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:

TGFβ signalling in context

Key Points

  • The effects of transforming growth factor-β (TGFβ) are different depending on the cellular context. Three kinds of contextual determinants shape the TGFβ response: the signal transduction components, the transcriptional cofactors of SMAD mediators and the epigenetic state of the cell.

  • TGFβ receptors signal through the canonical SMAD pathway that controls the expression of hundreds of genes, and several non-canonical pathways that regulate cell polarity, the cytoskeleton and microRNA maturation.

  • SMAD proteins regulate transcription with close ties to RNA polymerase II, and undergo activation, recycling and turnover in a highly orchestrated manner. SMAD proteins are a hub for pathway regulation and compute diverse inputs for a finely tuned response.

  • SMAD proteins recruit a histone reader protein to gain access to repressed chromatin, and histone modifiers, DNA modifiers and nucleosome remodellers to regulate transcription.

  • Master regulators of pluripotency in embryonic stem cells (ES cells) and of lineage determination in progenitor cells dominantly direct signal-activated SMAD proteins to target sites in the genome. In the context of differentiated cells, signal-activated SMAD proteins are directed to different subsets of target genes by different DNA-binding partners.

  • The TGFβ, bone morphogenetic protein (BMP) and WNT pathways converge in the context of ES cells, progenitor cells and cells undergoing epithelial–mesenchymal transition (EMT) or induced pluripotent stem cell (iPS cell) reprogramming. TGFβ kills premalignant cells, and this selects for mutant clones that avert this effect and instead use TGFβ for invasion and metastasis in this context.

Abstract

The basic elements of the transforming growth factor-β (TGFβ) pathway were revealed more than a decade ago. Since then, the concept of how the TGFβ signal travels from the membrane to the nucleus has been enriched with additional findings, and its multifunctional nature and medical relevance have relentlessly come to light. However, an old mystery has endured: how does the context determine the cellular response to TGFβ? Solving this question is key to understanding TGFβ biology and its many malfunctions. Recent progress is pointing at answers.

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: Contextual determinants of TGFβ action.
Figure 2: TGFβ receptors and signal transducers.
Figure 3: The SMAD signalling cycle.
Figure 4: SMAD access to chromatin.
Figure 5: TGFβ action in ES cells, lineage progenitors and differentiated cells.
Figure 6: TGFβ action in EMT, MET and iPS cell transition.
Figure 7: TGFβ action in tumour suppression and tumour progression.

Similar content being viewed by others

References

  1. Shi, Y. & Massagué, J. Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell 113, 685–700 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Massagué, J. How cells read TGF-β signals. Nature Rev. Mol. Cell Biol. 1, 169–178 (2000).

    Article  Google Scholar 

  3. Huminiecki, L. et al. Emergence, development and diversification of the TGF-β signalling pathway within the animal kingdom. BMC Evol. Biol. 9, 28 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Mullen, A. C. et al. Master transcription factors determine cell-type-specific responses to TGF-β signaling. Cell 147, 565–576 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Xi, Q. et al. A poised chromatin platform for TGF-β access to master regulators. Cell 147, 1511–1524 (2011). Identifies TRIM33 as a partner of Nodal-activated SMAD3 that binds to and disables repressive histone marks in master regulators of ES cell differentiation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Feng, X. H. & Derynck, R. Specificity and versatility in TGF-β signaling through Smads. Annu. Rev. Cell Dev. Biol. 21, 659–693 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Massagué, J., Seoane, J. & Wotton, D. Smad transcription factors. Genes Dev. 19, 2783–2810 (2005).

    Article  PubMed  CAS  Google Scholar 

  8. Trompouki, E. et al. Lineage regulators direct BMP and Wnt pathways to cell-specific programs during differentiation and regeneration. Cell 147, 577–589 (2011). Demonstrates, together with reference 4, that master regulators of pluripotency in the context of ES cells and of lineage determination in progenitor cells direct signal-activated SMAD proteins to many sites in the genome.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kang, Y., Chen, C. R. & Massagué, J. A self-enabling TGFβ response coupled to stress signaling: Smad engages stress response factor ATF3 for Id1 repression in epithelial cells. Mol. Cell 11, 915–926 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Padua, D. et al. TGFβ primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell 133, 66–77 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kavsak, P. et al. Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGFβ receptor for degradation. Mol. Cell 6, 1365–1375 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Wu, J. W. et al. Structural mechanism of Smad4 recognition by the nuclear oncoprotein Ski: insights on Ski-mediated repression of TGF-β signaling. Cell 111, 357–367 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Heldin, C. H., Landstrom, M. & Moustakas, A. Mechanism of TGF-β signaling to growth arrest, apoptosis, and epithelial–mesenchymal transition. Curr. Opin. Cell Biol. 21, 166–176 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Massagué, J. TGFβ in cancer. Cell 134, 215–230 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Wu, M. Y. & Hill, C. S. Tgf-β superfamily signaling in embryonic development and homeostasis. Dev. Cell 16, 329–343 (2009).

