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:

The snail superfamily of zinc-finger transcription factors

Key Points

  • Snail family members are thought to act as transcriptional repressors. The zinc fingers correspond to the C2H2 type (cysteine/histidine) and bind to a site that contains six bases, CAGGTG. This motif is identical to the E box, the core-binding site of basic helix–loop–helix transcription factors.

  • Snail homologues have been found in many species including humans, other vertebrates, non-vertebrate chordates (ascidians and Amphioxus), insects, nematodes, annelids and molluscs.

  • They constitute a superfamily that can be subdivided into two related but independent groups: the Snail and the Scratch families. Independent duplication events of one Snail and one Scratch gene that were present in the metazoan ancestor, gave rise to three genes of each group in Drosophila and two for each group in most vertebrates.

  • They show a conserved function in mesoderm development from flies to mammals, and vertebrate family members participate in the specification and migration of the neural crest. The role in delamination and migration is mediated by the triggering of the epithelial–mesenchymal transition (EMT), a phenotypic change that renders epithelial cells migratory and invasive.

  • Snail induces EMT by directly repressing the transcription of E-cadherin.

  • EMT processes also occur during the malignant conversion of epithelial tumours, and pathological activation of Snail has been shown at the invasive front of chemically induced mouse skin tumours and in human breast carcinomas.

  • The expression of the two vertebrate family members, Snail and Slug, diverges at the sites of EMT in different species. Whereas in the chick, Slug is expressed in the premigratory neural crest and the primitive streak, and Snail is absent from these tissues, in the mouse embryo, Snail is expressed in these cells. Expression in zebrafish and Xenopus is more similar to that in the mouse. Interesting questions emerge, such as functional equivalence and the evolutionary mechanisms that led to those differences.

  • Different signalling pathways —TGF-β, BMP, FGF and Wnt — have been implicated in the induction of Snail family members during the process of EMT.

  • Different superfamily members (scratch in C. elegans and Slug in mammals) are involved in the regulation of cell death and survival.

  • Escargot and mouse Snail are involved in the control of polyploidy in several tissues, including imaginal discs cells in Drosophila, mouse trophoblast cells and human megakaryocytes.

  • A deficiency in the three Drosophila Snail family members leads to an inappropriate subcellular localization of determinants that are crucial for cell-fate decisions during asymmetric cell division.

  • An ancestral function in the development of sensory and/or neuronal structures is proposed for the whole superfamily, which includes both the Snail and Scratch genes. An additional ancestral function in the control of cell death/survival is also proposed.

  • The role of EMT seems to be exclusively associated with the members of the Snail family with representatives analysed in lophotrochozoans, ecdysozoans and deuterostomes. This role has been co-opted for cell migration during mesoderm and neural-crest formation, and tumour progression, when these processes emerged.

Abstract

The Snail superfamily of zinc-finger transcription factors is involved in processes that imply pronounced cell movements, both during embryonic development and in the acquisition of invasive and migratory properties during tumour progression. Different family members have also been implicated in the signalling cascade that confers left–right identity, as well as in the formation of appendages, neural differentiation, cell division and cell survival.

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: Phylogenetic tree of the Snail superfamily.
Figure 2: Sequence comparison of the main conserved domains and consensus sequences for the individual zinc fingers of the Snail superfamily.
Figure 3: Expression of Snail family members in Drosophila, amphioxus, chick and mouse embryos.
Figure 4: Snail genes occupy a central position in triggering EMT in physiological and pathological situations.
Figure 5: Different genetic pathways involving Snail function.

Similar content being viewed by others

References

  1. Wolpert, L. Quoted in Slack, J. M. W. From Egg to Embryo: Determinative Events in Early Development, Vol. 1 (Cambridge Univ. Press, Cambridge, UK, 1986).

    Google Scholar 

  2. Alberga, A., Boulay, J. L., Kempe, E., Dennefield, C. & Haenlin, M. The snail gene required for mesoderm formation in Drosophila is expressed dynamically in derivatives of all three germ layers. Development 111, 983–992 (1991).

    CAS  PubMed  Google Scholar 

  3. Nieto, M. A., Bennet, M. F., Sargent, M. G. & Wilkinson, D. G. Cloning and developmental expression of Sna, a murine homologue of the Drosophila snail gene. Development 116, 227–237 (1992).

    CAS  PubMed  Google Scholar 

  4. Smith, D. E., Del Amo, F. F. & Gridley, T. Isolation of Sna, a mouse gene homologous to the Drosophila genes snail and escargot: its expression pattern suggests multiple roles during postimplantation development. Development 116, 1033–1039 (1992).

    CAS  PubMed  Google Scholar 

  5. Hammerschmidt, M. & Nüsslein-Volhard, C. The expression of a zebrafish gene homologous to Drosophila snail suggests a conserved function in invertebrate and vertebrate gastrulation. Development 119, 1107–1118 (1993).

