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.

  • Letter
  • Published:

YAP is essential for tissue tension to ensure vertebrate 3D body shape

Subjects

Abstract

Vertebrates have a unique 3D body shape in which correct tissue and organ shape and alignment are essential for function. For example, vision requires the lens to be centred in the eye cup which must in turn be correctly positioned in the head1. Tissue morphogenesis depends on force generation, force transmission through the tissue, and response of tissues and extracellular matrix to force2,3. Although a century ago D’Arcy Thompson postulated that terrestrial animal body shapes are conditioned by gravity4, there has been no animal model directly demonstrating how the aforementioned mechano-morphogenetic processes are coordinated to generate a body shape that withstands gravity. Here we report a unique medaka fish (Oryzias latipes) mutant, hirame (hir), which is sensitive to deformation by gravity. hir embryos display a markedly flattened body caused by mutation of YAP, a nuclear executor of Hippo signalling that regulates organ size. We show that actomyosin-mediated tissue tension is reduced in hir embryos, leading to tissue flattening and tissue misalignment, both of which contribute to body flattening. By analysing YAP function in 3D spheroids of human cells, we identify the Rho GTPase activating protein ARHGAP18 as an effector of YAP in controlling tissue tension. Together, these findings reveal a previously unrecognised function of YAP in regulating tissue shape and alignment required for proper 3D body shape. Understanding this morphogenetic function of YAP could facilitate the use of embryonic stem cells to generate complex organs requiring correct alignment of multiple tissues.

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

Figure 1: Organ/tissue collapse and misalignment in hir mutants.
Figure 2: Tissue tension is reduced in hir mutants.
Figure 3: Cell and tissue dynamics in hir mutants.
Figure 4: YAP regulation of tissue tension and FN assembly is mediated by ARHGAP18.

Similar content being viewed by others

References

  1. Chauhan, B. K. et al. Cdc42- and IRSp53-dependent contractile filopodia tether presumptive lens and retina to coordinate epithelial invagination. Development 136, 3657–3667 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Nelson, C. M. & Bissell, M. J. Of extracellular matrix, scaffolds, and signaling: tissue architecture regulates development, homeostasis, and cancer. Annu. Rev. Cell Dev. Biol. 22, 287–309 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Mammoto, T. & Ingber, D. E. Mechanical control of tissue and organ development. Development 137, 1407–1420 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Thompson, D. W. On Growth and Form (Cambridge Univ. Press, 1917)

    Google Scholar 

  5. Furutani-Seiki, M. et al. Neural degeneration mutants in the zebrafish, Danio rerio. Development 123, 229–239 (1996)

    CAS  PubMed  Google Scholar 

  6. Furutani-Seiki, M. et al. A systematic genome-wide screen for mutations affecting organogenesis in medaka, Oryzias latipes. Mech. Dev. 121, 647–658 (2004)

    Article  CAS  PubMed  Google Scholar 

  7. Sudol, M. et al. Characterization of the mammalian YAP (Yes-associated protein) gene and its role in defining a novel protein module, the WW domain. J. Biol. Chem. 270, 14733–14741 (1995)

    Article  CAS  PubMed  Google Scholar 

  8. Pan, D. The Hippo signaling pathway in development and cancer. Dev. Cell 19, 491–505 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhao, B., Tumaneng, K. & Guan, K.-L. L. The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. Nature Cell Biol. 13, 877–883 (2011)

    Article  CAS  PubMed  Google Scholar 

  10. Miesfeld, J. B. & Link, B. A. Establishment of transgenic lines to monitor and manipulate Yap/Taz-Tead activity in zebrafish reveals both evolutionarily conserved and divergent functions of the Hippo pathway. Mech. Dev. 133, 177–188 (2014)

