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:

How cells read TGF-β signals

Abstract

Cell proliferation, differentiation and death are controlled by a multitude of cell?cell signals, and loss of this control has devastating consequences. Prominent among these regulatory signals is the transforming growth factor-β (TGF-β) family of cytokines, which can trigger a bewildering diversity of responses, depending on the genetic makeup and environment of the target cell. What are the networks of cell-specific molecules that mould the TGF-β response to each cell's needs?

Key Points

  • A cell can respond to a transforming growth factor-β (TGF-β) signal in a multitude of ways. This review emphasizes both the complex network of cross-talking signals that constitute it, and the importance of cellular context in determining the outcome of a signal.

  • In organisms ranging from worms to humans, the TGF-β signal is transduced to the nucleus through the action of SMADs. Different TGF-β members act through specific SMADs. The specificity of recptor?SMAD interactions is dictated by discrete structural elements in the receptor kinase domain and the MAD homology domain of the SMAD.

  • The numerous members of the TGF-β family initiate signalling by assembling a membrane receptor complex. In this complex, two type II receptor subunits phosphorylate and activate two type I receptor subunits that then propagate the signal by phosphorylating SMAD proteins.

  • In the basal state, SMADs are retained in the cytoplasm so that they are accessible to activated receptors. Upon phosphorylation, SMADs move to the nucleus where they control transcription.

  • The SMADs can potentially activate many different genes, but their affinity for DNA is too low to do that alone. Cofactors are involved that simultaneously contact a SMAD and a specific DNA sequence. The combined DNA-binding specificity of a SMAD-cofactor complex dictates the choice of target gene.

  • Whereas some SMAD cofactors function solely as DNA-binding adaptors, others have intrinsic transcription factor activity. The latter, because they are themselves regulated by extracellular signals, provide a basis for integration of different inputs at the transcriptional level.

  • SMADs recruit not only co-activators but also corepressors. They are thought to mediate repression through binding to histone deacetylases, whose effects generally lead to chromatin condensation.

  • Recent data indicates that TGF-βs may also signal through the mitogen-activated protein kinase (MAPK) pathways.

  • Further control is provided by the regulation of ligand production and negative feedback, which occurs both at the level of the TGF-β receptors and through the action of antagonistic SMADs.

  • Inputs that control the level of a SMAD signal can have both quantitative and qualitative effects on the repsonse, because some inputs can activate different sets of genes at different signal thresholds.

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: The basic SMAD pathway.
Figure 2: A network controlling a pathway.
Figure 3: SMADs interact with DNA-binding cofactors to specify TGF-β gene responses.
Figure 4: Crosstalk between the SMAD and mitogen-activated protein kinase pathways.
Figure 5: Self-modifying signals.

Similar content being viewed by others

References

  1. Massagué, J. TGFβ signal transduction. Annu. Rev. Biochem. 67, 753?791 (1998).

    Article  PubMed  Google Scholar 

  2. Schier, A. F. & Shen, M. M. Nodal signalling in vertebrate development . Nature 403, 385?389 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. Whitman, M. SMADs and early developmental signaling by the TGFβ superfamily. Genes Dev. 12, 2445?2462 (1998).

    Article  CAS  PubMed  Google Scholar 

  4. Massagué, J., Blain, S. W. & Lo, R. S. TGF-β signaling in growth control, cancer and heritable disorders. Cell 103, 295? 309 (2000).

    Article  PubMed  Google Scholar 

  5. Wrana, J. L., Attisano, L., Wieser, R., Ventura, F. & Massagué, J. Mechanism of activation of the TGF-β receptor. Nature 370, 341?347 ( 1994).Elucidation of the mechanisms of receptor activation based on a combined biochemical and genetic approach.

    Article  CAS  PubMed  Google Scholar 

  6. Wieser, R., Wrana, J. L. & Massagué, J. GS domain mutations that constitutively activate TβR-I, the downstream signaling component in the TGF-β receptor complex. EMBO J. 14, 2199?2208 ( 1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lo, R. S. & Massagué, J. Ubiquitin-dependent degradation of TGF-β-activated SMAD2. Nature Cell Biol. 1, 472?478 (1999).