    Article  CAS  PubMed  Google Scholar 

  16. Affolter, M. & Basler, K. The Decapentaplegic morphogen gradient: from pattern formation to growth regulation. Nature Rev. Genet. 8, 663–674 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Kicheva, A. & Gonzalez-Gaitan, M. The Decapentaplegic morphogen gradient: a precise definition. Curr. Opin. Cell Biol. 20, 137–143 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Plouhinec, J. L., Zakin, L. & De Robertis, E. M. Systems control of BMP morphogen flow in vertebrate embryos. Curr. Opin. Genet. Dev. 21, 696–703 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chen, X. et al. Smad4 and FAST-1 in the assembly of activin-responsive factor. Nature 389, 85–89 (1997).

    Article  CAS  PubMed  Google Scholar 

  20. Hata, A. et al. OAZ uses distinct DNA- and protein-binding zinc fingers in separate BMP–Smad and Olf signaling pathways. Cell 100, 229–240 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Gomis, R. R. et al. A FoxO–Smad synexpression group in human keratinocytes. Proc. Natl Acad. Sci. USA 103, 12747–12752 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Orkin, S. H. & Hochedlinger, K. Chromatin connections to pluripotency and cellular reprogramming. Cell 145, 835–850 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Young, R. A. Control of the embryonic stem cell state. Cell 144, 940–954 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nieto, M. A. The ins and outs of the epithelial to mesenchymal transition in health and disease. Annu. Rev. Cell Dev. Biol. 27, 347–376 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Scheel, C. et al. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell 145, 926–940 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wrana, J. L., Attisano, L., Wieser, R., Ventura, F. & Massagué, J. Mechanism of activation of the TGF-β receptor. Nature 370, 341–347 (1994).

    Article  CAS  PubMed  Google Scholar 

  27. Huse, M. et al. The TGFβ receptor activation process: an inhibitor- to substrate-binding switch. Mol. Cell 8, 671–682 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Greenwald, J. et al. The BMP7/ActRII extracellular domain complex provides new insights into the cooperative nature of receptor assembly. Mol. Cell 11, 605–617 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Groppe, J. et al. Cooperative assembly of TGF-β superfamily signaling complexes is mediated by two disparate mechanisms and distinct modes of receptor binding. Mol. Cell 29, 157–168 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Moustakas, A. & Heldin, C. H. The regulation of TGFβ signal transduction. Development 136, 3699–3714 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. Pardali, E., Goumans, M. J. & ten Dijke, P. Signaling by members of the TGF-β family in vascular morphogenesis and disease. Trends Cell Biol. 20, 556–567 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Cheifetz, S. et al. The transforming growth factor-β system, a complex pattern of cross-reactive ligands and receptors. Cell 48, 409–415 (1987).

    Article  CAS  PubMed  Google Scholar 

  33. Zakin, L. & De Robertis, E. M. Extracellular regulation of BMP signaling. Curr. Biol. 20, R89–R92 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zilberberg, L. et al. Specificity of latent TGF-β binding protein (LTBP) incorporation into matrix: role of fibrillins and fibronectin. J. Cell Physiol. 227, 3828–3836 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Groppe, J. et al. Structural basis of BMP signalling inhibition by the cystine knot protein Noggin. Nature 420, 636–642 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Potti, T. A., Petty, E. M. & Lesperance, M. M. A comprehensive review of reported heritable noggin-associated syndromes and proposed clinical utility of one broadly inclusive diagnostic term: NOG-related-symphalangism spectrum disorder (NOG-SSD). Hum. Mutat. 32, 877–886 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Sneddon, J. B. et al. Bone morphogenetic protein antagonist gremlin 1 is widely expressed by cancer-associated stromal cells and can promote tumor cell proliferation. Proc. Natl Acad. Sci. USA 103, 14842–14847 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Shi, M. et al. Latent TGF-β structure and activation. Nature 474, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lindsay, M. E. & Dietz, H. C. Lessons on the pathogenesis of aneurysm from heritable conditions. Nature 473, 308–316 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Muller, P. et al. Differential diffusivity of Nodal and Lefty underlies a reaction-diffusion patterning system. Science 336, 721–724 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Schier, A. F. Nodal morphogens. Cold Spring Harb. Perspect. Biol. 1, a003459 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Lewis, K. A. et al. β-glycan binds inhibin and can mediate functional antagonism of activin signalling. Nature 404, 411–414 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Wiater, E., Harrison, C. A., Lewis, K. A., Gray, P. C. & Vale, W. W. Identification of distinct inhibin and transforming growth factor β-binding sites on β-glycan: functional separation of β-glycan co-receptor actions. J. Biol. Chem. 281, 17011–17022 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. López-Casillas, F., Wrana, J. L. & Massagué, J. β-glycan presents ligand to the TGFβ signaling receptor. Cell 73, 1435–1444 (1993).