    Article  CAS  PubMed  Google Scholar 

  11. Gee, S. T., Milgram, S. L., Kramer, K. L., Conlon, F. L. & Moody, S. A. Yes-associated protein 65 (YAP) expands neural progenitors and regulates Pax3 expression in the neural plate border zone. PLoS ONE 6, e20309 (2011)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lei, Q. Y. et al. TAZ promotes cell proliferation and epithelial-mesenchymal transition and is inhibited by the Hippo pathway. Mol. Cell. Biol. 28, 2426–2436 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhao, B., Li, L., Tumaneng, K., Wang, C. Y. & Guan, K.-L. L. A coordinated phosphorylation by Lats and CK1 regulates YAP stability through SCFβ-TRCP. Genes Dev. 24, 72–85 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Heisenberg, C.-P. P. & Bellaïche, Y. Forces in tissue morphogenesis and patterning. Cell 153, 948–962 (2013)

    Article  CAS  Google Scholar 

  15. Vicente-Manzanares, M., Ma, X., Adelstein, R. S. & Horwitz, A. R. Cytoskeletal motors: non-muscle myosin II takes centre stage in cell adhesion and migration. Nature Rev. Mol. Cell Biol. 10, 778–790 (2009)

    Article  CAS  Google Scholar 

  16. Köppen, M., Fernández, B. G., Carvalho, L., Jacinto, A. & Heisenberg, C.-P. P. Coordinated cell-shape changes control epithelial movement in zebrafish and Drosophila. Development 133, 2671–2681 (2006)

    Article  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Guevorkian, K., Colbert, M.-J., Durth, M., Dufour, S. & Brochard-Wyart, F. Aspiration of biological viscoelastic drops. Phys. Rev. Lett. 104, 218101 (2010)

    Article  ADS  PubMed  Google Scholar 

  19. Singh, P., Carraher, C. & Schwarzbauer, J. E. Assembly of fibronectin extracellular matrix. Annu. Rev. Cell Dev. Biol. 26, 397–419 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Rolo, A., Skoglund, P. & Keller, R. E. Morphogenetic movements driving neural tube closure in Xenopus require myosin IIB. Dev. Biol. 327, 327–338 (2009)

    Article  CAS  PubMed  Google Scholar 

  21. Maeda, M. et al. ARHGAP18, a GTPase-activating protein for RhoA, controls cell shape, spreading, and motility. Mol. Biol. Cell 22, 3840–3852 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. McDonald, J. A. et al. Fibronectin’s cell-adhesive domain and an amino-terminal matrix assembly domain participate in its assembly into fibroblast pericellular matrix. J. Biol. Chem. 262, 2957–2967 (1987)

    CAS  PubMed  Google Scholar 

  23. Iwasaki, T., Murata-Hori, M., Ishitobi, S. & Hosoya, H. Diphosphorylated MRLC is required for organization of stress fibers in interphase cells and the contractile ring in dividing cells. Cell Struct. Funct. 26, 677–683 (2001)

    Article  CAS  PubMed  Google Scholar 

  24. Dupont, S. et al. Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183 (2011)

    Article  CAS  PubMed  Google Scholar 

  25. Pinto, I. M., Rubinstein, B., Kucharavy, A., Unruh, J. R. & Li, R. Actin depolymerization drives actomyosin ring contraction during budding yeast cytokinesis. Dev. Cell 22, 1247–1260 (2012)

    Article  PubMed Central  Google Scholar 

  26. Sansores-Garcia, L. et al. Modulating F-actin organization induces organ growth by affecting the Hippo pathway. EMBO J. 30, 2325–2335 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Daley, W. P., Peters, S. B. & Larsen, M. Extracellular matrix dynamics in development and regenerative medicine. J. Cell Sci. 121, 255–264 (2008)

    Article  CAS  PubMed  Google Scholar 

  28. Morin-Kensicki, E. M. et al. Defects in yolk sac vasculogenesis, chorioallantoic fusion, and embryonic axis elongation in mice with targeted disruption of Yap65. Mol. Cell. Biol. 26, 77–87 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hilman, D. & Gat, U. The evolutionary history of YAP and the Hippo/YAP pathway. Mol. Biol. Evol. 28, 2403–2417 (2011)