    CAS  PubMed  Google Scholar 

  6. Nieto, M. A., Sargent, M., Wilkinson, D. G. & Cooke, J. Control of cell behaviour during vertebrate development by Slug, a zinc finger gene. Science 264, 835–839 (1994).The isolation of the first Slug gene and its involvement in delamination of the neural crest and of the early mesoderm.

    Article  CAS  PubMed  Google Scholar 

  7. Langeland, J. A., Tomsa, J. M., Jackman, W. R. Jr & Kimmel, C. B. An amphioxus snail gene: expression in paraxial mesoderm and neural plate suggests a conserved role in patterning the chordate embryo. Dev. Genes Evol. 208, 569–577 (1998).

    Article  CAS  PubMed  Google Scholar 

  8. Wada, S. & Saiga, H. Cloning and embryonic expression of Hrsna, a snail family gene of the ascidian Halocynthia roretzi: implication in the origins of mechanisms for mesoderm specification. Dev. Growth Differ. 41, 9–18 (1999).

    Article  CAS  PubMed  Google Scholar 

  9. Mayor, R., Guerrero, R., Young, R. M., Gomez-Skarmeta, J. L. & Cuellar, C. A novel function for the Xslug gene: control of dorsal mesendoderm development by repressing BMP-4. Mech. Dev. 97, 47–56 (2000).

    Article  CAS  PubMed  Google Scholar 

  10. 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).This study shows that Snail -mutant mice die at gastrulation owing to defective EMT.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Thisse, C., Thisse, B. & Postlethwait, J. H. Expression of snail2, a second member of the zebrafish snail family, in cephalic mesendoderm and presumptive neural crest of wild-type and spadetail mutant embryos. Dev. Biol. 172, 86–99 (1995).

    Article  CAS  PubMed  Google Scholar 

  12. Mancilla, A. & Mayor, R. Neural crest formation in Xenopus laevis: mechanisms of Slug induction. Dev. Biol. 177, 580–589 (1996).

    Article  CAS  PubMed  Google Scholar 

  13. Carl, T. F., Dufton, C., Hanken, J. & Klymkowsky, M.W. Inhibition of neural crest migration in Xenopus using antisense slug RNA. Dev. Biol. 213, 101–115 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. LaBonne, C. & Bronner-Fraser, M. Snail-related transcriptional repressors are required in Xenopus for both the induction of the neural crest and its subsequent migration. Dev. Biol. 221, 195–205 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Cano, A. et al. The transcription factor snail controls epithelial–mesenchymal transitions by repressing E-cadherin expression. Nature Cell Biol. 2, 76–83 (2000).This study shows that Snail triggers EMT through the repression of E-cadherin transcription. This also shows the correlation of Snail expression with the invasive phenotype in cell lines and in tumours induced in the skin of the mouse.

    Article  CAS  PubMed  Google Scholar 

  16. Batlle, E. et al. The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nature Cell Biol. 2, 84–89 (2000).The demonstration that Snail is a direct repressor of E-cadherin expression in tumour cells.

    Article  CAS  PubMed  Google Scholar 

  17. Hay, E. D. An overview of epithelio-mesenchymal transformation. Acta Anat. 154, 8–20 (1995).

    Article  CAS  PubMed  Google Scholar 

  18. Hemavathy, K., Ashraf, S. I. & Ip, Y. T. Snail/Slug family of repressors: slowly going to the fast lane of development and cancer. Gene 257, 1–12 (2000).

    Article  CAS  PubMed  Google Scholar 

  19. Gans, C. & Northcutt, R. G. Neural crest and the evolution of vertebrates: a new head. Science 220, 268–274 (1983).

    Article  CAS  PubMed  Google Scholar 

  20. Grau, Y., Carteret, C. & Simpson, P. Mutations and chromosomal rearrangements affecting the expression of snail, a gene involved in embryonic patterning in Drosophila melanogaster. Genetics 108, 347–360 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Nusslein-Volhard, C., Weischaus, E. & Kluding, H. Mutations affecting the pattern of the larval cuticle in Drosophila melanogaster. I. Zygotic loci on the second chromosome. Wilheim Roux's Arch. Dev. Biol. 193, 267–282 (1984).References 20 and 21 simultaneously described the first Snail family member.

    Article  CAS  Google Scholar 

  22. Knight, R. & Shimeld, S. Identification of conserved C2H2 zinc-finger gene families in the Bilateralia. Genome Biol. 2, research0016.1–0016.8 (2001).

