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.

  • Article
  • Published:

Crumbs2 promotes cell ingression during the epithelial-to-mesenchymal transition at gastrulation

Abstract

During gastrulation of the mouse embryo, individual cells ingress in an apparently stochastic pattern during the epithelial-to-mesenchymal transition (EMT). Here we define a critical role of the apical protein Crumbs2 (CRB2) in the gastrulation EMT. Static and live imaging show that ingressing cells in Crumbs2 mutant embryos become trapped at the primitive streak, where they continue to express the epiblast transcription factor SOX2 and retain thin E-cadherin-containing connections to the epiblast surface that trap them at the streak. CRB2 is distributed in a complex anisotropic pattern on apical cell edges, and the level of CRB2 on a cell edge is inversely correlated with the level of myosin IIB. The data suggest that the distributions of CRB2 and myosin IIB define which cells will ingress, and we propose that cells with high apical CRB2 are basally extruded from the epiblast by neighbouring cells with high levels of apical myosin.

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: Crumbs2 is required for mammalian gastrulation.
Figure 2: Crumbs2 promotes cell ingression at the primitive streak.
Figure 3: Live imaging defines defects in cell ingression in Crumbs2 mutants.
Figure 4: Streak-autonomous and non-cell-autonomous activities of Crumbs2.
Figure 5: CRB2 enrichment at the primitive streak.
Figure 6: CRB2 regulates the distribution of myosin IIB.
Figure 7: CRB2 is required for epiblast integrity only when cells delaminate at the streak.

Similar content being viewed by others

References

  1. Shook, D. & Keller, R. Mechanisms, mechanics and function of epithelial-mesenchymal transitions in early development. Mech. Dev. 120, 1351–1383 (2003).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  3. Williams, M., Burdsal, C., Periasamy, A., Lewandoski, M. & Sutherland, A. Mouse primitive streak forms in situ by initiation of epithelial to mesenchymal transition without migration of a cell population. Dev. Dynam. 241, 270–283 (2012).

    Article  Google Scholar 

  4. Voiculescu, O., Bodenstein, L., Lau, I. J. & Stern, C. D. Local cell interactions and self-amplifying individual cell ingression drive amniote gastrulation. eLife 3, e01817 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Herion, N. J., Salbaum, J. M. & Kappen, C. Traffic jam in the primitive streak: the role of defective mesoderm migration in birth defects. Birth Defects Res. A Clin. Mol. Teratol. 100, 608–622 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Thiery, J. P., Acloque, H., Huang, R. Y. & Nieto, M. A. Epithelial-mesenchymal transitions in development and disease. Cell 139, 871–890 (2009).

    Article  CAS  PubMed  Google Scholar 

  7. Scheel, C. & Weinberg, R. A. Cancer stem cells and epithelial-mesenchymal transition: concepts and molecular links. Semin. Cancer Biol. 22, 396–403 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Arnold, S. J. & Robertson, E. J. Making a commitment: cell lineage allocation and axis patterning in the early mouse embryo. Nat. Rev. Mol. Cell Biol. 10, 91–103 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Acloque, H. et al. Reciprocal repression between Sox3 and snail transcription factors defines embryonic territories at gastrulation. Dev. Cell 21, 546–558 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tepass, U., Theres, C. & Knust, E. Crumbs encodes an EGF-like protein expressed on apical membranes of Drosophila epithelial cells and required for organization of epithelia. Cell 61, 787–799 (1990).

    Article  CAS  PubMed  Google Scholar 

  11. Tepass, U. Crumbs, a component of the apical membrane, is required for zonula adherens formation in primary epithelia of Drosophila. Dev. Biol. 177, 217–225 (1996).

    Article  CAS  PubMed  Google Scholar 

  12. van de Pavert, S. A. et al. Crumbs homologue 1 is required for maintenance of photoreceptor cell polarization and adhesion during light exposure. J. Cell. Sci. 117, 4169–4177 (2004).

    Article  PubMed  Google Scholar 

  13. Whiteman, E. L. et al. Crumbs3 is essential for proper epithelial development and viability. Mol. Cell. Biol. 34, 43–56 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Xiao, Z. et al. Deficiency in Crumbs homolog 2 (Crb2) affects gastrulation and results in embryonic lethality in mice. Dev. Dynam. 240, 2646–2656 (2011).