    Article  CAS  PubMed  Google Scholar 

  8. Zhu, H., Kavsak, P., Abdollah, S., Wrana, J. L. & Thomsen, G. H. A SMAD ubiquitin ligase targets the BMP pathway and affects embryonic pattern formation. Nature 400, 687?693 (1999).

    Article  CAS  PubMed  Google Scholar 

  9. Raftery, L. A. & Sutherland, D. J. TGF-β family signal transduction in Drosophila development: from Mad to SMADs. Dev. Biol. 210, 251?268 ( 1999).A chronicle of the discovery of MAD, the founding member of the SMAD family.

    Article  CAS  PubMed  Google Scholar 

  10. Xu, L., Chen, Y. G. & Massagué, J. The nuclear import function of SMAD2 is masked by SARA and unmasked by TGFβ-dependent phosphorylation. Nature Cell Biol. 2, 559?562 ( 2000).

    Article  CAS  PubMed  Google Scholar 

  11. Hahn, S. A. et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 271, 350? 353 (1996).

    Article  CAS  PubMed  Google Scholar 

  12. Lagna, G., Hata, A., Hemmati-Brivanlou, A. & Massagué, J. Partnership between DPC4 and SMAD proteins in TGFβ signalling pathways . Nature 383, 832?836 (1996).Reveals the role of SMAD4 as a shared partner of receptor-activated SMADs.

    Article  CAS  PubMed  Google Scholar 

  13. Chen, X. et al. SMAD4 and FAST-1 in the assembly of activin-responsive factor . Nature 389, 85?89 (1997).Delineates the role of FAST as a SMAD DNA-binding cofactor

    Article  CAS  PubMed  Google Scholar 

  14. Liu, F., Pouponnot, C. & Massagué, J. Dual role of the SMAD4/DPC4 tumor suppressor in TGFβ-inducible transcriptional responses. Genes Dev. 11, 3157?3167 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. de Caestecker, M. P. et al. The SMAD4 activation domain (SAD) is a proline-rich, p300-dependent transcriptional activation domain. J. Biol. Chem. 275 , 2115?2122 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Janknecht, R., Wells, N. J. & Hunter, T. TGF-β-stimulated cooperation of SMAD proteins with the coactivators CBP/p300. Genes Dev. 12, 2114?2119 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Massagué, J. & Wotton, D. Transcriptional control by the TGF?β/SMAD signaling system. EMBO J. 19, 1745?1754 ( 2000).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Yahata, T. et al. The MSG1 non-DNA-binding transactivator binds to the p300/CBP coactivators, enhancing their functional link to the SMAD transcription factors . J. Biol. Chem. 275, 8825? 8834 (2000).

    Article  CAS  PubMed  Google Scholar 

  19. Tsukazaki, T., Chiang, T. A., Davison, A. F., Attisano, L. & Wrana, J. L. SARA, a FYVE domain protein that recruits SMAD2 to the TGFβ receptor. Cell 95, 779?791 (1998).Identification of a cytoplasmic regulator of SMAD movement to TGF-β receptors.

    Article  CAS  PubMed  Google Scholar 

  20. Conti, E., Uy, M., Leighton, L., Blobel, G. & Kuriyan, J. Crystallographic analysis of the recognition of a nuclear localization signal by the nuclear import factor karyopherin-α. Cell 94, 193?204 ( 1998).

    Article  CAS  PubMed  Google Scholar 

  21. Xiao, Z., Liu, X. & Lodish, H. F. Importin-β mediates nuclear translocation of SMAD 3. J. Biol. Chem. 275, 23425? 23428 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Morén, A., Itoh, S., Moustakas, A., Dijke, P. & Heldin, C. H. Functional consequences of tumorigenic missense mutations in the amino-terminal domain of SMAD4. Oncogene 19, 4396?4404 (2000).