    Google Scholar 

  23. Mauhin, V., Lutz, Y., Dennefeld, C. & Alberga, A. Definition of the DNA-binding site repertoire for the Drosophila transcription factor SNAIL. Nucleic Acids Res. 21, 3951–3957 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Fuse, N., Hirose, S. & Hayashi, S. Diploidy of Drosophila imaginal cells is maintained by a transcriptional repressor encoded by escargot. Genes Dev. 8, 2270–2281 (1994).The first indication of a Snail family member that is involved in endoreduplication.

    Article  CAS  PubMed  Google Scholar 

  25. Inukai, T. et al. Slug, a ces-1-related zinc finger transcription factor gene with antiapoptotic activity is a downstream target of the E2A-HLF oncoprotein. Mol. Cell 4, 343–352 (1999).This study provides a description of the anti-apoptotic effect of Slug in humans.

    Article  CAS  PubMed  Google Scholar 

  26. Kataoka, H. et al. A novel snail-related transcription factor Smuc regulates basic helix–loop–helix transcription factor activities via specific E-box motifs. Nucleic Acids Res. 28, 626–633 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nakayama, H., Scott, I. C. & Cross, J. C. The transition to endoreduplication in trophoblast giant cells is regulated by the mSna zinc finger transcription factor. Dev. Biol. 199, 150–163 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Pérez-Moreno, M. et al. A new role for E12/E47 in the repression of E-cadherin expression and epithelial–mesenchymal transition. J. Biol. Chem. 276, 27424–27431 (2001).

    Article  PubMed  Google Scholar 

  29. Fijiwara, S., Corbo, J. C. & Levine, M. The snail repressor establishes a muscle/notochord boundary in the Ciona embryo. Development 125, 2511–2520 (1998).

    Google Scholar 

  30. Hemavathy, K., Guru, S. C., Harris, J., Chen, J. D. & Ip, Y. T. Human Slug is a repressor that localizes to sites of active transcription. Mol. Cell. Biol. 20, 5087–5095 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Grimes, H. L., Chan, T. O., Zweidler-McKay, P. A., Tong, B. & Tsichlis, P. N. The Gfi1 proto-oncoprotein contains a novel transcriptional repressor domain, SNAG, and inhibits G1 arrest induced by inteleukin-2 withdrawal. Mol. Cell. Biol. 16, 6263–6272 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lespinet, O. et al. Characterization of two snail genes in the gastropod mollusk Patella vulgata. Implications for understanding the ancestral function of the snail–related genes in Bilateralia. Submitted.This study links Snail genes with EMT and left–right asymmetry in molluscs.

  33. Roark, M., Sturtevant, M. A., Emery, J., Vaessin, H. & Grell, E. scratch, a pan-neural gene encoding a zinc finger protein related to snail, promotes neural development. Genes Dev. 9, 2384–2398 (1995).

    Article  CAS  PubMed  Google Scholar 

  34. Nibu, Y. et al. dCtBP mediates transcriptional repression by Knirps, Kruppel and Snail in the Drosophila embryo. EMBO J. 17, 7009–7020 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nakakura, E. K. et al. Mammalian Scratch: a neural-specific Snail family transcriptional repressor. Proc. Natl Acad. Sci. USA 98, 4010–4015 (2001).

    Article  CAS  PubMed  Google Scholar 

  36. Manzanares, M., Locascio, A. & Nieto, M. A. The increasing complexity of the Snail superfamily in metazoan evolution. Trends Genet. 17, 178–181 (2001).The organization and new classification for members of the Snail superfamily.

    Article  CAS  PubMed  Google Scholar 

  37. Metzstein, M. M. & Horwitz, H. R. The C. elegans cell death specification gene ces-1 encodes a Snail family zinc finger protein. Mol. Cell 4, 309–319 (1999).The identification of the first Snail family member in nematodes.

    Article  CAS  PubMed  Google Scholar 

  38. Leptin, M. twist and snail as positive and negative regulators during Drosophila mesoderm development. Genes Dev. 5, 1568–1576 (1991).

    Article  CAS  PubMed  Google Scholar 

  39. Ip, Y. T., Park, R., Kosman, D. & Levine, M. The dorsal gradient morphogen regulates stripes of rhomboid expression in the presumptive neruoectoderm of the Drosophila embryo. Genes Dev. 6, 1728–1739 (1992).

    Article  CAS  PubMed  Google Scholar 

  40. Kasai, Y., Nambu, J. R., Lieberman, P. M. & Crews, S. T. Dorsal–ventral patterning in Drosophila: DNA binding of snail protein to the single-minded gene. Proc. Natl Acad. Sci. USA 89, 3414–3418 (1992).

    Article  CAS  PubMed  Google Scholar 

  41. Somer, R. J. & Tautz, D. Expression patterns of twist and snail in Tribolium (Coleoptera) suggest a homologous formation of mesoderm in long and short germ band insects. Dev. Genet. 15, 32–37 (1994).

