Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation.

Epithelial-to-mesenchymal transition (EMT) is fundamental to both embryogenesis and tumor metastasis. The Notch intercellular signaling pathway regulates cell fate determination throughout metazoan evolution, and overexpression of activating alleles is oncogenic in mammals. Here we demonstrate that Notch activity promotes EMT during both cardiac development and oncogenic transformation via transcriptional induction of the Snail repressor, a potent and evolutionarily conserved mediator of EMT in many tissues and tumor types. In the embryonic heart, Notch functions via lateral induction to promote a selective transforming growth factor-beta (TGFbeta)-mediated EMT that leads to cellularization of developing cardiac valvular primordia. Embryos that lack Notch signaling elements exhibit severely attenuated cardiac snail expression, abnormal maintenance of intercellular endocardial adhesion complexes, and abortive endocardial EMT in vivo and in vitro. Accordingly, transient ectopic expression of activated Notch1 (N1IC) in zebrafish embryos leads to hypercellular cardiac valves, whereas Notch inhibition prevents valve development. Overexpression of N1IC in immortalized endothelial cells in vitro induces EMT accompanied by oncogenic transformation, with corresponding induction of snail and repression of VE-cadherin expression. Notch is expressed in embryonic regions where EMT occurs, suggesting an intimate and fundamental role for Notch, which may be reactivated during tumor metastasis.

[1]  T. Gridley,et al.  Defects in somite formation in lunatic fringe-deficient mice , 1998, Nature.

[2]  D. Kingsley,et al.  The TGF-beta superfamily: new members, new receptors, and new genetic tests of function in different organisms. , 1994, Genes & development.

[3]  C. Mummery,et al.  RNA and protein localisations of TGF beta 2 in the early mouse embryo suggest an involvement in cardiac development. , 1993, Development.

[4]  J A Epstein,et al.  Neurofibromin modulation of ras activity is required for normal endocardial-mesenchymal transformation in the developing heart. , 1998, Development.

[5]  E. Cuppen,et al.  The Wnt/β-catenin pathway regulates cardiac valve formation , 2003, Nature.

[6]  S. Klewer,et al.  Heart-valve mesenchyme formation is dependent on hyaluronan-augmented activation of ErbB2–ErbB3 receptors , 2002, Nature Medicine.

[7]  G. Weinmaster,et al.  Notch1 is essential for postimplantation development in mice. , 1994, Genes & development.

[8]  W. Hahn,et al.  Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells , 2002, Nature Medicine.

[9]  D. Srivastava,et al.  HRT1, HRT2, and HRT3: a new subclass of bHLH transcription factors marking specific cardiac, somitic, and pharyngeal arch segments. , 1999, Developmental biology.

[10]  D. Srivastava Genetic assembly of the heart: implications for congenital heart disease. , 2001, Annual review of physiology.

[11]  J. Rossant,et al.  Expression analysis of a Notch homologue in the mouse embryo. , 1992, Developmental biology.

[12]  R. Greenspan,et al.  Expression pattern of Motch, a mouse homolog of Drosophila Notch, suggests an important role in early postimplantation mouse development. , 1992, Development.

[13]  Raymond B. Runyan,et al.  Temporal and distinct TGFbeta ligand requirements during mouse and avian endocardial cushion morphogenesis. , 2002, Developmental biology.

[14]  A. Balmain,et al.  Metastasis is driven by sequential elevation of H-ras and Smad2 levels , 2002, Nature Cell Biology.

[15]  Mary-Lee Dequéant,et al.  Periodic Notch inhibition by Lunatic Fringe underlies the chick segmentation clock , 2003, Nature.

[16]  Raphael Kopan,et al.  Notch: a membrane-bound transcription factor. , 2002, Journal of cell science.

[17]  E. Morrisey,et al.  GATA-5: a transcriptional activator expressed in a novel temporally and spatially-restricted pattern during embryonic development. , 1997, Developmental biology.