    Article  PubMed  Google Scholar 

  23. Jones, J. B. & Kern, S. E. Functional mapping of the MH1 DNA-binding domain of DPC4/SMAD4. Nucleic Acids Res. 28, 2363?2368 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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 

  25. Watanabe, M., Masuyama, N., Fukuda, M. & Nishida, E. Regulation of intracellular dynamics of SMAD4 by its leucine-rich nuclear export signal . EMBO Rep. 1, 176?182 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Masuyama, N., Hanafusa, H., Kusakabe, M., Shibuya, H. & Nishida, E. Identification of two SMAD4 proteins in Xenopus. Their common and distinct properties. J. Biol. Chem. 274, 12163?12170 ( 1999).

    Article  CAS  PubMed  Google Scholar 

  27. Chen, Y. G. et al. Determinants of specificity in TGF-β signal transduction . Genes Dev. 12, 2144?2152 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Graff, J. M., Bansal, A. & Melton, D. A. Xenopus Mad proteins transduce distinct subsets of signals for the TGF-β superfamily. Cell 85, 479?487 (1996).The opposing but complementary roles of SMAD1 and SMAD2 are revealed.

    Article  CAS  PubMed  Google Scholar 

  29. Baker, J. & Harland, R. M. A novel mesoderm inducer, mMadr-2, functions in the activin signal transduction pathway. Genes Dev. 10, 1880?1889 ( 1996).

    Article  CAS  PubMed  Google Scholar 

  30. Rodriguez Esteban, C. et al. The novel Cer-like protein Caronte mediates the establishment of embryonic left?right asymmetry. Nature 401 , 243?251 (1999).

    Article  CAS  PubMed  Google Scholar 

  31. Yokouchi, Y., Vogan, K. J., Pearse, R. V. & Tabin, C. J. Antagonistic signaling by Caronte, a novel Cerberus-related gene, establishes left?right asymmetric gene expression. Cell 98 , 573?583 (1999).

    Article  CAS  PubMed  Google Scholar 

  32. Saijoh, Y. et al. Left?right assymetric expression of lefty2 and nodal is induced by a signaling pathway that includes the transcription factor FAST2 . Mol. Cell 5, 35?47 (2000).References 31 and 32 report on regulation of BMP activity by Hedgehog in the extracellular space.

    Article  CAS  PubMed  Google Scholar 

  33. Feng, X. H. & Derynck, R. A kinase subdomain of transforming growth factor-β (TGF-β) type I receptor determines the TGF-β intracellular signaling activity. EMBO J. 16, 3912?3922 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lo, R. S., Chen, Y. G., Shi, Y. G., Pavletich, N. & Massagué, J. The L3 loop: a structural motif determining specific interactions between SMAD proteins and TGF-β receptors. EMBO J. 17, 996?1005 ( 1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yagi, K. et al. Alternatively spliced variant of SMAD2 lacking exon 3. Comparison with wild-type SMAD2 and SMAD3. J. Biol.Chem. 274, 703?709 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Ashcroft, G. S. et al. Mice lacking SMAD3 show accelerated wound healing and an impaired local inflammatory response. Nature Cell Biol. 1, 260?266 (1999).

    Article  CAS  PubMed  Google Scholar 

  37. Denissova, N. G., Pouponnot, C., Long, J., He, D. & Liu, F. Transforming growth factor β-inducible independent binding of SMAD to the SMAD7 promoter. Proc. Natl Acad. Sci. USA 97, 6397?6402 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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 

  39. Germain, S., Howell, M., Esslemont, G. M. & Hill, C. S. Homeodomain and winged-helix transcription factors recruit activated SMADs to distinct promoter elements via a common SMAD interaction motif. Genes Dev. 14, 435?451 ( 2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Yeo, C. Y., Chen, X. & Whitman, M. The role of FAST-1 and SMADs in transcriptional regulation by activin during early Xenopus embryogenesis. J. Biol. Chem. 274, 26584?26590 ( 1999).

    Article  CAS  PubMed  Google Scholar 

  41. Hua, X., Miller, Z. A., Wu, G., Shi, Y. & Lodish, H. F. Specificity in transforming growth factor β-induced transcription of the plasminogen activator inhibitor-1 gene: interactions of promoter DNA, transcription factor muE3, and SMAD proteins. Proc. Natl Acad. Sci. USA 96, 13130? 13135 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chen, X., Rubock, M. J. & Whitman, M. A transcriptional partner of MAD proteins in TGF-β signalling. Nature 383, 691? 696 (1996).