    Article  CAS  Google Scholar 

  15. Ramkumar, N. et al. Protein O-Glucosyltransferase 1 (POGLUT1) promotes mouse gastrulation through modification of the apical polarity protein CRUMBS2. PLoS Genet. 11, e1005551 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Hadjantonakis, A. K., Cox, L. L., Tam, P. P. & Nagy, A. An X-linked GFP transgene reveals unexpected paternal X-chromosome activity in trophoblastic giant cells of the mouse placenta. Genesis 29, 133–140 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Hayashi, S., Lewis, P., Pevny, L. & McMahon, A. P. Efficient gene modulation in mouse epiblast using a Sox2Cre transgenic mouse strain. Mech. Dev. 119, S97–S101 (2002).

    Article  PubMed  Google Scholar 

  18. Perantoni, A. O. et al. Inactivation of FGF8 in early mesoderm reveals an essential role in kidney development. Development 132, 3859–3871 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Rhee, J. M. et al. In vivo imaging and differential localization of lipid-modified GFP-variant fusions in embryonic stem cells and mice. Genesis 44, 202–218 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Röper, K. Anisotropy of Crumbs and aPKC drives myosin cable assembly during tube formation. Dev. Cell 23, 939–953 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Letizia, A., Ricardo, S., Moussian, B., Martín, N. & Llimargasm, M. A functional role of the extracellular domain of Crumbs in cell architecture and apicobasal polarity. J. Cell. Sci. 126, 2157–2163 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Zou, J., Wang, X. & Wei, X. Crb apical polarity proteins maintain zebrafish retinal cone mosaics via intercellular binding of their extracellular domains. Dev. Cell 22, 1261–1274 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tepass, U. The apical polarity protein network in Drosophila epithelial cells: regulation of polarity, junctions, morphogenesis, cell growth, and survival. Annu. Rev. Cell Dev. Biol. 28, 655–685 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Rozbicki, E. et al. Myosin-II-mediated cell shape changes and cell intercalation contribute to primitive streak formation. Nat. Cell Biol. 17, 397–408 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Liu, P. et al. Requirement for Wnt3 in vertebrate axis formation. Nat. Genet. 22, 361–365 (1999).

    Article  CAS  PubMed  Google Scholar 

  26. Ciruna, B. & Rossant, J. FGF signaling regulates mesoderm cell fate specification and morphogenetic movement at the primitive streak. Dev. Cell 1, 37–49 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Fletcher, G. C., Lucas, E. P., Brain, R., Tournier, A. & Thompson, B. J. Positive feedback and mutual antagonism combine to polarize Crumbs in the Drosophila follicle cell epithelium. Curr. Biol. 22, 1116–1122 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Ishiuchi, T. & Takeichi, M. Willin and Par3 cooperatively regulate epithelial apical constriction through aPKC-mediated ROCK phosphorylation. Nat. Cell Biol. 13, 860–866 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Fernandez-Gonzalez, R., Simoes Sde, M., Röper, J. C., Eaton, S. & Zallen, J. A. Myosin II dynamics are regulated by tension in intercalating cells. Dev. Cell 17, 736–743 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Slattum, G., McGee, K. M. & Rosenblatt, J. P115 RhoGEF and microtubules decide the direction apoptotic cells extrude from an epithelium. J. Cell. Biol. 186, 693–702 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Slattum, G. M. & Rosenblatt, J. Tumour cell invasion: an emerging role for basal epithelial cell extrusion. Nat. Rev. Cancer 14, 495–501 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Campbell, K., Knust, E. & Skaer, H. Crumbs stabilises epithelial polarity during tissue remodelling. J. Cell Sci. 122, 2604–2612 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ebarasi, L. et al. Defects of CRB2 cause steroid-resistant nephrotic syndrome. Am. J. Hum. Genet. 96, 153–161 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Slavotinek, A. et al. CRB2 mutations produce a phenotype resembling congenital nephrosis, Finnish type, with cerebral ventriculomegaly and raised α-fetoprotein. Am. J. Hum. Genet. 96, 162–169 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Grande, M. T. et al. Snail1-induced partial epithelial-to-mesenchymal transition drives renal fibrosis in mice and can be targeted to reverse established disease. Nat. Med. 21, 989–997 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Lovisa, S. et al. Epithelial-to-mesenchymal transition induces cell cycle arrest and parenchymal damage in renal fibrosis. Nat. Med. 21, 998–1009 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Arcila, M. E. et al. MAP2K1 (MEK1) mutations define a distinct subset of lung adenocarcinoma associated with smoking. Clin. Cancer Res. 21, 1935–1943 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Beltran, H. et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat. Med. 22, 298–305 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  41. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, p11 (2013).