[18]  E. Carver,et al.  The Mouse Snail Gene Encodes a Key Regulator of the Epithelial-Mesenchymal Transition , 2001, Molecular and Cellular Biology.

[19]  J. Seidman,et al.  Chamber-specific cardiac expression of Tbx5 and heart defects in Holt-Oram syndrome. , 1999, Developmental biology.

[20]  J. Sundberg,et al.  Notch signaling is essential for vascular morphogenesis in mice. , 2000, Genes & development.

[21]  Raymond B. Runyan,et al.  Slug is an essential target of TGFbeta2 signaling in the developing chicken heart. , 2000, Developmental biology.

[22]  R Bicknell,et al.  Delta4, an endothelial specific notch ligand expressed at sites of physiological and tumor angiogenesis. , 2001, Differentiation; research in biological diversity.

[23]  M. Bennett,et al.  Cloning and developmental expression of Sna, a murine homologue of the Drosophila snail gene. , 1992, Development.

[24]  A. G. Herreros,et al.  The transcription factor Snail is a repressor of E-cadherin gene expression in epithelial tumour cells , 2000, Nature Cell Biology.

[25]  E. Robey Regulation of T cell fate by Notch. , 1999, Annual review of immunology.

[26]  C. Haass,et al.  A γ‐secretase inhibitor blocks Notch signaling in vivo and causes a severe neurogenic phenotype in zebrafish , 2002, EMBO reports.

[27]  The vascular endothelial-cadherin promoter directs endothelial-specific expression in transgenic mice. , 1999 .

[28]  M. Lampugnani,et al.  A novel endothelial-specific membrane protein is a marker of cell-cell contacts , 1992, The Journal of cell biology.

[29]  O. Pourquié,et al.  Segmentation of the paraxial mesoderm and vertebrate somitogenesis. , 2000, Current topics in developmental biology.

[30]  E. Lander,et al.  A molecular signature of metastasis in primary solid tumors , 2003, Nature Genetics.

[31]  S. Dedhar,et al.  Inhibition of integrin linked kinase (ILK) suppresses β-catenin-Lef/Tcf-dependent transcription and expression of the E-cadherin repressor, snail, in APC−/− human colon carcinoma cells , 2001, Oncogene.

[32]  T. Mak,et al.  Disruption of the mouse RBP-J kappa gene results in early embryonic death. , 1995, Development.

[33]  S. Hokari,et al.  Mechanisms involved in valvuloseptal endocardial cushion formation in early cardiogenesis: Roles of transforming growth factor (TGF)‐β and bone morphogenetic protein (BMP) , 2000, The Anatomical record.

[34]  G. Moreno-Bueno,et al.  Correlation of Snail expression with histological grade and lymph node status in breast carcinomas , 2002, Oncogene.

[35]  J. Rossant,et al.  Interaction between Notch signalling and Lunatic fringe during somite boundary formation in the mouse , 1999, Current Biology.

[36]  J. I. Izpisúa Belmonte,et al.  Notch activity induces Nodal expression and mediates the establishment of left-right asymmetry in vertebrate embryos. , 2003, Genes & development.

[37]  Julian Lewis,et al.  Notch signaling in the development of the inner ear: lessons from Drosophila. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[38]  J. Thiery Epithelial–mesenchymal transitions in tumour progression , 2002, Nature Reviews Cancer.

[39]  Á. Raya,et al.  Activation of Notch signaling pathway precedes heart regeneration in zebrafish , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[40]  R. Markwald,et al.  Structural development of endocardial cushions. , 1977, The American journal of anatomy.

[41]  F. D. Miller,et al.  Functional gamma‐secretase inhibitors reduce beta‐amyloid peptide levels in brain , 2000, Journal of neurochemistry.

[42]  M. Quintanilla,et al.  Transforming growth factor beta-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions. , 2003, The Journal of biological chemistry.