    Article  PubMed  Google Scholar 

  45. Gritsman, K. et al. The EGF-CFC protein one-eyed pinhead is essential for nodal signaling. Cell 97, 121–132 (1999).

    Article  CAS  PubMed  Google Scholar 

  46. Gallione, C. et al. Overlapping spectra of SMAD4 mutations in juvenile polyposis (JP) and JP-HHT syndrome. Am. J. Med. Genet. A 152A, 333–339 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Marchuk, D. A. Genetic abnormalities in hereditary hemorrhagic telangiectasia. Curr. Opin. Hematol. 5, 332–338 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. Xu, L., Kang, Y., Col, S. & Massagué, J. Smad2 nucleocytoplasmic shuttling by nucleoporins CAN/Nup214 and Nup153 feeds TGFβ signaling complexes in the cytoplasm and nucleus. Mol. Cell 10, 271–282 (2002).

    Article  CAS  PubMed  Google Scholar 

  49. Chen, X. & Xu, L. Specific nucleoporin requirement for Smad nuclear translocation. Mol. Cell. Biol. 30, 4022–4034 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hill, C. S. Nucleocytoplasmic shuttling of Smad proteins. Cell Res. 19, 36–46 (2009).

    Article  CAS  PubMed  Google Scholar 

  51. Dai, F., Lin, X., Chang, C. & Feng, X. H. Nuclear export of Smad2 and Smad3 by RanBP3 facilitates termination of TGF-β signaling. Dev. Cell 16, 345–357 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. BabuRajendran, N. et al. Structure of Smad1 MH1/DNA complex reveals distinctive rearrangements of BMP and TGF-β effectors. Nucleic Acids Res. 38, 3477–3488 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shi, Y. et al. Crystal structure of a Smad MH1 domain bound to DNA: insights on DNA binding in TGF-β signaling. Cell 94, 585–594 (1998).

    Article  CAS  PubMed  Google Scholar 

  54. Korchynskyi, O. & ten Dijke, P. Identification and functional characterization of distinct critically important bone morphogenetic protein-specific response elements in the Id1 promoter. J. Biol. Chem. 277, 4883–4891 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Yoon, S. J., Wills, A. E., Chuong, E., Gupta, R. & Baker, J. C. HEB and E2A function as SMAD/FOXH1 cofactors. Genes Dev. 25, 1654–1661 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Koinuma, D. et al. Promoter-wide analysis of Smad4 binding sites in human epithelial cells. Cancer Sci. 100, 2133–2142 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Fei, T. et al. Genome-wide mapping of SMAD target genes reveals the role of BMP signaling in embryonic stem cell fate determination. Genome Res. 20, 36–44 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Morikawa, M. et al. ChIP-seq reveals cell type-specific binding patterns of BMP-specific Smads and a novel binding motif. Nucleic Acids Res. 39, 8712–8727 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Alarcón, C. et al. Nuclear CDKs drive Smad transcriptional activation and turnover in BMP and TGF-β pathways. Cell 139, 757–769 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Gao, S. et al. Ubiquitin ligase Nedd4L targets activated Smad2/3 to limit TGF-β signaling. Mol. Cell 36, 457–468 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Fuentealba, L. C. et al. Integrating patterning signals: Wnt/GSK3 regulates the duration of the BMP/Smad1 signal. Cell 131, 980–993 (2007). Shows the integration of dorsoventral BMP and anteroposterior WNT signal gradients at the level of SMAD1 phosphorylation during embryonic pattern formation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Sapkota, G., Alarcón, C., Spagnoli, F. M., Brivanlou, A. H. & Massagué, J. Balancing BMP signaling through integrated inputs into the Smad1 linker. Mol. Cell 25, 441–454 (2007).

