 and NKX2.5 or TBX5 in whole-mount embryos. c, Transverse sections showing PRRX1 and NKX2.5 or TBX5 expression at the levels indicated by dotted lines in b. Boxes indicate the position of the corresponding enlarged images. The arrowhead indicates the centre of the posterior pole displaced to the left. d, Dual fluorescent in situ hybridization for PRRX1 and the atrial (AMHC) or ventricular (VMHC) myosins. High power images of transverse sections as indicated on the left showing the absence of myocardial PRRX1 expression. e, Downregulation of PRRX1 transcription after electroporation with siRNA. Data represent mean ± s.e.m.; ***P < 0.001; Student’s t-test; n = 3 biological replicates. fh, Quantification of heart location from experiments shown in Fig. 3c, d. RNAi, RNA interference. i, AMHC and VMHC in control embryos and in those electroporated with PRRX1 siRNA. j, Morphometric analysis of the atrium in chick embryos electroporated with control siRNA or PRRX1 siRNA. The graph represents the area of AMHC expression (μm2; n = 7 embryos per condition). Data represent mean ± s.e.m.; **P < 0.01; Student’s t-test. k, Expression of FGF8, required for outflow tract formation, is not altered by PRRX1 downregulation. l, PRRX1 and ISLET1 coexpression in the lateral right sinus venosus horn (RSH) (arrows). Scale bars, 500 μm (a), 10 μm (enlarged images in l) or 100 μm (all other panels). A, atrium; AIP; anterior intestinal portal; CF, cardiac folds; LPM, lateral plate mesoderm; LSH/RSH; left/right sinus venosus horns; OFT, outflow tract; SH, sinus venosus horns; V, ventricle.

Source data

Extended Data Figure 8 PRRX1 is essential in the posterior pole of the heart.

a, Double fluorescent in situ hybridization for TBX18 and PRRX1 in chick embryos. b, TBX18 downregulation in embryos electroporated with PRRX1 siRNA. c, Maximum-intensity projection of dual fluorescent staining of PRRX1 and phalloidin (a mycotoxin that stains actin stress fibres). Stars indicate the area of asymmetry and colocalization. d, Heart looping assessed by MF20 immunofluorescence in embryos treated with vehicle or blebbistatin. e, Maximum-intensity projection showing the cells coexpressing PALLD (palladin) and PRRX1. f, PALLD is decreased upon PRRX1 downregulation. Data represent mean ± s.e.m. of four pools (five embryos each) of control and PRRX1-knockdown embryos; *P < 0.05, **P < 0.01; Student’s t-test. g, Visualization of electroporated areas in chick embryos shown in Fig. 4b. Embryos were co-electroporated on the right side with an eGFP-reporter construct plus an empty vector or a vector containing the NODAL-coding sequence. h, Heart location in embryos containing control BSA- or BMP4-loaded beads. i, Diagram of the PRRX1 promoter. Proximal (P) and distal (D) binding sites for SMAD1/SMAD5/SMAD8 and constructs used in j and k. j, ChIP with antibodies against SMAD5 or IgG (negative control). Data represent mean ± s.e.m.; **P < 0.01, ***P < 0.001; ns, not significant; Student’s t-test; n = 5 biological replicates. k, Luciferase activity of PRRX1 promoter. Both distal (D) and proximal (P) SMAD5-binding sites are functional. Fragments containing these sites can activate the promoter in primary chick fibroblasts upon BMP4 treatment. Mutations in these sites prevent the response to BMP4. Data represent mean ± s.e.m.; ***P < 0.001; ns, not significant; one-way ANOVA; n = 4 biological replicates. Dotted lines show the level of the sections. Scale bars, 100 μm or 10 μm (enlarged images and sections in a, b). A, atrium; AIP, anterior intestinal portal; CF, cardiac folds; LPM, lateral plate mesoderm; LSH/RSH, left/right sinus venosus horns; OFT, outflow tract; SV, sinus venosus; V, ventricle; VV, vitelline veins.