    Article  CAS  PubMed  Google Scholar 

  43. Zhang, Y., Feng, X. H. & Derynck, R. SMAD3 and SMAD4 cooperate with c-Jun/c-Fos to mediate TGF-β-induced transcription. Nature 394, 909?913 (1998).

    Article  CAS  PubMed  Google Scholar 

  44. Tsuji, K., Ito, Y. & Noda, M. Expression of the PEBP2αA/AML3/CBFA1 gene is regulated by BMP4/7 heterodimer and its overexpression suppresses type I collagen and osteocalcin gene expression in osteoblastic and nonosteoblastic mesenchymal cells. Bone 22, 87?92 (1998).

    Article  CAS  PubMed  Google Scholar 

  45. Pardali, E. et al. SMAD and AML proteins synergistically confer transforming growth factor β1 responsiveness to human germ-line IgA genes. J. Biol. Chem. 275, 3552?3560 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Hanai, J. et al. Interaction and functional cooperation of PEBP2/CBF with SMADs. Synergistic induction of the immunoglobulin germline Cα promoter. J. Biol. Chem. 274, 31577?31582 (1999).

    Article  CAS  PubMed  Google Scholar 

  47. Nishita, M. et al. Interaction between Wnt and TGF-β signalling pathways during formation of Spemann's organizer. Nature 403 , 781?785 (2000). Evidence for direct cooperation between mediators of the TGF-β and Wnt pathways.

    Article  CAS  PubMed  Google Scholar 

  48. Labbe, 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  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wotton, D., Lo, R. S., Lee, S. & Massagué, J. A SMAD transcriptional corepressor. Cell 97, 29 ?39 (1999).The first SMAD transcriptional corepressor and its competition with co-activators.

    Article  CAS  PubMed  Google Scholar 

  50. Luo, K. et al. The ski oncoprotein interacts with the SMAD proteins to repress TGF-β signaling. Genes Dev. 13, 2196 ?2206 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sun, Y., Liu, X., Ng-Eaton, E., Lodish, H. F. & Weinberg, R. A. SnoN and Ski protooncoproteins are rapidly degraded in response to transforming growth factor-β signaling. Proc. Natl Acad. Sci. USA 96, 12442?12447 (1999).References 50 and 51 report on the proto-oncogene SKI as a corepressor of SMADs in the basal state.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Perou, C. M. et al. Distinctive gene expression patterns in human mammary epithelial cells and breast cancers. Proc. Natl Acad. Sci. USA 96, 9212?9217 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  54. Lo, R. S., Wotton, D. & Massagué, J. EGF signaling via Ras stabilizes the SMAD transcriptional corepressor TGIF. EMBO J. (in the press).

  55. Gripp, K. W. et al. Mutations in TGIF cause holoprosencephaly and link nodal signalling to human neural axis determination. Nature Genet. 25 , 205?208 (2000).

    Article  CAS  PubMed  Google Scholar 

  56. Sampath, K. et al. Induction of the zebrafish ventral brain and floorplate requires cyclops/nodal signalling. Nature 395, 185 ?189 (1998).

    Article  CAS  PubMed  Google Scholar 

  57. Nomura, M. & Li, E. SMAD2 role in mesoderm formation, left?right patterning and craniofacial development. Nature 393 , 786?790 (1998).

    Article  CAS  PubMed  Google Scholar 

  58. Hocevar, B. A., Brown, T. L. & Howe, P. H. TGF-β induces fibronectin synthesis through a c-Jun N-terminal kinase-dependent, SMAD4-independent pathway. EMBO J. 18, 1345?1356 ( 1999).An example of rapid activation of JNK by TGF-β and its effects on gene expression.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hanafusa, H. et al. Involvement of the p38 mitogen-activated protein kinase pathway in transforming growth factor-β-induced gene expression. J. Biol. Chem. 274, 27161?27167 (1999).