    Article  CAS  PubMed  Google Scholar 

  63. Aragón, E. et al. A Smad action turnover switch operated by WW domain readers of a phosphoserine code. Genes Dev. 25, 1275–1288 (2011). Shows, together with reference 59, how the nuclear kinases CDK8, CDK9 and GSK3 drive a cycle of SMAD utilization and disposal that is an integral part of canonical BMP and TGFβ pathways.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Conaway, R. C., Sato, S., Tomomori-Sato, C., Yao, T. & Conaway, J. W. The mammalian Mediator complex and its role in transcriptional regulation. Trends Briochem. Sci. 30, 250–255 (2005).

    Article  CAS  Google Scholar 

  65. Bruce, D. L. & Sapkota, G. P. Phosphatases in SMAD regulation. FEBS Lett. 586, 1897–1905 (2012).

    Article  CAS  PubMed  Google Scholar 

  66. Sapkota, G. et al. Dephosphorylation of the linker regions of Smad1 and Smad2/3 by small C-terminal domain phosphatases has distinct outcomes for bone morphogenetic protein and transforming growth factor-β pathways. J. Biol. Chem. 281, 40412–40419 (2006).

    Article  CAS  PubMed  Google Scholar 

  67. Ghosh, A., Shuman, S. & Lima, C. D. The structure of Fcp1, an essential RNA polymerase II CTD phosphatase. Mol. Cell 32, 478–490 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lin, X. et al. PPM1A functions as a Smad phosphatase to terminate TGFβ signaling. Cell 125, 915–928 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Simonsson, M., Kanduri, M., Gronroos, E., Heldin, C. H. & Ericsson, J. The DNA binding activities of Smad2 and Smad3 are regulated by coactivator-mediated acetylation. J. Biol. Chem. 281, 39870–39880 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Lonn, P. et al. PARP-1 attenuates Smad-mediated transcription. Mol. Cell 40, 521–532 (2010).

    Article  PubMed  CAS  Google Scholar 

  71. Ebisawa, T. et al. Smurf1 interacts with transforming growth factor-β type I receptor through Smad7 and induces receptor degradation. J. Biol. Chem. 276, 12477–12480 (2001).

    Article  CAS  PubMed  Google Scholar 

  72. Al-Salihi, M. A., Herhaus, L., Maccartney, T. & Sapkota, G. USP11 augments TGFb signalling by deubiquitylating ALK5. Open Biol. 2, 120063 (2010).

    Article  CAS  Google Scholar 

  73. Eichhorn, P. J. et al. USP15 stabilizes TGF-β receptor I and promotes oncogenesis through the activation of TGF-β signaling in glioblastoma. Nature Med. 18, 429–435 (2012).

    Article  CAS  PubMed  Google Scholar 

  74. Zhang, L. et al. USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-β type I receptor. Nature Cell Biol. 14, 717–726 (2012).

    Article  CAS  PubMed  Google Scholar 

  75. Inui, M. et al. USP15 is a deubiquitylating enzyme for receptor-activated SMADs. Nature Cell Biol. 13, 1368–1375 (2011).

    Article  CAS  PubMed  Google Scholar 

  76. Paulsen, M., Legewie, S., Eils, R., Karaulanov, E. & Niehrs, C. Negative feedback in the bone morphogenetic protein 4 (BMP4) synexpression group governs its dynamic signaling range and canalizes development. Proc. Natl Acad. Sci. USA 108, 10202–10207 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Stroschein, S. L., Wang, W., Zhou, S., Zhou, Q. & Luo, K. Negative feedback regulation of TGF-β signaling by the SnoN oncoprotein. Science 286, 771–774 (1999).

    Article  CAS  PubMed  Google Scholar 

  78. Koinuma, D. et al. Arkadia amplifies TGF-β superfamily signalling through degradation of Smad7. EMBO J. 22, 6458–6470 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Levy, L. et al. Arkadia activates Smad3/Smad4-dependent transcription by triggering signal-induced SnoN degradation. Mol. Cell. Biol. 27, 6068–6083 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kretzschmar, M., Doody, J. & Massagué, J. Opposing BMP and EGF signalling pathways converge on the TGF-β family mediator Smad1. Nature 389, 618–622 (1997).

    Article  CAS  PubMed  Google Scholar 

  81. Matsuura, I. et al. Cyclin-dependent kinases regulate the antiproliferative function of Smads. Nature 430, 226–231 (2004).