    Article  Google Scholar 

  42. Corbo, J. C., Erives, A., Di Gregorio, A., Chang, A. & Levine, M. Dorsoventral patterning of the vertebrate neural tube is conserved in a protochordate. Development 124, 2335–2344 (1997).The isolation of the first snail gene in a non-vertebrate chordate.

    CAS  PubMed  Google Scholar 

  43. Goldstein, B., Leviten, M. W. & Weisblat, D. A. Dorsal and Snail homologs in leech development. Dev. Genes Evol. 211, 329–337 (2001).The isolation of the first Snail homologue in lophotrochozoans.

    Article  CAS  PubMed  Google Scholar 

  44. Holland, L. Z. & Holland, N. D. Evolution of neural crest and placodes: amphioxus as a model for the ancestral vertebrate? J. Anat. 199, 85–98 (2001).Although Slug -mutant mice do not show gross abnormalities in either neural crest or mesoderm formation, further analysis of these mutants will provide information on the specific roles of Slug in mammals in these and other tissues.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. LaBonne, C. & Bronner-Fraser, M. Neural crest induction in Xenopus: evidence for a two-signal model. Development 125, 2403–2414 (1998).

    CAS  PubMed  Google Scholar 

  46. Del Barrio, M. G. & Nieto, M. A. Overexpression of Snail family members highlights their ability to promote chick neural crest formation. Development 129, 1583–1594 (2002).

    CAS  PubMed  Google Scholar 

  47. Erickson, C. A. in Principles of Tissue Engineering, 2nd Edn 19–31 (Academic, San Diego, USA, 2000).

    Book  Google Scholar 

  48. Sela-Donenfeld, D. & Kalcheim, C. Regulation of the onset of neural crest migration by coordinated activity of BMP4 and Noggin in the dorsal neural tube. Development 126, 4749–4762 (1999).

    CAS  PubMed  Google Scholar 

  49. Thisse, C., Thisse, B., Schilling, T. F. & Postlethwait, J. H. Structure of the zebrafish snail1 gene and its expression in wild-type, spadetail and no tail mutant embryos. Development 119, 1203–1215 (1993).

    CAS  PubMed  Google Scholar 

  50. Sefton, M., Sanchez, 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 

  51. Velmaat, J. M. et al. Snail an immediate early target gene of parathyroid hormone related peptide signaling in parietal endoderm formation. Int. J. Dev. Biol. 44, 297–307 (2000).

    Google Scholar 

  52. Romano, L. & Runyan, R. B. Slug is an essential target of TGFβ2 signaling in the developing chicken heart. Dev. Biol. 223, 91–102 (2000).

    Article  CAS  PubMed  Google Scholar 

  53. Burdsal, C. A., Damsky, C. H. & Pedersen, R. A. The role of E-cadherin and integrins in mesoderm differentiation and migration at the mammalian primitive streak. Development 118, 829–844 (1993).

    CAS  PubMed  Google Scholar 

  54. Oda, H., Tsukita, S. & Takeichi, M. Dynamic behavior of the cadherin-based cell–cell adhesion system during Drosophila gastrulation. Dev. Biol. 203, 435–450 (1998).

    Article  CAS  PubMed  Google Scholar 

  55. Comijn, J. et al. The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol. Cell 7, 1267–1278 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Nakagawa, S. & Takeichi, M. Neural crest cell–cell adhesion controlled by sequential and subpopulation-specific expression of novel cadherins. Development 121, 1321–1332 (1995).

    CAS  PubMed  Google Scholar 

  57. Nakagawa, S. & Takeichi, M. Neural crest emigration from the neural tube depends on regulated cadherin expression. Development 125, 2963–2971 (1998).

    CAS  PubMed  Google Scholar 

  58. Garcia de Herreros, A. in Common Molecules in Development and Carcinogenesis (Juan March Foundation, Madrid, Spain, 2001).

    Google Scholar 

  59. Navarro, P., Lozano, E. & Cano, A. Expression of E- or P-cadherin is not sufficient to modify the morphology and the tumorigenic behavior of murine spindle carcinoma cells. Possible involvement of plakoglobin. J. Cell Sci. 105, 923–934 (1993).

    CAS  PubMed  Google Scholar 

  60. Liu, J. P. & Jessell, T. M. A role for rhoB in the delamination of neural crest from the dorsal neural tube. Development 125, 5055–5067 (1998).

    CAS  PubMed  Google Scholar 

  61. Barrett, K., Leptin, M. & Settleman, J. The Rho GTPase and a putative RhoGEF mediate a signaling pathway for the cell shape changes in Drosophila gastrulation. Cell 91, 905–915 (1997).