    Article  CAS  PubMed  Google Scholar 

  60. Sano, Y. et al. ATF-2 is a common nuclear target of SMAD and TAK1 pathways in transforming growth factor-β signaling. J. Biol. Chem. 274, 8949?8957 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Takatsu, Y. et al. TAK1 participates in c-Jun N-terminal kinase signaling during Drosophila development. Mol. Cell. Biol. 20, 3015?3026 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Shibuya, H. et al. TAB1: an activator of the TAK1 MAPKKK in TGF-β signal transduction. Science 272, 1179? 1182 (1996).

    Article  CAS  PubMed  Google Scholar 

  63. Yamaguchi, K. et al. XIAP, a cellular member of the inhibitor of apoptosis protein family, links the receptors to TAB1?TAK1 in the BMP signaling pathway . EMBO J. 18, 179?187 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Engel, M. E., McDonnell, M. A., Law, B. K. & Moses, H. L. Interdependent SMAD and JNK signaling in transforming growth factor-β-mediated transcription. J. Biol. Chem. 274, 37413 ?37420 (1999).

    Article  CAS  PubMed  Google Scholar 

  65. Wong, C. et al. SMAD3?SMAD4 and AP-1 complexes synergize in transcriptional activation of the c-Jun promoter by transforming growth factor-β. Mol. Cell. Biol. 19, 1821?1830 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Massagué, J. & Chen, Y. G. Controlling TGF-β signaling. Genes Dev. 14, 627? 644 (2000).

    PubMed  Google Scholar 

  67. ten Dijke, P., Miyazono, K. & Heldin, C. H. Signaling inputs converge on nuclear effectors in TGF-β signaling. Trends Biochem. Sci. 25, 64?70 (2000).

    Article  CAS  PubMed  Google Scholar 

  68. Tanimoto, H., Itoh, S., ten Dijke, P. & Tabata, T. Hedgehog creates a gradient of DPP activity in Drosophila wing imaginal discs. Mol. Cell 5, 59?71 ( 2000).Crosstalk between the Hedgehog and the BMP system at the level of Decapentaplegic receptors.

    Article  CAS  PubMed  Google Scholar 

  69. Hayashi, H. et al. The MAD-related protein SMAD7 associates with the TGFβ receptor and functions as an antagonist of TGFβ signaling. Cell 89, 1165?1173 ( 1997).

    Article  CAS  PubMed  Google Scholar 

  70. Nakao, A. et al. Identification of SMAD7, a TGFβ-inducible antagonist of TGF-β signaling. Nature 389, 631? 635 (1997).First evidence of antagonistic SMADs relaying feedback signals.

    Article  CAS  PubMed  Google Scholar 

  71. Ulloa, L., Doody, J. & Massagué, J. Inhibition of transforming growth factor-β/SMAD signalling by the interferon-γ/STAT pathway. Nature 397, 710?713 (1999). Negative regulation of the SMAD pathway by the STAT pathway.

    Article  CAS  PubMed  Google Scholar 

  72. Bitzer, M. et al. A mechanism of suppression of TGF-β/SMAD signaling by NFκB/RelA. Genes Dev. 14, 187? 197 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Kretzschmar, M., Doody, J. & Massagué, J. Opposing BMP and EGF signalling pathway converge on the TGFβ family mediator SMAD1. Nature 389, 618?622 (1997).Negative effects of Ras/ERK pathway on SMAD function.

    Article  CAS  PubMed  Google Scholar 

  74. Kretzschmar, M., Doody, J., Timokhina, I. & Massagué, J. A mechanism of repression of TGF-β/SMAD signaling by oncogenic ras. Genes Dev. 13, 804?816 ( 1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Halfon, M. S. et al. Ras pathway specificity is determined by the integration of multiple signal-activated and tissue-restricted transcription factors. Cell 103, 63?74 ( 2000).

    Article  CAS  PubMed  Google Scholar 

  76. Onichtchouk, D. et al. Silencing of TGF-β signalling by the pseudoreceptor BAMBI . Nature 401, 480?485 (1999).

    Article  CAS  PubMed  Google Scholar 

  77. Tsuneizumi, K. et al. Daughters against dpp modulates dpp organizing activity in Drosophila wing development. Nature 389, 627?631 (1997).