    Article  CAS  PubMed  Google Scholar 

  82. Labbé, E., Letamendia, A. & Attisano, L. Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-β and wnt pathways. Proc. Natl Acad. Sci. USA 97, 8358–8363 (2000).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Labbé, E. et al. Transcriptional cooperation between the transforming growth factor-β and Wnt pathways in mammary and intestinal tumorigenesis. Cancer Res. 67, 75–84 (2007).

    Article  PubMed  Google Scholar 

  84. Nakano, N. et al. Requirement of TCF7L2 for TGF-β-dependent transcriptional activation of the TMEPAI gene. J. Biol. Chem. 285, 38023–38033 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Seoane, J., Le, H. V., Shen, L., Anderson, S. A. & Massagué, J. Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell 117, 211–223 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Naka, K. et al. TGF-β–FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature 463, 676–680 (2010).

    Article  CAS  PubMed  Google Scholar 

  87. Vincent, T. et al. A SNAIL1–SMAD3/4 transcriptional repressor complex promotes TGF-β mediated epithelial–mesenchymal transition. Nature Cell Biol. 11, 943–950 (2009). Evidence that formation of a SNAIL1–SMAD3–SMAD4 transcriptional complex provides a mechanism for repression of epithelial genes in the context of EMT.

    Article  CAS  PubMed  Google Scholar 

  88. Varelas, X. & Wrana, J. L. Coordinating developmental signaling: novel roles for the Hippo pathway. Trends Cell Biol. 22, 88–96 (2012).

    Article  CAS  PubMed  Google Scholar 

  89. Agricola, E., Randall, R. A., Gaarenstroom, T., Dupont, S. & Hill, C. S. Recruitment of TIF1γ to chromatin via its PHD finger–bromodomain activates its ubiquitin ligase and transcriptional repressor activities. Mol. Cell 43, 85–96 (2011).

    Article  CAS  PubMed  Google Scholar 

  90. Ross, S. et al. Smads orchestrate specific histone modifications and chromatin remodeling to activate transcription. EMBO J. 25, 4490–4502 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zerlanko, B. J., Bartholin, L., Melhuish, T. A. & Wotton, D. Premature senescence and increased TGFβ signaling in the absence of Tgif1. PLoS ONE 7, e35460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Taniguchi, K., Anderson, A. E., Sutherland, A. E. & Wotton, D. Loss of Tgif function causes holoprosencephaly by disrupting the SHH signaling pathway. PLoS Genet. 8, e1002524 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Thillainadesan, G. et al. TGF-β-dependent active demethylation and expression of the p15ink4b tumor suppressor are impaired by the ZNF217/CoREST complex. Mol. Cell 46, 636–649 (2012). Demonstrates a remarkable case in which a SMAD transcriptional complex recruits a base excision repair complex to remove repressive DNA methylation from a TGFβ target gene.

    Article  CAS  PubMed  Google Scholar 

  94. Gomis, R. R., Alarcón, C., Nadal, C., Van Poznak, C. & Massagué, J. C/EBPβ at the core of the TGFβ cytostatic response and its evasion in metastatic breast cancer cells. Cancer Cell 10, 203–214 (2006).

    Article  CAS  PubMed  Google Scholar 

  95. Seoane, J. et al. TGFβ influences Myc, Miz-1 and Smad to control the CDK inhibitor p15INK4b. Nature Cell Biol. 3, 400–408 (2001).

    Article  CAS  PubMed  Google Scholar 

  96. Xi, Q., He, W., Zhang, X. H., Le, H. V. & Massagué, J. Genome-wide impact of the BRG1 SWI/SNF chromatin remodeler on the transforming growth factor-β transcriptional program. J. Biol. Chem. 283, 1146–1155 (2008).

    Article  CAS  PubMed  Google Scholar 

  97. Wilson, B. G. & Roberts, C. W. SWI/SNF nucleosome remodellers and cancer. Nature Rev. Cancer 11, 481–492 (2011).

    Article  CAS  Google Scholar 

  98. Hatakeyama, S. TRIM proteins and cancer. Nature Rev. Cancer 11, 792–804 (2011).

    Article  CAS  Google Scholar 

  99. He, W. et al. Hematopoiesis controlled by distinct TIF1γ and Smad4 branches of the TGFβ pathway. Cell 125, 929–941 (2006).

    Article  CAS  PubMed  Google Scholar 

  100. Dupont, S. et al. FAM/USP9x, a deubiquitinating enzyme essential for TGFβ signaling, controls Smad4 monoubiquitination. Cell 136, 123–135 (2009).

    Article  CAS  PubMed  Google Scholar 

  101. Morsut, L. et al. Negative control of Smad activity by ectodermin/Tif1γ patterns the mammalian embryo. Development 137, 2571–2578 (2010).