    Article  CAS  PubMed  Google Scholar 

  30. Sasai, Y. Cytosystems dynamics in self-organization of tissue architecture. Nature 493, 318–326 (2013)

    Article  ADS  CAS  PubMed  Google Scholar 

  31. Porazinski, S. R., Wang, H. & Furutani-Seiki, M. Essential techniques for introducing medaka to a zebrafish laboratory–towards the combined use of medaka and zebrafish for further genetic dissection of the function of the vertebrate genome. Methods Mol. Biol. 770, 211–241 (2011)

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  PubMed  Google Scholar 

  33. Hirose, Y. Varga, Z. M., Kondoh, H. & Furutani-Seiki, M. Single cell lineage and regionalization of cell populations during Medaka neurulation. Development 131, 2553–2563 (2004)

    Article  CAS  PubMed  Google Scholar 

  34. Iwanami, N. et al. WDR55 is a nucleolar modulator of ribosomal RNA synthesis, cell cycle progression, and teleost organ development. PLoS Genet. 4, e1000171 (2008)

    Article  PubMed  PubMed Central  Google Scholar 

  35. Naruse, K. et al. A medaka gene map: the trace of ancestral vertebrate proto-chromosomes revealed by comparative gene mapping. Genome Res. 14, 820–828 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Itoh, M., Nagafuchi, A., Moroi, S. & Tsukita, S. Involvement of ZO-1 in cadherin-based cell adhesion through its direct binding to alpha catenin and actin filaments. J. Cell Biol. 138, 181–192 (1997)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Alexandre, P., Reugels, A. M., Barker, D., Blanc, E. & Clarke, J. D. Neurons derive from the more apical daughter in asymmetric divisions in the zebrafish neural tube. Nature Neurosci. 13, 673–679 (2010)

    Article  CAS  PubMed  Google Scholar 

  38. Mayer, M., Depken, M., Bois, J. S., Jülicher, F. & Grill, S. W. Anisotropies in cortical tension reveal the physical basis of polarizing cortical flows. Nature 467, 617–621 (2010)

    Article  ADS  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Raff, T. Perry, A. Ward, M. Wills, J. Caunt, J. Clarke, L. Hurst and C. Tickle for critical reading and comments. We thank M. Tada, M. Furuse, N. Wada, Y. Nakai, J. Robinson and R. Kelsh for contributions to the paper and University of Bath for fish and bioimaging facilities. This work was funded by the ERATO/SORST projects of JST, Japan (H.K.), National Institutes of Health R01EY014167 (B.A.L.) and Medical Research Council, UK (M.F.-S.).

Author information

Authors and Affiliations

Authors

Contributions

S.P., H.W., Y.A., M.B., T.M., H.M., S.H., T.S., S.F.G.K., Y.O., S.A., A.M., S.L., J.B.M., B.A.L., T.S., A.C.M., A.O.U., S.B. and M.F.-S. performed experiments. S.P., H.W., Y.A., M.B., T.M. and M.F.-S. conceived the study. S.B., N.S., H.N., S.M., H.K., C.-P.H., H.N. and M.F.- S. supervised the study. C.-P.H. and M.F.- S. wrote the paper. All authors interpreted data.

Corresponding authors

Correspondence to Hiroshi Nishina, Carl-Philipp Heisenberg or Makoto Furutani-Seiki.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 YAP is mutated in hir mutants.