    Article  CAS  PubMed  Google Scholar 

  78. Hata, A., Lagna, G., Massagué, J. & Hemmati-Brivanlou, A. SMAD6 inhibits BMP/SMAD1 signaling by specifically competing with the SMAD4 tumor suppressor. Genes Dev. 12, 186? 197 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Yamada, M., Szendro, P. I., Prokscha, A., Schwartz, R. J. & Eichele, G. Evidence for a role of SMAD6 in chick cardiac development. Dev. Biol. 215, 48?61 (1999).

    Article  CAS  PubMed  Google Scholar 

  80. Zhao, J., Shi, W., Chen, H. & Warburton, D. SMAD7 and SMAD6 differentially modulate transforming growth factor-β-induced inhibition of embryonic lung morphogenesis. J. Biol. Chem. 275 , 23992?23997 (2000).

    Article  CAS  PubMed  Google Scholar 

  81. Galvin, K. M. et al. A role for SMAD6 in development and homeostasis of the cardiovascular system. Nature Genet. 24, 171? 174 (2000).

    Article  CAS  PubMed  Google Scholar 

  82. Imamura, T. et al. SMAD6 inhibits signalling by the TGFβ superfamily. Nature 389, 622?626 ( 1997).

    Article  CAS  PubMed  Google Scholar 

  83. López-Casillas, F., Wrana, J. L. & Massagué, J. Betaglycan presents ligand to the TGF-β signaling receptor. Cell 73, 1435? 1444 (1993).

    Article  PubMed  Google Scholar 

  84. Lewis, K. A. et al. Betaglycan binds inhibin and can mediate functional antagonism of activin signalling. Nature 404, 411? 414 (2000).The surprising versatility of a TGF-β accessory receptor is revealed.

    Article  CAS  PubMed  Google Scholar 

  85. Zúñiga, A., Haramis, A. P., McMahon, A. P. & Zeller, R. Signal relay by BMP antagonism controls the SHH/FGF4 feedback loop in vertebrate limb buds. Nature 401, 598? 602 (1999).

    Article  PubMed  Google Scholar 

  86. Gazzerro, E., Gangji, V. & Canalis, E. Bone morphogenetic proteins induce the expression of noggin, which limits their activity in cultured rat osteoblasts. J. Clin. Invest. 102, 2106?2114 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Bilezikjian, L. M., Corrigan, A. Z., Blount, A. L. & Vale, W. W. Pituitary follistatin and inhibin subunit messenger ribonucleic acid levels are differentially regulated by local and hormonal factors. Endocrinology 137, 4277?4284 ( 1996).

    Article  CAS  PubMed  Google Scholar 

  88. Crawford, S. E. et al. Thrombospondin-1 is a major activator of TGF-β1 in vivo. Cell 93, 1159? 1170 (1998).

    Article  CAS  PubMed  Google Scholar 

  89. Green, J. B. A., New, H. V. & Smith, J. C. Responses of embryonic Xenopus cells to activin and FGF are separated by multiple dose thresholds and correspond to distinct axes of the mesoderm. Cell 71, 731? 739 (1992).

    Article  CAS  PubMed  Google Scholar 

  90. Dyson, S. & Gurdon, J. B. The interpretation of position in a morphogen gradient as revealed by occupancy of activin receptors. Cell 93, 557?568 ( 1998).A saga showing the morphogenetic potential of activin signalling during Xenopus embryogenesis.

    Article  CAS  PubMed  Google Scholar 

  91. Zhao, J. & Buick, R. N. Regulation of transforming growth factor-β receptors in H-ras oncogene-transformed rat intestinal epithelial cells. Cancer Res. 55, 6181? 6188 (1995).

    CAS  PubMed  Google Scholar 

  92. Cornell, R. A. & Kimelman, D. Activin-mediated mesoderm induction requires FGF. Development 120, 453?462 (1994).

    CAS  PubMed  Google Scholar 

  93. LaBonne, C. & Whitman, M. Mesoderm induction by activin requires FGF-mediated intracellular signals. Development 120 , 463?472 (1994).