    Google Scholar 

  42. Alves, C. H. et al. Loss of CRB2 in the mouse retina mimics human retinitis pigmentosa due to mutations in the CRB1 gene. Hum. Mol. Genet. 22, 35–50 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Barrow, J. R. et al. Ectodermal Wnt3/β-catenin signaling is required for the establishment and maintenance of the apical ectodermal ridge. Genes Dev. 17, 394–409 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mehalow, A. K. et al. CRB1 is essential for external limiting membrane integrity and photoreceptor morphogenesis in the mammalian retina. Hum. Mol. Genet. 12, 2179–2189 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Muzumdar, M. D., Tasic, B., Miyamichi, K., Li, L. & Luo, L. A global double-fluorescent Cre reporter mouse. Genesis 45, 593–605 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Lakso, M. et al. Efficient in vivo manipulation of mouse genomic sequences at the zygote stage. Proc. Natl Acad. Sci. USA 93, 5860–5865 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lee, J. D., Silva-Gagliardi, N. F., Tepass, U., McGlade, C. J. & Anderson, K. V. The FERM protein Epb4.1l5 is required for organization of the neural plate and for the epithelial-mesenchymal transition at the primitive streak of the mouse embryo. Development 134, 2007–2016 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Laprise, P. et al. The FERM protein Yurt is a negative regulatory component of the Crumbs complex that controls epithelial polarity and apical membrane size. Dev. Cell 11, 363–374 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Franci, C. et al. Expression of Snail protein in tumor-stroma interface. Oncogene 25, 5134–5144 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Lemmers, C. et al. hINADl/PATJ, a homolog of discs lost, interacts with crumbs and localizes to tight junctions in human epithelial cells. J. Biol. Chem. 277, 25408–25415 (2002).

    Article  CAS  PubMed  Google Scholar 

  51. 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 

  52. Migeotte, I., Grego-Bessa, J. & Anderson, K. V. Rac1 mediates morphogenetic responses to intercellular signals in the gastrulating mouse embryo. Development 138, 3011–3020 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Silva, J. et al. Promotion of reprogramming to ground state pluripotency by signal inhibition. PLoS Biol. 6, e253 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the MSKCC Molecular Cytology and Mouse Genetics Core Facilities for valuable technical support. We thank the Hadjantonakis laboratory for the X-linked GFP and GFP-GPI strains. We thank M. Lewandoski for Brachyury-Cre mice. We thank J. Zallen, A.-K. Hadjantonakis, A. Hall, I. Migeotte, H. Kakkar and members of the Anderson laboratory for their helpful suggestions. The work was supported by NIH R37 HD03455 to K.V.A. and the MSKCC Cancer Center Support Grant (P30 CA008748).

Author information

Authors and Affiliations

Authors

Contributions

N.R. carried out the experiments; T.O. participated in and analysed the live imaging experiments. N.F.S.-G., C.J.M. and J.W. provided essential reagents and advice. N.R. and K.V.A. designed experiments, analysed the data and wrote the paper.

Corresponding author

Correspondence to Kathryn V. Anderson.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Expression of Crumbs2 RNA and CRB2 protein in the epiblast.

(A) Whole mount in situ hybridization for Crumbs2 RNA expression in a wild-type E7.5 embryo, n = 6 wild type embryos. (B,C) Transverse section through the wild-type embryo at E7.5 (B) and E7.75 (C) after in situ hybridization for Crumbs2, showing its apical localization and posterior enrichment in the epiblast. Apical enrichment of Crumbs RNA has also been seen in Drosophila follicle cells54. (D,D ) Single optical section from whole mount E6.75 embryo immunostained for CRB2 (green) and E-cadherin (red), showing the posterior enrichment of CRB2 (D) and (D ) its sub-cellular localization apical to E-cadherin in a higher magnification view of the boxed region in (D), n = 7 wild type embryos. CRB2 was localized to an apical domain of the epiblast that overlapped with apical F-actin and in caps apical to the adherens junctions (see also Fig. 6b, b). Neither Crumbs2 RNA nor CRB2 protein was detected in the wild-type endoderm or mesoderm. Scale bars AC, 75 μm; D,D , 50 μm. The images shown here are representative.