[43]  Yukinori Endo,et al.  Bimodal functions of Notch-mediated signaling are involved in neural crest formation during avian ectoderm development. , 2002, Development.

[44]  S. Artavanis-Tsakonas,et al.  Notch signaling: cell fate control and signal integration in development. , 1999, Science.

[45]  J. Lewis,et al.  Notch signalling and the control of cell fate choices in vertebrates. , 1998, Seminars in cell & developmental biology.

[46]  T. Gridley Notch signaling during vascular development , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[47]  Yvonne A. Evrard,et al.  lunatic fringe is an essential mediator of somite segmentation and patterning , 1998, Nature.

[48]  Raymond B. Runyan,et al.  TGFβ Type III and TGFβ Type II receptors have distinct activities during epithelial–mesenchymal cell transformation in the embryonic heart , 2001, Developmental dynamics : an official publication of the American Association of Anatomists.

[49]  J. Campos-Ortega,et al.  Overexpression of a zebrafish homologue of the Drosophila neurogenic gene Delta perturbs differentiation of primary neurons and somite development , 1997, Mechanisms of Development.

[50]  D. Stainier,et al.  UDP-glucose dehydrogenase required for cardiac valve formation in zebrafish. , 2001, Science.

[51]  Manuel J. Aybar,et al.  Snail precedes Slug in the genetic cascade required for the specification and migration of the Xenopus neural crest , 2003, Development.

[52]  B. Hogan,et al.  Organogenesis and pattern formation in the mouse: RNA distribution patterns suggest a role for bone morphogenetic protein-2A (BMP-2A). , 1990, Development.

[53]  H. Weintraub,et al.  The intracellular domain of mouse Notch: a constitutively activated repressor of myogenesis directed at the basic helix-loop-helix region of MyoD. , 1994, Development.

[54]  Hans Clevers,et al.  Notch1 functions as a tumor suppressor in mouse skin , 2003, Nature Genetics.

[55]  K. Irvine,et al.  Fringe modulates Notch–ligand interactions , 1997, Nature.

[56]  Raymond B. Runyan,et al.  Invasion of mesenchyme into three-dimensional collagen gels: a regional and temporal analysis of interaction in embryonic heart tissue. , 1983, Developmental biology.

[57]  J. Rossant,et al.  Notch1 is required for the coordinate segmentation of somites. , 1995, Development.

[58]  Francisco Portillo,et al.  The transcription factor Snail controls epithelial–mesenchymal transitions by repressing E-cadherin expression , 2000, Nature Cell Biology.

[59]  D. Stainier,et al.  The zebrafish gene cloche acts upstream of a flk-1 homologue to regulate endothelial cell differentiation. , 1997, Development.

[60]  David J Anderson,et al.  Transient Notch Activation Initiates an Irreversible Switch from Neurogenesis to Gliogenesis by Neural Crest Stem Cells , 2000, Cell.

[61]  R R Markwald,et al.  Molecular regulation of atrioventricular valvuloseptal morphogenesis. , 1995, Circulation research.

[62]  M. Nieto,et al.  The snail superfamily of zinc-finger transcription factors , 2002, Nature Reviews Molecular Cell Biology.

[63]  Didier Y. R. Stainier,et al.  Zebrafish genetics and vertebrate heart formation , 2001, Nature Reviews Genetics.

[64]  C. Drake,et al.  Vasculogenesis in the day 6.5 to 9.5 mouse embryo. , 2000, Blood.

[65]  Kenneth M. Yamada,et al.  The Zinc-Finger Protein Slug Causes Desmosome Dissociation, an Initial and Necessary Step for Growth Factor–induced Epithelial–Mesenchymal Transition , 1997, The Journal of cell biology.

[66]  U. Lendahl,et al.  Notch and neurogenesis , 1998, Journal of neuroscience research.

[67]  S. Lehnert,et al.  TGF beta in murine morphogenetic processes: the early embryo and cardiogenesis. , 1990, Development.