    CAS  PubMed  Google Scholar 

  94. Oft, M. et al. TGF-β1 and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells. Genes Dev. 10, 2462?2477 ( 1996).TGF-β is shown to instigate tumorigenic behaviour in transformed mammary cells.

    Article  CAS  PubMed  Google Scholar 

  95. Nakashima, K. et al. Synergistic signaling in fetal brain by STAT3?SMAD1 complex bridged by p300. Science 284, 479 ?482 (1999).

    Article  CAS  PubMed  Google Scholar 

  96. Shi, M. J. & Stavnezer, J. CBF α3 (AML2) is induced by TGF-β1 to bind and activate the mouse germline Igα promoter . J. Immunol. 161, 6751? 6760 (1998).

    CAS  PubMed  Google Scholar 

  97. Otto, F. et al. Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell 89, 765?771 ( 1997).

    Article  CAS  PubMed  Google Scholar 

  98. Komori, T. et al. Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts. Cell 89, 755?764 ( 1997).

    Article  CAS  PubMed  Google Scholar 

  99. Lee, B. et al. Missense mutations abolishing DNA binding of the osteoblast-specific transcription factor OSF2/CBFA1 in cleidocranial dysplasia. Nature Genet. 16, 307?310 ( 1997).

    Article  CAS  PubMed  Google Scholar 

  100. Mundlos, S. et al. Mutations involving the transcription factor CBFA1 cause cleidocranial dysplasia. Cell 89, 773? 779 (1997).

    Article  CAS  PubMed  Google Scholar 

  101. Laurent, M. N., Blitz, I. L., Hashimoto, C., Rothbacher, U. & Cho, K. W. The Xenopus homeobox gene twin mediates Wnt induction of goosecoid in establishment of Spemann's organizer. Development 124, 4905? 4916 (1997).

    CAS  PubMed  Google Scholar 

  102. Henry, G. L. & Melton, D. A. Mixer, a homeobox gene required for endoderm development. Science 281, 91?96 (1998).

    Article  CAS  PubMed  Google Scholar 

  103. Huse, M., Chen, Y. G., Massagué, J. & Kuriyan, J. Crystal structure of the cytoplasmic domain of the type I TGF-β receptor in complex with FKBP12. Cell 96, 425? 436 (1999).

    Article  CAS  PubMed  Google Scholar 

  104. Shi, Y., Hata, A., Lo, R. S., Massagué, J. & Pavletich, N. P. A structural basis for mutational inactivation of the tumour suppressor SMAD4. Nature 388, 87?93 (1997).

    Article  CAS  PubMed  Google Scholar 

  105. Wu, G. et al. Structural basis of SMAD2 recognition by the SMAD Anchor for Receptor Activation. Science 287, 92? 97 (2000).

    Article  CAS  PubMed  Google Scholar 

  106. Kim, J., Johnson, K., Chen, H. J., Carroll, S. & Laughon, A. Drosophila Mad binds to DNA and directly mediates activation of vestigial by Decapentaplegic. Nature 388, 304?308 (1997).

    Article  CAS  PubMed  Google Scholar 

  107. Liu, F. et al. A human Mad protein acting as a BMP-regulated transcriptional activator. Nature 381, 620? 623 (1996).

    Article  CAS  PubMed  Google Scholar 

  108. Dou, C. et al. BF-1 interferes with transforming growth factor-β signaling by associating with SMAD partners. Mol. Cell. Biol. 20, 6201?6211 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Kim, R. H. et al. A novel SMAD nuclear interacting protein, SNIP1, suppresses p300-dependent TGF-β signal transduction. Genes Dev. 14, 1605?1616 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASE LINKS