Source data

Extended Data Figure 9 SNAIL1/snail1 and PRRX1 in heart looping in vertebrate embryos.

a, PRRX1 and SNAIL1 expression in whole-mount chick embryos and transverse sections of chick embryos. b, Maximum-intensity projection of confocal images showing heart location by triple immunofluorescence in embryos electroporated with control fluorescein-labelled morpholinos, with a mix of two fluorescein-morpholinos against SNAIL1 or a combination of SNAIL1 morpholinos plus PRRX1 RNA interference. An RFP construct was used as a reporter for electroporated cells. c, Schematic drawing and knockdown-efficiency assay of fluorescein-labelled morpholinos directed to SNAIL1 splicing sites. d, Quantitative analysis of heart location. e, snail1a is not expressed in the LPM in zebrafish embryos. f, snail1b expression in whole-mounted embryos and transverse sections at the levels of the dotted lines. g, Schematic drawing and knockdown efficiency assay of the snail1b splice site morpholino (snail1bMO). h, Quantification of heart location in control and snail1b morphants. i, snail1b is not expressed in the pericardium (P) in the zebrafish embryo. Scale bars, 500 μm (a, left panel), 50 μm (i) or 100 μm (all other panels).

Source data

Extended Data Figure 10 Model for heart looping in vertebrates.

a, Cartoon depicting the comparative expression of Prrx1/PRRX1 and Snail1/SNAIL1 in zebrafish, chick and mouse embryos. b, Transverse sections showing the lack of PRRX2 expression in the sinus venosus horns of chick at HH9–10 and Prrx2 expression in mouse embryos at 8.5 dpc. Scale bars, 100 μm. c, Schematic representation of the proposed model for heart looping in vertebrates in relation to the expression of prrx1a/PRRX1/Prrx1 and snail1b/SNAIL1/Snail1. d, A prominent right-handed pathway drives differential L/R EMT and heart looping in vertebrates. In addition to the left-specific Nodal–Pitx2/PITX2 pathway that confers left identity and represses the right-handed pathway, BMP signalling activates EMT in the LPM in a L/R asymmetric manner to drive heart laterality and the repression of leftward information on the right through the repression of Pitx2/PITX2. This conserved cellular mechanism is implemented through the activation of different EMT inducers in different vertebrates. Nomenclature in the diagrams is unified for all species for simplicity. Correct names are as in the legend. A, atrium; AIP, anterior intestinal portal; C, coelom; DSpLM, dorsal splanchnic lateral mesoderm; Ec, ectoderm; FG, foregut; LA, left atrium; LSH, left sinus venosus horn; LV, left ventricle; LVV, left vitelline vein; NC, neural crest; N, notochord; NP, neural plate; NT, neural tube; OFT, outflow tract; PV, pericardial vesicle; RA, right atrium; RSH, right sinus venosus horn; RV, right ventricle; RVV, right vitelline vein; SoLM, somatic lateral mesoderm; S, somite; SV, sinus venosus; V, ventricle; VSpLM, ventral splanchnic lateral mesoderm; YS, yolk sac.

Supplementary information

Supplementary Information (PDF 200 kb)

Asymmetric cell movements during heart looping in zebrafish

The Tg(tbx5a:eGFP) line shows both the heart tube and late cardiac progenitors contributing to the posterior pole. Embryos were microinjected with Control MO at 1 cell stage and the time-lapse 4D videos were recorded from 28 hpf to 44 hpf taking one picture every 10 minutes. The video shows a ventral view (right side on the left) of surface rendering of a maximum intensity projection. Note the bilaterally asymmetric cell contribution, with more cells from the right side. (MOV 6879 kb)

prrx1a knockdown prevents heart looping in zebrafish

Tg(tbx5a:eGFP) embryos were microinjected with prrx1aMO1 at 1 cell stage and the time lapse 4D videos were recorded from 28 hpf to 44 hpf stage taking one picture every 10 minutes. The video shows images similar to those described in legend to Video S1. Note the decrease in the Tbx5a+ progenitor cells and the absence of heart looping. (MOV 4406 kb)