Supplementary Figure 2 Distinct gastrulation defects in Crumbs2 and Rac1 mutants.

(A) Single optical transverse sections of E8.0 wild type and Crumbs2 mutant embryos immunostained for Laminin (green) and E-cadherin (red), showing broader streak (marked by the break in Laminin) and accumulation of E-cadherin positive cells in the mutant streak, n ≥ 15 mutant embryos. (B) Single optical section from transverse section of E7.75 wild-type and Rac1 epiblast-specific deletion embryos stained for SNAIL1, Phalloidin and Laminin, showing the accumulation of SNAIL1-expressing cells in the Rac1 conditional primitive streak, n = 3 mutant embryos. Scale bars A, 50 μm; B, 20 μm.

Supplementary Figure 3 Mesoderm defects in epiblast-specific deletion of Crumbs2.

(A) In situ hybridization shows the expression of Brachyury, Meox1 and Uncx4.1 at E8.5, n ≥ 3 embryos per genotype. Brachyury expression shows the reduced number of axial mesoderm cells in the discontinuous midline of the mutant. Meox1 expression shows reduction in paraxial mesoderm in the mutant. Striped Uncx4.1 expression shows that somitogenesis clock is active although the mutants do not form normal somites. (B) The progression of Brachyury (T) expression in the axial mesoderm, visualized by whole mount immunostaining, n ≥ 3 mutant embryos per stage. The T expression pattern appears relatively normal at E7.5; after that time it appears that the axial mesoderm extends in the anterior-posterior axis but new cells fail to be added to the axial mesoderm, leading to a thin, broken line of Brachyury-positive cells by E8.5. Scale bar A, 150 μm; B, 110 μm. The images shown here are representative.

Supplementary Figure 4 Crumbs2 chimeric embryos.

(A) Whole mount image of a low contribution chimera that resembled an E8.5 wild-type embryo, n = 20 chimeric embryos. (B) Whole mount image of a high contribution chimera that recapitulated the phenotype of Crumbs2 mutants, n = 9 chimeric embryos. Percentage chimerism was determined in sections. (C) En face view of the anterior epiblast/early neural epithelium of chimeric embryos at E8.5 immunostained for CRB2 (red) and β-catenin (gray), n = 5 chimeric embryos. Mutant cells are GFP-positive (green). Arrows point to the lack of CRB2 expression in wild-type cells at the edges shared with mutant cells, whereas β-catenin expression is maintained at these edges. Scale bar in A,B, 150 μm; C, 10 μm. The images shown here are representative.

Supplementary Figure 5 Geometry and distribution of apical CRB2 in cell edges of the primitive streak region.

Histograms of cell shape distributions at the primitive streak. (A) Cells at the primitive streak have a variety of cell shapes; the most common cells have 4–6 edges. (B) Percentage of the polygons with 3, 4, 5 or 6 edges, and the number of their edges enriched with CRB2, documenting the nature of its anisotropic distribution at the streak. n = 3 embryos, 50 cells per embryo; total = 176 cells.

Supplementary Figure 6 Localization of PatJ, a Crumbs complex protein, Crumbs2 and Myosin IIB in the early embryo.

(A) Extended projection en face view of immunostaining for PatJ and ZO1, n ≥ 5 wild type embryos. In the streak region of an E8.0 wild-type embryo. PatJ is distributed anisotropically on cell edges, with some strong apical puncta, and a complex pattern of enrichment at cell edges, similar to the pattern of CRB2 expression at the streak (Fig. 7c). (B) PatJ is not detectable at the streak of Crumbs2 or Poglut1wsnp mutants, n ≥ 3 mutant embryos per genotype. (C,D) Reciprocal localization of Crumb2 and Myosin IIB in the early embryo and in the epiblast adjacent to the streak. (C) Extended projection view of wild-type streak at early bud stage (E7.0). Note the striking mulitcellular rosette with high Myosin IIB at the vertex (arrow), which could have surrounded an extruded cell. Also note cells with constricted apical surfaces with high CRB2 surrounded by high myosin (yellow arrows). (D) CRB2 (red) and Myosin IIB (green) are anisotropically distributed in the side epiblast of wild type (B) at E8.5. Proximal is up, n ≥ 5 wild type embryos per stage (C,D). Scale bar in A,B, 25 μm; C,D, 20 μm. The images shown here are representative.