    Article  CAS  PubMed  Google Scholar 

  102. Vincent, D. F. et al. Inactivation of TIF1γ cooperates with Kras to induce cystic tumors of the pancreas. PLoS Genet. 5, e1000575 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Bardeesy, N. et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev. 20, 3130–3146 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Ozdamar, B. et al. Regulation of the polarity protein Par6 by TGFβ receptors controls epithelial cell plasticity. Science 307, 1603–1609 (2005).

    Article  CAS  PubMed  Google Scholar 

  105. Viloria-Petit, A. M. et al. A role for the TGFβ–Par6 polarity pathway in breast cancer progression. Proc. Natl Acad. Sci. USA 106, 14028–14033 (2009). Shows a non-canonical mode of TGFβ signalling that involves direct phosphorylation of a cell polarity regulator by the TGFβ type II receptor and that facilitates cancer cell invasion.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Yi, J. J., Barnes, A. P., Hand, R., Polleux, F. & Ehlers, M. D. TGF-β signaling specifies axons during brain development. Cell 142, 144–157 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Morrell, N. W. Pulmonary hypertension due to BMPR2 mutation: a new paradigm for tissue remodeling? Proc. Am. Thorac. Soc. 3, 680–686 (2006).

    Article  CAS  PubMed  Google Scholar 

  108. Foletta, V. C. et al. Direct signaling by the BMP type II receptor via the cytoskeletal regulator LIMK1. J. Cell Biol. 162, 1089–1098 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Lee-Hoeflich, S. T. et al. Activation of LIMK1 by binding to the BMP receptor, BMPRII, regulates BMP-dependent dendritogenesis. EMBO J. 23, 4792–4801 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Park, K. S. & Gumbiner, B. M. Cadherin-6B stimulates an epithelial mesenchymal transition and the delamination of cells from the neural ectoderm via LIMK/cofilin mediated non-canonical BMP receptor signaling. Dev. Biol. 366, 232–243 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Holm, T. M. et al. Noncanonical TGFβ signaling contributes to aortic aneurysm progression in Marfan syndrome mice. Science 332, 358–361 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Mu, Y., Gudey, S. K. & Landstrom, M. Non-Smad signaling pathways. Cell Tissue Res. 347, 11–20 (2012).

    Article  CAS  PubMed  Google Scholar 

  113. 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 

  114. Bruna, A. et al. High TGFβ-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer Cell 11, 147–160 (2007).

    Article  CAS  PubMed  Google Scholar 

  115. Ikushima, H. et al. Autocrine TGF-β signaling maintains tumorigenicity of glioma-initiating cells through Sry-related HMG-box factors. Cell Stem Cell 5, 504–514 (2009). Shows that canonical TGFβ–SMAD signalling in the context of glioma stem cells drives expression of the pluripotency factor SOX2 to promote self-renewal.

    Article  CAS  PubMed  Google Scholar 

  116. Peñuelas, S. et al. TGF-β increases glioma-initiating cell self-renewal through the induction of LIF in human glioblastoma. Cancer Cell 15, 315–327 (2009). Shows that canonical SMAD signalling in the context of glioma cells drives expression of pluripotency cytokine LIF and that this promotes tumour reinitiation.

    Article  PubMed  CAS  Google Scholar 

  117. Kang, Y. et al. Breast cancer bone metastasis mediated by the Smad tumor suppressor pathway. Proc. Natl Acad. Sci. USA 102, 13909–13914 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Davis, B. N., Hilyard, A. C., Nguyen, P. H., Lagna, G. & Hata, A. Smad proteins bind a conserved RNA sequence to promote microRNA maturation by Drosha. Mol. Cell 39, 373–384 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Davis, B. N., Hilyard, A. C., Lagna, G. & Hata, A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature 454, 56–61 (2008). Discovers, together with reference 118, a non-canonical mode of TGFβ signalling that involves a direct interaction of signal-activated SMAD proteins with a subset of miRNA precursors in the Drosha maturation complex.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Chen, X. et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133, 1106–1117 (2008).

    Article  CAS  PubMed  Google Scholar 

  121. Ying, Q. L., Nichols, J., Chambers, I. & Smith, A. BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell 115, 281–292 (2003).