a, In situ hybridization of sox3 showed that the lens placode (arrowhead) is specified in hir mutant embryos (n = 3) at st. 21. At st. 22, the nascent lens invaginated in WT (n = 21), but not in hir mutant embryos (n = 13, arrowhead). b, Two frames from time-lapse imaging of retina of embryos injected with membrane EGFP and nuclear red fluorescent protein (MNFP) mRNAs. In WT (n = 10), the nascent lens invaginates from st. 21 (b1, margins of the lens indicated by arrowheads with retina to the right), whereas in hir (n = 7) the lens mostly detached from the retina (b2′, arrowheads show lens remnants attached to the retina). Scale bars, 80 μm in a; 30 μm in b. c, Nine recombinants in 1,908 meiosis mapped hir close to the YAP gene on chromosome 13 (R: recombinant, C: non-recombinant embryos). d, YAP cDNA encodes six protein binding domains/motifs and one transcription activation (TAc) domain; a non-sense mutation in WW1 domain in hir. e, RT–PCR analysis of YAP mRNA during development. β-actin as control. f, mRNA of normal YAP and its variants were injected into hir mutants. The numbers represent: hir phenotype rescue judged via brain thickness, heart migration and Cuvier’s duct formation; mutants (judged by genotyping when necessary); survived injected embryos of hir+/− crosses. High dose (400 pg) mRNA of YAPhir variant was injected into WT embryos to examine dominant-negative effects. The rescue by YAP4SA variant required only 20% of the amount required to rescue using normal YAP mRNA.

Extended Data Figure 2 Morpholino knockdown in medaka and zebrafish.

a, Design of medaka YAP TB and SB MOs relative to translation start (ATG), exons (numbered boxes) and introns. Primers (arrows) used to assess the efficiency of SB MO KD. b, Upper panel, proper splicing of YAP transcripts (579 bp) was nearly fully blocked (343 bp, <5% of normal level) by YAP SB MO (5 ng), assessed by RT–PCR; lower panel, β-actin control. c, WT embryos injected with YAP TB MO and standard control MO. c1c3 Dorsal and c1′c3′ lateral views (also Supplementary Table 1). Arrowheads indicate location of heart progenitors. Body flattening and bilateral cardiac progenitor cell migration was affected in a dose-dependent manner. c2, c2′, Bilateral cardiac progenitor cells fused at the midline but did not migrate anteriorly; c1, c1′ their migration arrests next to the ears at the high dose. The two distinct YAP morpholinos (YAP TB and SB MOs) mimicked the hir phenotype (hir mutants have a cardia bifida phenotype (arrowheads in Fig. 1a1 and a1′)) in a dose-dependent manner. To further verify specificity of the YAP MOs, YAP TB MO was co-injected with human YAP mRNA that does not hybridize with the YAP TB MO. Injection of YAP TB (but not YAP SB) MO into hir mutant embryos enhanced the blastopore closure phenotype of hir mutants (Fig. 1b, c, Supplementary Table 2). These maternal YAP KD hir mutant embryos failed to close the blastopore. Less than half the amount (2 ng) of YAP TB MO was required for causing this phenotype in hir mutants compared to that required for WT embryos (5 ng). This blastopore closure phenotype was rescued by medaka YAP mRNA (200 pg) co-injection. dg, Zebrafish (ZF) WT embryos injected with three distinct ZFYAP MOs (TB, 5′ UTR and SB) exhibit the blastopore closure phenotype as in medaka (Supplementary Table 3). Efficiencies of ZF YAP and TAZ SB MO KD (1.5 ng each) were assessed by RT–PCR using primers in d, f, respectively as in a, b. As reported by Gee et al., co-injection of ZF YAP mRNAs did not rescue the ZF YAP MO phenotype in zebrafish11. h, Co-injection of ZF TAZ MO (total 2 ng) enhanced slow epiboly of YAP TB KD-injected embryos; control = 89 ± 4.16% (n = 20), YAP KD = 70.09 ± 4.7% (n = 11), YAP/TAZ KD = 52.5 ± 2.64% (n = 10). Error bars show ± s.e.m. P < 0.001, one-way ANOVA. y axis shows percentage epiboly. i, j, TUNEL for cell death and phosphohistone H3 (PH3) antibody staining for cell proliferation (see methods for sample sizes). Stained cells in the neural tube were counted. Error bars indicate ± s.e.m. P < 0.05, P < 0.01, P < 0.001, one-way ANOVA.

