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:

Intrinsically disordered proteins in cellular signalling and regulation

Key Points

  • Intrinsically disordered proteins (IDPs) and intrinsically disordered regions (IDRs) of proteins that may also contain structured domains mediate crucial signalling processes in eukaryotic cells.

  • Disorder is advantageous in cell signalling because disordered sequences have the potential to bind to multiple partners, often using different structures.

  • Disordered regions are relatively accessible, often contain multiple binding motifs and are frequently the sites for post-translational modification, an important mediator of the control of signalling pathways.

  • Disordered proteins have central roles in the formation of higher-order signalling assemblies and in the operation of circadian clocks.

Abstract

Intrinsically disordered proteins (IDPs) are important components of the cellular signalling machinery, allowing the same polypeptide to undertake different interactions with different consequences. IDPs are subject to combinatorial post-translational modifications and alternative splicing, adding complexity to regulatory networks and providing a mechanism for tissue-specific signalling. These proteins participate in the assembly of signalling complexes and in the dynamic self-assembly of membrane-less nuclear and cytoplasmic organelles. Experimental, computational and bioinformatic analyses combine to identify and characterize disordered regions of proteins, leading to a greater appreciation of their widespread roles in biological processes.

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: Intrinsic disorder in signalling.
Figure 2: Variable binding affinities of IDPs.
Figure 3: Response to multisite phosphorylation in intrinsically disordered regions.
Figure 4: Autoinhibition through interactions with intrinsically disordered regions.
Figure 5: Disorder mediates stress-induced translational silencing.

Similar content being viewed by others

References

  1. Wright, P. E. & Dyson, H. J. Intrinsically unstructured proteins: re-assessing the protein structure–function paradigm. J. Mol. Biol. 293, 321–331 (1999).

    Article  CAS  PubMed  Google Scholar 

  2. Dunker, A. K., Brown, C. J., Lawson, J. D., Iakoucheva, L. M. & Obradovic, Z. Intrinsic disorder and protein function. Biochemistry 41, 6573–6582 (2002).

    Article  CAS  PubMed  Google Scholar 

  3. Dyson, H. J. & Wright, P. E. Intrinsically unstructured proteins and their functions. Nature Rev. Mol. Cell Biol. 6, 197–208 (2005).

    Article  CAS  Google Scholar 

  4. van der Lee, R. et al. Classification of intrinsically disordered regions and proteins. Chem. Rev. 114, 6589–6631 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dunker, A. K., Cortese, M. S., Romero, P., Iakoucheva, L. M. & Uversky, V. N. Flexible nets. The roles of intrinsic disorder in protein interaction networks. FEBS J. 272, 5129–5148 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Kim, P. M., Sboner, A., Xia, Y. & Gerstein, M. The role of disorder in interaction networks: a structural analysis. Mol. Systems Biol. 4, 179 (2008).

    Article  Google Scholar 

  7. Iakoucheva, L. M., Brown, C. J., Lawson, J. D., Obradovic, Z. & Dunker, A. K. Intrinsic disorder in cell-signaling and cancer-associated proteins. J. Mol. Biol. 323, 573–584 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Liu, J. et al. Intrinsic disorder in transcription factors. Biochemistry 45, 6873–6888 (2006).

    Article  CAS  PubMed  Google Scholar 

  9. Galea, C. A., Wang, Y., Sivakolundu, S. G. & Kriwacki, R. W. Regulation of cell division by intrinsically unstructured proteins: intrinsic flexibility, modularity, and signaling conduits. Biochemistry 47, 7598–7609 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Gsponer, J., Futschik, M. E., Teichmann, S. A. & Babu, M. M. Tight regulation of unstructured proteins: from transcript synthesis to protein degradation. Science 322, 1365–1368 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vavouri, T., Semple, J. I., Garcia-Verdugo, R. & Lehner, B. Intrinsic protein disorder and interaction promiscuity are widely associated with dosage sensitivity. Cell 138, 198–208 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Babu, M. M., van der Lee, R., de Groot, N. S. & Gsponer, J. Intrinsically disordered proteins: regulation and disease. Curr. Opin. Struct. Biol. 21, 432–440 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Tompa, P. The interplay between structure and function in intrinsically unstructured proteins. FEBS Lett. 579, 3346–3354 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Frey, S., Richter, R. P. & Görlich, D. FG-rich repeats of nuclear pore proteins form a three-dimensional meshwork with hydrogel-like properties. Science 314, 815–817 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Tompa, P. Structure and Function of Intrinsically Disordered Proteins (Chapman & Hall/CRC, 2009).

    Book  Google Scholar 

  16. Guharoy, M., Szabo, B., Martos, S. C., Kosol, S. & Tompa, P. Intrinsic structural disorder in cytoskeletal proteins. Cytoskeleton 70, 550–571 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Kato, M. et al. Cell-free formation of RNA granules: low complexity sequence domains form dynamic fibers within hydrogels. Cell 149, 753–767 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Weber, S. C. & Brangwynne, C. P. Getting RNA and protein in phase. Cell 149, 1188–1191 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Oldfield, C. J. et al. Coupled folding and binding with α-helix-forming molecular recognition elements. Biochemistry 44, 12454–12470 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Pontius, B. W. Close encounters: why unstructured, polymeric domains can increase rates of specific macromolecular association. Trends Biochem. Sci. 18, 181–186 (1993).