    Article  CAS  PubMed  Google Scholar 

  122. Liu, F., Pouponnot, C. & Massagué, J. Dual role of the Smad4/DPC4 tumor suppressor in TGFβ-inducible transcriptional complexes. Genes Dev. 11, 3157–3167 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Brabletz, S. & Brabletz, T. The ZEB/miR-200 feedback loop – a motor of cellular plasticity in development and cancer? EMBO Rep. 11, 670–677 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Derynck, R. & Akhurst, R. J. Differentiation plasticity regulated by TGF-β family proteins in development and disease. Nature Cell Biol. 9, 1000–1004 (2007).

    Article  CAS  PubMed  Google Scholar 

  125. Tan, E. J. et al. Regulation of transcription factor Twist expression by the DNA architectural protein high mobility group A2 during epithelial-to-mesenchymal transition. J. Biol. Chem. 287, 7134–7145 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Thuault, S. et al. HMGA2 and Smads co-regulate SNAIL1 expression during induction of epithelial-to-mesenchymal transition. J. Biol. Chem. 283, 33437–33446 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Nawshad, A., Medici, D., Liu, C. C. & Hay, E. D. TGFβ3 inhibits E-cadherin gene expression in palate medial-edge epithelial cells through a Smad2–Smad4–LEF1 transcription complex. J. Cell Sci. 120, 1646–1653 (2007).

    Article  CAS  PubMed  Google Scholar 

  128. Samavarchi-Tehrani, P. et al. Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming. Cell Stem Cell 7, 64–77 (2010).

    Article  CAS  PubMed  Google Scholar 

  129. McDonald, O. G., Wu, H., Timp, W., Doi, A. & Feinberg, A. P. Genome-scale epigenetic reprogramming during epithelial-to-mesenchymal transition. Nature Struct. Mol. Biol. 18, 867–874 (2011).

    Article  CAS  Google Scholar 

  130. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  131. Li, R. et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63 (2010). Reports, together with reference 128, the opposing roles of BMP-induced MET and TGFβ-induced EMT during somatic cell reprogramming.

    Article  CAS  PubMed  Google Scholar 

  132. Ichida, J. K. et al. A small-molecule inhibitor of tgf-β signaling replaces Sox2 in reprogramming by inducing Nanog. Cell Stem Cell 5, 491–503 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Maherali, N. & Hochedlinger, K. Tgfβ signal inhibition cooperates in the induction of iPSCs and replaces Sox2 and cMyc. Curr. Biol. 19, 1718–1723 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Roberts, A. B. et al. Smad3 is key to TGF-β-mediated epithelial-to-mesenchymal transition, fibrosis, tumor suppression and metastasis. Cytokine Growth Factor Rev. 17, 19–27 (2006).

    Article  CAS  PubMed  Google Scholar 

  135. Yang, L., Pang, Y. & Moses, H. L. TGF-β and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 31, 220–227 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Li, M. O. & Flavell, R. A. TGF-β: a master of all T cell trades. Cell 134, 392–404 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Littman, D. R. & Rudensky, A. Y. Th17 and regulatory T cells in mediating and restraining inflammation. Cell 140, 845–858 (2010).

    Article  CAS  PubMed  Google Scholar 

  138. Yang, L. et al. IL-21 and TGF-β are required for differentiation of human TH17 cells. Nature 454, 350–352 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Zhou, L. et al. TGF-β-induced Foxp3 inhibits TH17 cell differentiation by antagonizing RORγt function. Nature 453, 236–240 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Koinuma, D. et al. Chromatin immunoprecipitation on microarray analysis of Smad2/3 binding sites reveals roles of ETS1 and TFAP2A in transforming growth factor β signaling. Mol. Cell. Biol. 29, 172–186 (2009).

    Article  CAS  PubMed  Google Scholar 

  141. Chen, C. R., Kang, Y., Siegel, P. M. & Massagué, J. E2F4/5 and p107 as Smad cofactors linking the TGFβ receptor to c-myc repression. Cell 110, 19–32 (2002).

    Article  CAS  PubMed  Google Scholar 

  142. Arany, P. R. et al. Smad3 deficiency alters key structural elements of the extracellular matrix and mechanotransduction of wound closure. Proc. Natl Acad. Sci. USA 103, 9250–9255 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Guasch, G. et al. Loss of TGFβ signaling destabilizes homeostasis and promotes squamous cell carcinomas in stratified epithelia. Cancer Cell 12, 313–327 (2007). Demonstrates that in the context of normal skin epithelial cells TGFβ induces growth inhibition, but in the context of pre-malignant (that is, a KRAS mutant) cells TGFβ drives apoptosis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Levy, L. & Hill, C. S. Alterations in components of the TGF-β superfamily signaling pathways in human cancer. Cytokine Growth Factor Rev. 17, 41–58 (2006).