    Article  CAS  PubMed  Google Scholar 

  62. Morize, P., Christiansen, A. E., Costa, M., Parks, S. & Wieschaus, E. Hyperactivation of the folded gastrulation pathway induces specific cell shape changes. Development 125, 589–597 (1998).

    CAS  PubMed  Google Scholar 

  63. Spagnoli, F. M., Cicchini, C., Tripodi, M. & Weiss, M. C. Inhibition of MMH (Met murine hepatocyte) cell differentiation by TGF-β is abrogated by pre-treatment with the heritable differentiation effector FGF1. J. Cell Sci. 113, 3639–3647 (2000).

    CAS  PubMed  Google Scholar 

  64. Liem, K. F. Jr, Tremml, G., Roelink, H. & Jessell, T. Dorsal differentiation of neural plate cells induced by BMP-mediated signals from epidermal ectoderm. Cell 82, 969–979 (1995).

    Article  CAS  PubMed  Google Scholar 

  65. Dickinson, M. E., Selleck, M. A., McMahon, A. P. & Bronner-Fraser, M. Dorsalization of the neural tube by the non-neural ectoderm. Development 121, 2099–2106 (1995).

    CAS  PubMed  Google Scholar 

  66. Marchant, L., Linker, C., Ruiz, P. & Mayor, R. The inductive properties of mesoderm suggest that the neural crest cells are specified by a gradient of BMP. Dev. Biol. 198, 319–329 (1998).

    Article  CAS  PubMed  Google Scholar 

  67. Nguyen, V. H. et al. Ventral and lateral regions of the zebrafish gastrula, including neural crest progenitors, are established by a bmp2b/swirl pathway of genes. Dev. Biol. 199, 93–110 (1998).

    Article  CAS  PubMed  Google Scholar 

  68. Ikeya, M., Lee, S. M. K., Johnson, J. E., McMahon, A. P. & Takada, S. Wnt signalling required for expansion of neural crest and CNS progenitors. Nature 389, 966–970 (1997).

    Article  CAS  PubMed  Google Scholar 

  69. Mayor, R., Guerrero, N. & Martinez, C. Role of FGF and Noggin in neural crest induction. Dev. Biol. 189, 1–12 (1997).

    Article  CAS  PubMed  Google Scholar 

  70. Dorsky, R. I., Moon, R. T. & Raible, D. W. Control of neural crest fate by the Wnt signalling pathway. Nature 396, 370–373 (1998).

    Article  CAS  PubMed  Google Scholar 

  71. Streit, A. & Stern, C. D. Establishment and maintenance of the border of the neural plate and in the chick: involvement of FGF and BMP activity. Mech. Dev. 82, 51–66 (1999).

    Article  CAS  PubMed  Google Scholar 

  72. Alvarez, I. S., Araujo, M. & Nieto, M. A. Neural induction in whole chick embryo cultures by FGF. Dev. Biol. 199, 42–54 (1998).

    Article  CAS  PubMed  Google Scholar 

  73. Savagner, P., Yamada, K. M. & Thiery, J. P. The zinc finger protein Slug causes desmosome dissociation, an initial and necessary step growth factor-induced epithelial–mesenchymal transition. J. Cell Biol. 137, 1403–1419 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Montero, J. A. et al. Role of FGFs in the control of programmed cell death during limb development. Development 128, 2076–2084 (2001).

    Google Scholar 

  75. Ros, M., Sefton, M. & Nieto, M. A. Slug, a zinc finger gene previously implicated in the early patterning of the mesoderm and the neural crest, is also involved in chick limb development. Development 124, 1821–1829 (1997).

    CAS  PubMed  Google Scholar 

  76. Buxton, P. G., Kostakopoulou, K., Brickell, P., Thorogood, P. & Ferretti, P. Expression of the transcription factor slug correlates with growth of the limb bud and is regulated by FGF-4 and retinoic acid. Int. J. Dev. Biol. 41, 559–568 (1997).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  78. Vallin, J. et al. Cloning and characterization of three Xenopus Slug promoters reveal direct regulation by Lef/β-catenin signaling. J. Biol. Chem. 276, 30350–30358 (2001).

    Article  CAS  PubMed  Google Scholar 

  79. Kwonseop, K., Zifan, L. & Hay, E. D. Direct evidence for a role of β-catenin/LEF–1 signaling pathway in induction of EMT. Submitted.

  80. Tan, C. et al. Inhibition of integrin linker kinase (ILK) suppresses β-catenin-Lef/Tcf-dependent transcription and expression of the E-cadherin repressor snail, in APC−/− human colon carcinoma cells. Oncogene 20, 133–140 (2001).