Supplementary Figure 7 Crumbs2 phenotypes in the E7.75-E8.0 anterior epiblast.

(A) Transverse sections through E8.0 wild-type and Crumbs2 embryos immunostained for pHH3 (mitotic cells) and acetylated-tubulin. Anterior is to the left. Arrows point to examples of non-apical pHH3 positive nuclei. The mitotic indices were not significantly different (WT = 9.924 ± 0.9685 (mean ± s.e.m.), n = 3 embryos (2 sections/embryo) and Crumbs2 mutant = 7.002 ± 0.5897 (mean ± s.e.m.), n = 3 embryos (2–3 sections/embryo), P-value = 0.02, not significant), although the percentage of non-apical mitoses (arrows) was significantly higher in the mutants; non-apical mitosis in wild type = 2.6% and Crumbs2 mutants = 23.5%. Scale bar, 40 μm. (B) Transverse sections through the epiblast of E7.75 wild type and Crumbs2−/−mutant embryos immunostained for aPKCλ and Par3 (green), n = 3 embryos per genotype (2–3 sections per embryo); apical markers are localized correctly in the mutants despite the thicker epiblast layer (DAPI, blue). Scale bars, A, 40 μm; B, 10 μm. The images shown here are representative.

Supplementary information

Supplementary Information

Supplementary Information (PDF 948 kb)

Wild-type GFP streak cell translocates from the apical to the basal side of the epiblast.

The epiblast cells randomly expressing GFP in the mouse embryo at E7.5 are imaged from the primitive streak side. A 3D rendered surface (yellow) is built for visualization of the individual cell shape changes. During ingression basal protrusions are formed, the cell body translocates basally and the cell leaves the epiblast in less than 2 h. Note apical process retraction. Time is h:min. Scale bar, 10 μm. (MOV 4161 kb)

Another wild-type GFP streak cell ingresses from the apical to the basal side of the epiblast.

The cell with basal protrusions constricts its apical membrane, moves toward basal plane and exits the epiblast. Time is h:min. Scale bar, 6 μm. (MOV 2943 kb)

Cell shape dynamics of wild-type GFP streak cell during ingression.

The ingressing cell shape highlighted by the 3D rendered surface is highly dynamic. During translocation, the initial cigar-shaped cell body changes to more discoid shape. Multiple protrusions accompanied the ingression. Time is h:min. Scale bar, 10 μm. (MOV 4447 kb)

Mutant GFP streak cell does not translocate from the apical to the basal side of the epiblast.

The epiblast cells randomly expressing GFP in the mouse mutant embryo at E7.5 were imaged from the primitive streak side. A 3D rendered cell surface shows the bottle-like shape of a Crumbs2−/− cell. During the imaging time, basal protrusions are formed and the cell body is located basally, but the cell does not leave the epiblast within 2 h. Note apical process does not retract. Time is h:min. Scale bar, 10 μm. (MOV 4621 kb)

Another mutant GFP streak cell does not ingress from the apical side of the epiblast.

During the time-lapse observation a 3D rendered cell (yellow) maintains its bottle-like shape. Note failure of the apical surface of the cell to retract highlighted by black plane in the upper part of the box. Time is h:min. Scale bar, 10 μm. (MOV 3630 kb)

A group of Crumbs2 null mutant streak cells fail to ingress remaining attached to the apical side of the epiblast.

Bottle-like shape of Crumbs2 null mutant cell with long thin apical extensions at the streak is persistent within multiple cells. Crumbs2 mutant cells accumulate at the streak. Note first frames of the video show cells without rendered cell surfaces (green). Time is h:min. Scale bar, 15 μm. (MOV 5644 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ramkumar, N., Omelchenko, T., Silva-Gagliardi, N. et al. Crumbs2 promotes cell ingression during the epithelial-to-mesenchymal transition at gastrulation. Nat Cell Biol 18, 1281–1291 (2016). https://doi.org/10.1038/ncb3442

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3442

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