Source data

Extended Data Figure 3 Anisotropic enveloping layer cell shape analysis in hir mutants.

a1, Schematic of sectional view of blastoderm margin of a gastrulating embryo (TJ, tight junction; AR actomyosin ring; YSL, yolk syncytial layer; EVL, enveloping layer); a2a6, EVL shape was visualized in phalloidin-stained fixed medaka embryos at 75% epiboly (st. 16, 21 hpf) and compared among, 2 WT (n = 14); 3 hir (n = 9); 4 maternal YAP KD hir mutants (mYAPKDhir) by TB MO-injection into hir embryos (n = 12), 5 MRLC-AA (dominant negative form) mRNA-injected WT (n = 6); and 6, MRLC-DD (constitutive active form) mRNA-injected hir embryos (n = 4). b, EVL shape anisotropy quantification by the length/width ratio (LWR, shown in a2) of marginal EVL cells (up to 4 rows back from the EVL/YSL boundary, shown in Fig. 2d bracket). While EVL shape anisotropy was reduced in hir mutant embryos (a3) to a level comparable to that of MRLC blocked embryos (a5), activation of MRLC in hir (a6) did not rescue it. Parentheses indicate number of cells measured. Scale bar 30 µm. Error bars represent ± s.e.m. P < 0.001, one-way ANOVA.

Source data

Extended Data Figure 4 Flattening of the hir neural tube is associated with string-like cell arrangements.

a, Increasing height (indicated by brackets in a1 and a5) of WT neural tube (outlined, n = 10) was associated with cell stacking. Time in minutes from st. 21 shown bottom left of each sub-panel. Red fluorescent cells, for example, cell 1 in a1, labelled by photo-converting Kaede fluorescent protein, rounded up at the ventricular zone arrowhead in a2 and divided along the ventricular zone (perpendicular cell division in a3) to generate stacked daughter cells 1-1, 1-2, making the neural tube thicker in a5. b, Width/height ratio of spinal cord, measured from time-lapse imaging of single embryos (WT, hir n = 3 each), showed that flattening occurred progressively in hir. Error bars are ± s.e.m. (see Source Data). c, Single-cell tracking of clones (labelled by membrane-GFP and nuclear-RFP) of the growing neural tube at the level of the fifth somite. Lower panels for WT and hir show magnified views of shaded regions in upper panels. The flatter and wider neural tube of the hir mutant at st. 27 was associated with long chain-like cell arrangements (asterisks, bottom panels of hir) tracked from a single neuroepithelial cell at st. 22, compared with the thick cell group generated by cell stacking in WT embryos. Scale bars, 40 µm.

Source data

Extended Data Figure 5 Flattening of the hir neural tube is associated with cell stacking failure.

ad, Single-cell analysis in hir neural tube shows cell stacking failure occurred after mitosis (a, b) and during mitosis (c, d). Neural progenitor cells divided with spindle orientation ‘perpendicular’ or ‘parallel’ to the ventricular zone (‘perpendicular’ or ‘parallel’ cell division, respectively). a, While daughter cells (asterisks) in WT remained stacked after 45 min following perpendicular cell division (first row), those in hir exhibited cell slippage (second and third rows). Telophase neuroepithelial cells in the neural tube, first column; magnified views in second to fourth columns. Dotted lines show division planes. Two types of cell slippage were observed: ventral slippage (VS) where the dorsal daughter cell slipped towards the ventral (second row), and dorsal slippage (DS) where the ventral daughter cell slipped towards the dorsal (third row). After parallel cell division, daughter cells did not change their positions in hir (fourth row). b, Cell stacking was reduced and cell slippage increased after perpendicular cell division, but cells after parallel cell division remained unaltered in hir mutants. Cell numbers in parentheses. Error bars, ± s.e.m. P < 0.05, P < 0.01, t-test (see Source Data). c, During perpendicular mitosis, daughter cells did not stack properly in hir mutants. Cell division orientation (θ) was measured in time-lapse sequences as the acute angle of the telophase cell axis against that of the ventricular zone (for example, dotted line 26° in c). d, Rose diagrams showing frequency and angle of parallel cell divisions. At st. 25–26 (50–54 hpf) perpendicular cell divisions generated stacked cells against gravitational forces in WT (n = 3 embryos at both stages). Far fewer stacked cells were observed in hir (n = 4 embryos at st. 22–24, n = 3 embryos at st. 25–26). These results are illustrated in Fig. 3a. Scale bars, 15 μm in a, 40 µm in c.