    Article  CAS  PubMed  Google Scholar 

  81. Willert, K. & Nusse, R. β-catenin: a key mediator of Wnt signalling. Curr. Opin. Genet. Dev. 8, 95–102 (1998).

    Article  CAS  PubMed  Google Scholar 

  82. Oft, M., Heider, K. & Beug, H. TGF-β signalling is necessary for carcinoma cell invasiveness and metastasis. Curr. Biol. 8, 1243–1252 (1998).

    Article  CAS  PubMed  Google Scholar 

  83. Akhurst, R. J. & Balmain, A. Genetic events and the role of TGF-β in epithelial tumour progression. J. Pathol. 187, 82–90 (1999).

    Article  CAS  PubMed  Google Scholar 

  84. Isaac, A., Sargent, M. G. & Cooke, J. Control of vertebrate left–right asymmetry by a Snail-related zinc finger gene. Science 275, 1301–1304 (1997).The first description of the involvement of Snail in left–right asymmetry.

    Article  CAS  PubMed  Google Scholar 

  85. Jiang, R., Lan, Y., Norton, C. R., Sundberg, J. P. & Gridley, T. The Slug gene is not essential for mesoderm or neural crest development in mice. Dev. Biol. 198, 277–285 (1998).This study showed the phenotype of Slug -mutant mice.

    Article  CAS  PubMed  Google Scholar 

  86. Linker, C., Bronner-Fraser, M. & Mayor, R. Relationship between gene expression domains of Xsnail, Xslug, and Xtwist and cell movement in the prospective neural crest of Xenopus. Dev. Biol. 224, 215–225 (2000).

    Article  CAS  PubMed  Google Scholar 

  87. Seher, T. C. & Leptin, M. Tribbles, a cell-cycle brake that coordinates proliferation and morphogenesis during Drosophila gastrulation. Curr. Biol. 10, 623–629 (2000).

    Article  CAS  PubMed  Google Scholar 

  88. Grosshans, J. & Wieschaus, E. A genetic link between morphogenesis and cell division during formation of the ventral furrow in Drosophila. Cell 101, 523–531 (2000).

    Article  CAS  PubMed  Google Scholar 

  89. Hayashi, S., Hirose, S., Metcalfe, T. & Shirras, A. D. Control of imaginal cell development by the escargot gene of Drosophila. Development 118, 105–115 (1993).

    CAS  PubMed  Google Scholar 

  90. Ballester, A., Frampton, J., Vilaboa, N. & Calés, C. Heterologous expression of the transcriptional regulator Escargot inhibits megakaryocytic endomitosis. J. Biol. Chem. 276, 43413–43418 (2001).

    Article  CAS  PubMed  Google Scholar 

  91. Ashraf, S. I. & Ip, Y. T. The Snail protein family regulates expression of inscutable and string, genes involved in asymmetry and cell division in Drosophila. Development 128, 4757–4767 (2001).

    CAS  PubMed  Google Scholar 

  92. Cai, Y., Chia, W. & Yang, X. A family of snail-related zinc finger proteins regulates two distinct and parallel mechanisms that mediate Drosophila neuroblast asymmetric divisions. EMBO J. 20, 1704–1714 (2001).The first demonstration of the involvement of Snail family members in asymmetric cell division.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Capdevilla, J., Vogan, K. J., Tabin, C. J. & Izpisua-Belmonte, J. C. Mechanisms of left–right determination in vertebrates. Cell 101, 9–21 (2000).

    Article  Google Scholar 

  94. Monsoro-Burq, A. H. & LeDouarin, N. BMP4 plays a role in left–right patterning in chick embryos by maintaining Sonic hedgehog asymmetry. Mol. Cell 7, 789–799 (2001).

    Article  CAS  PubMed  Google Scholar 

  95. Ashraf, S. I., Hu, X., Rote, J. & Ip, Y. T. The mesoderm determinant snail collaborates with related zinc-finger proteins to control Drosophila neurogenesis. EMBO J. 18, 6426–6438 (1999).The isolation of worniu, a third member of the snail family in Drosophila and the demonstration of the role of snail genes in neural differentiation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Nakakura, E. K. et al. Mammalian Scratch participates in neuronal differentiation in P19 embryonal carcinoma cells. Mol. Brain Res. 95, 162–166 (2001).

    Article  CAS  PubMed  Google Scholar 

  97. Fuse, N., Matakatsu, H., Taniguchi, M. & Hayashi, S. Snail-type zinc finger proteins prevent neurogenesis in Scutoid and transgenic animals in Drosophila. Dev. Genes Evol. 209, 573–580 (1999).

    Article  CAS  PubMed  Google Scholar 

  98. Tanaka-Matakatsu, M., Uemura, T., Oda, H., Takeichi, M. & Hayashi, S. Cadherin-mediated cell adhesion and motility in Drosophila trachea regulated by the transcription factor Escargot. Development 122, 3697–3705 (1996).