TGF-β

SMADs

type I receptors

type II receptors

GS region

SMURF1

BMPs

GDFs

AMH

SMAD1

SMAD5

SMAD8

Activin

Nodal

SMAD2

SMAD3

SMAD4

p300

CBP

MSG-1

SARA

FYVE domain

importin-α

Xnr-1

Xnr-2

Vg1

BMP4

SMAD7

OAZ

FAST

Mixer

JUNB

TFE3

AML

LEF1

Wnts

β-catenin

Xtwn

TGIF

SKI

SnoN

ERK

HPE

cyclops

JNK

p38

MKK4

MKK3

TAK1

Interleukin-1

Rho family

Jun

Fos

ATF2

fibronectin

TNF-α

HH

DPP

thickveins

IFN-γ

JAK

STAT

NK-κB

Ras

EGF

MAD

wingless

twist

tinman

pointed

Drosophila EGF receptor

eve

BAMBI

SMAD6

LAP

Follistatin

Noggin

Chordin

Caronte

Cerberus

Gremlin

Betaglycan

Endoglin

Inhibin

Thrombospondin

CBFA1

CBFA2

CBFA3

IgCα

cleidocranial dyplasia

BMP7

Goosecoid

FURTHER INFORMATION

Massagué lab homepage

ENCYCLOPEDIA OF LIFE SCIENCES

Bone morphogenetic proteins and their receptors

Vertebrate embryo: Establishment of left?right asymmetry

Xenopus embryo: Neural induction

Glossary

SMADS

A family of transcription factors that mediate TGF-β signals. The term SMAD is derived from the founding members of this family, the Drosophila protein MAD (Mothers Against Decapentaplegic) and the Caenorhabditis elegans protein SMA (Small body size).

GS REGION

Regulatory region in TGF-β receptors.

UBIQUITYLATION

The attachment of the protein ubiquitin to lysine residues of other molecules, often as a tag for their rapid cellular degradation.

PROTEASOME

Protein complex responsible for degrading intracellular proteins that have been tagged for destruction by the addition of ubiquitin.

UBIQUITIN LIGASE

An enzyme that couples the small protein ubiquitin to lysine residues on a target protein, marking that protein for destruction by the proteasome.

NUCLEAR LOCALIZATION SIGNAL

A 7?9 residue sequence within a protein, rich in basic residues, which acts as a signal for localization of the protein within the nucleus.

MESODERM

The middle of the three embryonic germ layers, and the source of structures including bone, muscle, connective tissue and dermis.

MH1 AND MH2 DOMAIN

Conserved amino-terminal and carboxy-terminal globular domains, respectively, of SMAD proteins.

ENHANCER ELEMENT

Sequence in the regulatory region of a gene, recognized by factors that enhance the activity of the transcriptional promoter.

HYPOMORPHIC ALLELE

A mutant gene having a similar but weaker function than the wild-type gene.

IMAGINAL DISC

Single-cell layer epithelial structures of the Drosophila larva that give rise to wings, legs and other appendages.

CHONDROCYTE

Differentiated cell of cartilage tissue.

OSTEOBLAST

A mesenchymal cell with capacity to differentiate into bone tissue.

ECTODERM

The outer of the three embryonic germ layers, which gives rise to epidermis and neural tissue.

ASTROCYTES

Star-shaped cells that support the tissue of the central nervous system.

IMMUNOGLOBULIN-α CONSTANT REGION

Region of an antibody molecule that is constant within ? and defines ? each of the basic classes of immunoglobulin.

ANTIBODY CLASS SWITCHING

Process by which the region of an immunoglobulin heavy-chain gene that encodes the antigen recognizing (variable) portion is recombined with the constant region of a different immunoglobulin class.

MESENCHYME

Loosely organized, undifferentiated mesodermal cells.

MESOENDODERM

Gives rise to both the mesoderm and the endodermal tissue of the embryo.

DORSAL MARGINAL ZONE

Region of the Xenopus embryo that gives rise to the dorsal mesoderm.

GASTRULA

Multilayered embryo with an outer cell layer (ectoderm), an inner cell layer (endoderm), and an intermediate cell layer (mesoderm).

Rights and permissions

Reprints and permissions

About this article

Cite this article

Massagué, J. How cells read TGF-β signals. Nat Rev Mol Cell Biol 1, 169–178 (2000). https://doi.org/10.1038/35043051

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/35043051

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