Source data

Extended Data Figure 6 Detachment of lens is associated with loss of filopodia in hir.

a, Representative live images of filopodia (arrowheads) from single lens cells (asterisks) expressing Lifeact–GFP in a mosaic manner; a1, WT; a2, hir and a3, 70kDaFN mRNA-injected WT embryos at st. 21.5 when lenses are detaching in hir mutants (see Extended Data Fig. 1b for larger views). a3, Non-mosaic expression of 70kDaFN mRNA in WT embryos was confirmed by co-injected H2A–red fluorescent protein (RFP) in the nucleus (red). L, lens; R, retina. b, Filopodia number per cell was determined (see Extended Data Fig. 7b4 for YAPS87A injected hir embryos). n, number of analysed embryos. Error bars indicate ± s.e.m. P < 0.01, P < 0.001, one-way ANOVA (Extended Data Fig. 6 Source Data). c, Transverse section of integrin-β1 IHC. Strong integrin-β1 localization between lens and retina in st. 22 WT (n = 2) (c1, arrowhead); no such localization in hir (n = 3) (c2). At st. 23 in hir (n = 3), weak localization where rounded up lens reattached to retina (c2′, arrowhead). Scale bars, 10 µm in a; 40 µm in c.

Source data

Extended Data Figure 7 The hir mutation acts cell non-autonomously.

a, Mosaic expression of EGFP–YAPS87A by mRNA injection at 16-cell stage in hir mutant embryos rescued the hir eye phenotype in a2 compared to a1 (WT) and a3 (hir). The boxed area in a2 is magnified in the lower panels (a2′a2′′′) fluorescence, merged and bright-field views, respectively. Arrowheads in a2′ indicate EGFP–YAPS87A-expressing clones. b, Non-cell autonomous rescue of filopodia in hir mutant lens cells. YAPS87A+ mCherry-CAAX (labels membrane red) mRNA, and Lifeact-EGFP mRNA (labels F-actin green) were injected into different cells at 8–16 cell stage. b1, In the invaginated (arrow) hir mutant lens (boxed area magnified in b2 and b3, n = 10) rescued by mosaic expression of YAPS87A (red), YAPS87A non-expressing mutant cells recovered filopodia (arrowheads in b4, magnified view of b3). Filopodia number/cell was compared between WT and hir in Extended Data Fig. 6b. c1, Cells from donor embryos injected with rhodamine (red, top left) were transplanted to a recipient embryo (top right, blastula stage st. 12) at the location fated to be eyes (bottom, animal pole view). c2, WT; c3, hir and c4, WT cells transplanted into hir mutant eye, causing the lens (arrowhead) to invaginate into the retina as in WT at st. 23 (note that this confocal sectional view represents a fraction of transplanted cells in the whole eye, see Supplementary Table 4 for the frequency of rescue). Scale bars, 40 µm.