    CAS  PubMed  Google Scholar 

  99. Fuse, N., Hirose, S. & Hayashi, S. Determination of wing cell fate by the escargot and snail genes in Drosophila. Development 122, 1059–1067 (1996).

    CAS  PubMed  Google Scholar 

  100. Manzanares, M. et al. Conservation and elaboration of Hox gene regulation during evolution of the vertebrate head. Nature 408, 854–857 (2000).

    Article  CAS  PubMed  Google Scholar 

  101. Holland, P. W. H., García-Fernández, J., Williams, N. A. & Sidow, A. Gene duplications at the origin of vertebrate development. Development S125–S133 (1994).

  102. Wolfe, K. Yesterday's polyploids and the mystery of diploidization. Nature Rev. Genet. 2, 333–341 (2001).

    Article  CAS  PubMed  Google Scholar 

  103. Postlethwait, J. H. et al. Vertebrate genome evolution and the zebrafish gene map. Nature Genet. 18, 345–349 (1998).

    Article  CAS  PubMed  Google Scholar 

  104. Robinson-Rechavi, M., Marchand, O., Escriva, H. & Laudet, V. An ancestral whole-genome duplication may not have been responsible for the abundance of duplicated fish genes. Curr. Biol. 11, R458–R459 (2001).

  105. Perl, A. K., Wilgenbus, P., Dahl, U., Semb, H. & Christofori, G. A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature 392, 190–193 (1998).The demonstration of a causal effect between E-cadherin loss and epithelial tumour progression.

    Article  CAS  PubMed  Google Scholar 

  106. Poser, I. et al. Loss of E-cadherin expression in melanoma cells involves upregulation of the transcriptional repressor Snail. J. Biol. Chem. 276, 24661–24666 (2001).

    Article  CAS  PubMed  Google Scholar 

  107. Yokoyama, K. et al. Reverse correlation of E-cadherin and snail expression in oral squamous cell carcinoma cells in vitro. Oral Oncol. 37, 65–71 (2001).

    Article  CAS  PubMed  Google Scholar 

  108. Cheng, C. W. et al. Mechanisms of inactivation of E-cadherin in breast carcinoma: modification of the two-hit hypothesis of tumor suppressor gene. Oncogene 20, 3814–3823 (2001).

    Article  CAS  PubMed  Google Scholar 

  109. Blanco, M. J. et al. Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Submitted.

  110. Boulay, J. L., Dennefield, C. & Alberga, A. The Drosophila development gene snail encodes a protein with nucleic acid binding domains. Nature 330, 395–398 (1987).The cloning of Drosophila snail.

    Article  CAS  PubMed  Google Scholar 

  111. Whiteley, M., Noguchi, P. D., Sensaburgh, S. M., Odenwald, W. F. & Kassis, J. A. The Drosophila gene escargot encodes a zinc finger motif found in snail-related genes. Mech. Dev. 36, 117–127 (1992).

    Article  CAS  PubMed  Google Scholar 

  112. Smith, S., Metcalfe, J. A. & Elgar, G. Identification and analysis of two snail genes in the pufferfish (Fugu rubripes) and mapping of human SNA to 20q. Gene 247, 119–128 (2000).

    Article  CAS  PubMed  Google Scholar 

  113. Sargent, M. G. & Bennet, M. F. Identification in Xenopus of a structural homologue of the Drosophila gene snail. Development 109, 967–973 (1990). | PubMed |The isolation of the first vertebrate Snail homologue.

    CAS  PubMed  Google Scholar 

  114. Mayor, R., Morgan, R. & Sargent M. G. Induction of the prospective neural crest of Xenopus. Development 121, 767–777 (1995).

    CAS  PubMed  Google Scholar 

  115. Paznekas, W. A., Okajima, K., Schertzer, M., Wood, S. & Jabs, E. W. Genomic organization, expression, and chromosome location of the human SNAIL gene (SNAI1) and a related processed pseudogene (SNAI1P). Genomics 62, 42–49 (1999).

    Article  CAS  PubMed  Google Scholar 

  116. Twigg, S. R. F. & Wilkie, A. O. M. Characterisation of the human snail (SNAI1) gene and exclusion as a major disease gene in craniosynostosis. Hum. Genet. 105, 320–326 (1999).

    CAS  PubMed  Google Scholar 

  117. Rhim, H., Savagner, P., Thibaudeau, G., Thiery, J. P. & Pavan, W. P. Localization of a neural crest transcription factor, Slug, to mouse chromosome 16 and human chromosome 8. Mamm. Genome 8, 872–873 (1997).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

I am very grateful to people in the lab for their work along the years and for their encouraging discussions. Work in the lab is, at present, supported by grants from the Spanish Ministries of Science and Technology (DGESIC) and Health (FIS), and from the Local Government (Comunidad Autónoma de Madrid).