    Article  CAS  PubMed  Google Scholar 

  145. Arnal-Estapé, A. et al. HER2 silences tumor suppression in breast cancer cells by switching expression of C/EBPβ isoforms. Cancer Res. 70, 9927–9936 (2010).

    Article  PubMed  CAS  Google Scholar 

  146. Labelle, M., Begum, S. & Hynes, R. O. Direct signaling between platelets and cancer cells induces an epithelial–mesenchymal-like transition and promotes metastasis. Cancer Cell 20, 576–590 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Sethi, N., Dai, X., Winter, C. G. & Kang, Y. Tumor-derived JAGGED1 promotes osteolytic bone metastasis of breast cancer by engaging notch signaling in bone cells. Cancer Cell 19, 192–205 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Mohammad, K. S. et al. TGF-β-RI kinase inhibitor SD-208 reduces the development and progression of melanoma bone metastases. Cancer Res. 71, 175–184 (2011).

    Article  CAS  PubMed  Google Scholar 

  149. Anido, J. et al. TGF-β receptor inhibitors target the CD44high/Id1high glioma-initiating cell population in human glioblastoma. Cancer Cell 18, 655–668 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author would like to thank the members of his laboratory for their contributions and critical reading the manuscript. The authors' research in this field is supported by grants from the US National Institutes of Health (NIH).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joan Massagué.

Ethics declarations

Competing interests

The author declares no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Joan Massagué's homepage

Glossary

Myoblasts

Mesenchymal progenitor cells that are committed to differentiate into muscle cells.

Epithelial–mesenchymal transition

(EMT). A phenotypic change that is characteristic of some developing tissues and certain forms of cancer. During EMT, cells lose intercellular junctions and apical–basal polarity, become migratory and, in the case of cancer, become invasive.

Homeobox genes

A family of genes encoding transcription factors that are essential for patterning along the anterior–posterior body axis.

Pro-B cells

Cells in the earliest stage of B cell development in the bone marrow. They are characterized by incomplete immunoglobulin heavy-chain rearrangements and are defined as CD19+ cytoplasmic immunoglobulin M (IgM)− or, sometimes, as B220+CD43+ (according the Hardy classification scheme).

Latent TGFβ complex

A complex that includes a bioactive transforming growth factor-β (TGFβ) dimer non-covalently bound to the cleaved propetide of the TGFβ biosynthetic precursor. This cleavage product in turn covalently binds to latent TGFβ-binding protein (LTBP).

Nucleoporins

Family of proteins that constitute the nuclear pore complex, which is a structure that spans the nuclear envelope in eukaryotic cells.

Polyubiquitylation

Post-translational modification of proteins that involves the covalent attachment and polymerization of ubiquitin moieties to Lys chain amino groups.

WW domains

Protein interaction domains that are found in many proteins. The WW domain is characterized by a pair of Trp residues 20–22 amino acids apart, and an invariant Pro residue within a region of 40 amino acids. WW domains interact with Pro-rich regions, including those containing phospho-Ser or phospho-Thr.

Mediator complex

A multiprotein complex that functions as a transcriptional co-activator and binds to the carboxyterminal domain of the RNA polymerase II (Pol II) holoenzyme. This complex acts as a bridge between the Pol II and transcription factors.

SWI/SNF

(Switch/sucrose nonfermentable). A chromatin-remodelling complex family that was first identified genetically in yeast as a group of genes required for mating type switching and growth on alternative sugar sources to sucrose. This complex is required for the transcriptional activation of 7% of the genome. SWI/SNF complexes exist in multiple forms made up proteins referred to as BRGl-associated factors (BAFs).

MicroRNA

(miRNA). An approximately 21–22 ribonucleotide RNA that arises from the action of the Dicer double-stranded ribonucleases on short stem–loop precursors. miRNAs initiate blocking of the targeted mRNAs, which have nucleotide sequences that are complementary to the miRNA.

Drosha

A ribonuclease III enzyme that initiates processing of microRNAs.

Autocrine

Autocrine signalling refers to when the target cell is the signal-releasing cell itself.

Primitive streak

Structure that forms during early stages of embryonic development. It establishes the first axis of symmetry and marks the beginning of gastrulation.

Melanomas

Malignant tumours derived from melanocyte precursors.

Gliomas

The most common types of malignant tumour in the brain.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Massagué, J. TGFβ signalling in context. Nat Rev Mol Cell Biol 13, 616–630 (2012). https://doi.org/10.1038/nrm3434

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm3434

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