Extended Data Figure 8 F-actin and FN localizations in hir.

a, Whole-mount imaging of WT (n = 5) and hir (n = 4) embryos stained for F-actin (red) and FN (green). a1, a1′, Whole dorsal view of embryos anterior up, only FN shown; a2a4, a2′a4′, magnified view of area indicated by asterisks in a1, a1′; merged a2, a2′, F-actin a3, a3′ and FN a4, a4′. Arrowheads indicate cortical F-actin and FN fibrils in WT and corresponding region in hir (a3, a4, a3′, a4′); arrows show ectopic F-actin aggregates and aberrant FN fibrils in a3′, a4′. b, Immunostaining of 2D cultured RPE1 cells transfected with control (Cont, n = 21) and YAP siRNAs (n = 19) stained with Phalloidin (b1, b1′), β-catenin (b2, b2′) and merged with DAPI (b3, b3′); phalloidin (b4, 4′), FN (b5, b5′) and merged with DAPI (b6, b6′). In marked contrast to the 3D spheroids, FN deposits were not altered in YAP KD cells (b5, b5′) despite increased F-actin stress fibres (b1, b1′ and b4, b4′). c, The medaka fku mutants exhibit lens dislocation (arrows). Live dorsal view of the head of c1, WT; c2, fku and c3, hir mutant embryos at st. 24. c4, The fku mutation was mapped to LG21 to the region encompassing the FN1 gene (0 recombinants/1,130 meiosis). Positional cloning identified a non-sense mutation of Glu593 (GAA to TAA) in FN1 (2,503 amino acids). FN1 morpholino KD in WT embryos mimicked the fku mutant phenotype. d, Constitutive-active MRLC-DD mRNA markedly increased body thickness of WT embryos, but did not rescue the flattened body (brackets in lower panels) and dislocated lens phenotypes of hir (n = 48). Upper panels, live lateral view (insets, dorsal views of left eyes); lower panels, frontal sections stained with phalloidin (red) and TO-PRO-3 (blue) at st. 25. Scale bars 30 μm, except a2, 15 μm and b1, 50 μm.

Extended Data Figure 9 in vivo analysis of ARHGAP18 function.

a, Quantitative RT–PCR analysis showed that ARHGAP18 mRNA expression in the hir mutant is significantly reduced to 76% of WT level. EF1α used as an internal control. Data are shown as means ± s.e.m. (n = 10 each; P < 0.001 Student’s t-test (two-tailed)). b, myrARHGAP18 mRNA (150 pg) injection rescued the hir phenotype (21 rescued/39 hir/112 survived embryos). Upper panels, live dorsal view; lower panels, frontal sections stained with phalloidin (red) and TO-PRO-3 (blue) at st. 23; b1, uninjected hir, b2, injected hir and b3, WT. The lens (asterisk) invaginated into retina (arrows, upper panel) and the neural tube became thicker (brackets in lower panels) in the myrARHGAP18 mRNA-injected hir mutant embryos. b2′, FN staining of myrARHGAP18 mRNA-injected hir mutant embryos; boxed area magnified in subsequent panel to the right; invaginated lenses had fine FN fibrils (arrowheads) between lens and retina as in WT (see Fig. 3b1′′). c, Phylogenetic analysis identified 16 ARHGAP18 paralogues in vertebrate lineages. Arrowheads show medaka orthologues. d, siRNA screening of 40 human ARHGAP genes in HeLa cells showed that KD of five ARHGAP genes exhibited the rounding up phenotype similar to ARHGAP18 inactivation. Scale bars, 30 µm.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-6, 2 Supplementary Discussions and Supplementary Figure 1. (PDF 537 kb)

Video 1: Formation of the eye by coordinated invagination of the lens and retina in WT

Dorsal bright-field view, anterior up, between st.19 and st.23 (14 h duration). In WT, the nascent lenses and retina undergo coordinated morphogenesis to locate the lens properly in the eye. (MOV 16738 kb)

Video 2: Dislocation of the lens in hir mutants

Dorsal bright-field view, anterior up, between st.20 and st.24 (17 h duration). The mutant lens placodes dislocate, round up and migrate anteriorly where they loosely reattach to the retina. (MOV 13956 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

Porazinski, S., Wang, H., Asaoka, Y. et al. YAP is essential for tissue tension to ensure vertebrate 3D body shape. Nature 521, 217–221 (2015). https://doi.org/10.1038/nature14215

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14215

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