Down regulation of prrx1a produces randomized heart jogging

Tg(tbx5a:eGFP) embryos were microinjected with prrx1aMO1 at 1 cell stage and the time lapse 4D videos were recorded from 27hpf to 48hpf taking one picture every 10 minutes. The video shows a maximum intensity projection of confocal images. While the jogging is to the left side as in control embryos the heart fails to loop and remains as a straight tube in the midline.Tg(tbx5a:eGFP) embryos were microinjected with prrx1aMO1 at 1 cell stage and the time lapse 4D videos were recorded from 27hpf to 48hpf taking one picture every 10 minutes. The video shows a maximum intensity projection of confocal images. While the jogging is to the left side as in control embryos the heart fails to loop and remains as a straight tube in the midline. (MOV 4837 kb)

Down regulation of prrx1a produces randomized heart jogging

Tg(tbx5a:eGFP) embryos were microinjected with prrx1aMO1 at 1 cell stage and the time lapse 4D videos were recorded from 27hpf to 48hpf taking one picture every 10 minutes. The video shows a maximum intensity projection of confocal images. While the jogging is to the right side the heart fails to loop and remains as an straight tube in the midline. (MOV 4727 kb)

Asymmetric actomyosin forces during heart looping in zebrafish

Tg(tbx5a:eGFP) embryos were microinjected with Control MO plus LifeAct:RFP mRNA at 1 cell-stage and the time-lapse 4D videos were recorded from 28hpf to 38hpf stage taking one picture every 10 minutes. The video shows ventral views (right side on the left) of a maximum intensity projection and was generated with a colocalization channel. Note how actomyosin fibres make contact at the posterior pole of the heart on the right side. (MOV 1640 kb)

Prrx1a drives the asymmetric actomyosin forces necessary for the heart looping in zebrafish

Tg(tbx5a:eGFP) embryos were microinjected with prrx1aMO1 plus LifeAct:RFP mRNA at 1 cell-stage and the time-lapse 4D videos were recorded and presented as described in Video S5. Note the disruption of actomyosin fibres and the absence of the cable-like structure upon prrx1a downregulation. (MOV 813 kb)

prrx1a regulates cell shape and actomyosin cytoskeleton-Control

3D reconstruction at 30-32 hpf in a Tg(actb2:myl12.1-mCherry) zebrafish embryo injected with LifeAct:GFP mRNA and control morpholino (Ctrol MO) at 1 cell-stage embryo to visualize F-actin (lifeAct:GFP, green), myosin II (myl12.1 mCherry, red) and Prrx1 expression (anti-Prrx1, white). (MOV 4822 kb)

prrx1a regulates cell shape and actomyosin cytoskeleton-prrx1aMO1.

3D reconstruction at 30-32 hpf in a Tg(actb2:myl12.1-mCherry) zebrafish embryo injected with LifeAct:GFP mRNA and prrx1a morpholino (prrx1aMO1) at 1 cell-stage embryo to visualize, F-actin (lifeAct:GFP, green), myosin II (myl12.1-mCherry, red) and Prrx1 expression (anti-Prrx1, white). (MOV 4810 kb)

Heart looping in zebrafish-control

Tg(tbx5a:eGFP) embryos were treated with vehicle from 26hpf and the time lapse 4D videos were recorded from 26hpf to 44hpf stage taking one picture every 10 minutes. The video shows a maximum intensity projection. Note the proper dextral looping of the heart. (MOV 4435 kb)

Actomyosin inhibition prevents heart looping in zebrafish

Tg(tbx5a:eGFP) embryos were treated with blebbistatin from 26hpf and the time lapse 4D videos were recorded and presented as described for Video S9. Note how upon blebbistatin treatment the dextral looping of the heart is impeded and the heart remains as a straight tube. (MOV 5648 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ocaña, O., Coskun, H., Minguillón, C. et al. A right-handed signalling pathway drives heart looping in vertebrates. Nature 549, 86–90 (2017). https://doi.org/10.1038/nature23454

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature23454

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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