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASES

FlyBase

CtBP

deadpan

RhoGEF2

Escargot

Fog

rhomboid

scratch

single-minded

String

snail

Tribbles

worniu

 LocusLink

BCL-XL

ILK

 Swiss-Prot

β-catenin

cytokeratin-18

desmoplakin

E-cadherin

FGFR1

fibronectin

Gfi1

HLF

Lef-1

Muc-1

Nodal

PTH(rP)

RhoB

SIP1

Slug

Smuc

snail1

snail2

TGF-β1

TGF-β2

vimentin

 WormBase

ces-1

CES-2

CED-3

CED-4

ced-9

EGL-1

Glossary

GASTRULATION

The morphogenetic movements of the early embryo that lead to the generation of the third embryonic layer — the mesoderm.

MESODERM

The third embryonic layer generated during gastrulation, which occupies an intermediate position between the ectoderm and the endoderm. It will give rise to the skeleton, muscles and connective tissue.

TRIPOBLAST

An animal that is composed of three embryonic cell layers: ectoderm, endoderm and mesoderm.

NEURAL CREST

A cell population that originates in the dorsal part of the neural tube and gives rise to many derivatives, including most of the peripheral nervous system, the cranio-facial skeleton and pigmented cells of the body.

EPITHELIAL–MESENCHYMAL TRANSITION

The transformation of an epithelial cell into a mesenchymal cell with migratory and invasive properties.

BODY PLAN

The organization of the embryonic tissues to generate an individual with specific characters.

CHORDATE

An animal with a notochord. These include ascidians, amphioxus and all vertebrates.

NEMATODE

An unsegmented worm.

ANNELID

A segmented worm.

BASIC HELIX–LOOP–HELIX PROTEIN

A transcription factor with a basic domain that binds to a hexanucleotide called the E box, and a hydrophobic domain (the helix–loop–helix) that allows the formation of homo- and heterodimers. They can also have leucine repeats called a leucine zipper.

LOPHOTROCHOZOAN

This group includes two important animal groups, the Lophophorata (brachiopods, flat worms and nemerteans) and the Trochozoa (molluscs and annelids).

EPIDERMAL PLACODE

An epidermal thickening in the embryonic head that differentiates into neurons, as well as into other cell types, at the sites at which the sense organs will form.

PRIMITIVE STREAK

A structure that is formed at the posterior end of amniote embryos at gastrulation stages. An area of mesoderm formation.

METAZOA

The animal kingdom. Includes sponges, diploblasts, protostomes and deuterostomes.

PROTOSTOME

An animal in which the mouth develops from the first opening that develops in the embryo. These include ecdysozoans and lophotrochozoans.

DEUTEROSTOME

An animal in which the anus develops from the first opening of the embryo, and the mouth is formed later. These include echinoderms and chordates.

E-CADHERIN

The main cell–cell adhesion molecule, which is central in maintaining the integrity of epithelial tissues, both in physiology and pathology.

ADHERENS JUNCTION

A cell–cell and cell–extracellular matrix adhesion complex that is composed of integrins and cadherins that are attached to cytoplasmic actin filaments.

PARIETAL ENDODERM

The extraembryonic tissue that is derived from the primitive endoderm and visceral endoderm, and is composed of motile cells that secrete high amounts of extracellular matrix.

PRIMITIVE ENDODERM

The extraembryonic tissue that gives rise to the visceral and parietal endoderm.

VISCERAL ENDODERM

The extraembryonic cell layer that is involved in nutrient uptake and transport.

INVASIVENESS

The ability to degrade and migrate through the extracellular matrix.

CDC25

A family of protein phosphatases that dephosphorylate cyclin-dependent kinases during cell-cycle progression.

IMAGINAL DISCS

The primordia of different adult structures that are present in the larvae of insects with complete metamorphosis.

TROPHOBLAST

The extraembryonic epithelial tissue that is crucial for formation of the placenta.

ENDOREDUPLICATION

The process by which the cells pass to rounds of DNA duplication in the absence of a mitotic division.

ASYMMETRIC CELL DIVISION

A process by which a cell gives rise to two different descendants after division.

GANGLION MOTHER CELL

One of the daughters of a Drosophila neuroblast after asymmetric cell division. It divides once more to give rise to two post-mitotic neurons.

LEFT–RIGHT ASYMMETRY

The differences along the left–right axis of the body.

HEART SITUS

The position of the heart with respect to the left–right axis of the body.

ECDYSOZOANS

One of the important groups within the animal kingdom, it includes arthropods and nematodes.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nieto, M. The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3, 155–166 (2002). https://doi.org/10.1038/nrm757

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm757

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