    Article  CAS  PubMed  Google Scholar 

  21. Stein, A., Pache, R. A., Bernadó, P., Pons, M. & Aloy, P. Dynamic interactions of proteins in complex networks: a more structured view. FEBS J. 276, 5390–5405 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Gsponer, J. & Babu, M. M. The rules of disorder or why disorder rules. Progress Biophys. Mol. Biol. 99, 94–103 (2009).

    Article  CAS  Google Scholar 

  23. Lee, C. W., Ferreon, J. C., Ferreon, A. C., Arai, M. & Wright, P. E. Graded enhancement of p53 binding to CREB-binding protein (CBP) by multisite phosphorylation. Proc. Natl Acad. Sci. USA 107, 19290–19295 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Borg, M. et al. Polyelectrostatic interactions of disordered ligands suggest a physical basis for ultrasensitivity. Proc. Natl Acad. Sci. USA 104, 9650–9655 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Van Roey, K., Dinkel, H., Weatheritt, R. J., Gibson, T. J. & Davey, N. E. The switches. ELM resource: a compendium of conditional regulatory interaction interfaces. Sci. Signal. 6, rs7 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Van Roey, K., Gibson, T. J. & Davey, N. E. Motif switches: decision-making in cell regulation. Curr. Opin. Struct. Biol. 22, 378–385 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Oates, M. E. et al. D2P2: database of disordered protein predictions. Nucleic Acids Res. 41, D508–D516 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Marsh, J. A. & Forman-Kay, J. D. Ensemble modeling of protein disordered states: experimental restraint contributions and validation. Proteins 80, 556–572 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Salmon, L. et al. NMR characterization of long-range order in intrinsically disordered proteins. J. Am. Chem. Soc. 132, 8407–8418 (2010).

    Article  CAS  PubMed  Google Scholar 

  30. Sibille, N. & Bernadó, P. Structural characterization of intrinsically disordered proteins by the combined use of NMR and SAXS. Biochem. Soc. Trans. 40, 955–962 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Varadi, M. et al. pE-DB: a database of structural ensembles of intrinsically disordered and of unfolded proteins. Nucleic Acids Res. 42, D326–D335 (2014).

    Article  CAS  PubMed  Google Scholar 

  32. Ferreon, A. C., Moran, C. R., Gambin, Y. & Deniz, A. A. Single-molecule fluorescence studies of intrinsically disordered proteins. Methods Enzymol. 472, 179–204 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Hofmann, H. et al. Polymer scaling laws of unfolded and intrinsically disordered proteins quantified with single-molecule spectroscopy. Proc. Natl Acad. Sci. USA 109, 16155–16160 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Das, R. K. & Pappu, R. V. Conformations of intrinsically disordered proteins are influenced by linear sequence distributions of oppositely charged residues. Proc. Natl Acad. Sci. USA 110, 13392–13397 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Dyson, H. J. & Wright, P. E. Coupling of folding and binding for unstructured proteins. Curr. Opin. Struct. Biol. 12, 54–60 (2002).

    Article  CAS  PubMed  Google Scholar 

  36. Mohan, A. et al. Analysis of molecular recognition features (MoRFs). J. Mol. Biol. 362, 1043–1059 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Neduva, V. & Russell, R. B. Linear motifs: Evolutionary interaction switches. FEBS Lett. 579, 3342–3345 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Tompa, P., Davey, N. E., Gibson, T. J. & Babu, M. M. A million peptide motifs for the molecular biologist. Mol. Cell 55, 161–169 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Demarest, S. J. et al. Mutual synergistic folding in recruitment of CBP/p300 by p160 nuclear receptor coactivators. Nature 415, 549–553 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Waters, L. et al. Structural diversity in p160/CREB-binding protein coactivator complexes. J. Biol. Chem. 281, 14787–14795 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Qin, B. Y. et al. Crystal Structure of IRF-3 in complex with CBP. Structure 13, 1269–1277 (2005).

    Article  CAS  PubMed  Google Scholar 

  42. Spolar, R. S. & Record, M. T. Coupling of local folding to site-specific binding of proteins to DNA. Science 263, 777–784 (1994).

    Article  CAS  PubMed  Google Scholar 

  43. Fuxreiter, M., Simon, I., Friedrich, P. & Tompa, P. Preformed structural elements feature in partner recognition by intrinsically unstructured proteins. J. Mol. Biol. 338, 1015–1026 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Sugase, K., Dyson, H. J. & Wright, P. E. Mechanism of coupled folding and binding of an intrinsically disordered protein. Nature 447, 1021–1025 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Shammas, S., Travis, A. J. & Clarke, J. Remarkably fast coupled folding and binding of the intrinsically disordered transactivation domain of cMyb to CBP KIX. J. Phys. Chem. B 117, 13346–13356 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gianni, S., Morrone, A., Giri, R. & Brunori, M. A folding-after-binding mechanism describes the recognition between the transactivation domain of c-Myb and the KIX domain of the CREB-binding protein. Biochem. Biophys. Res. Commun. 428, 205–209 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Shammas, S. L., Travis, A. J. & Clarke, J. Allostery within a transcription coactivator is predominantly mediated through dissociation rate constants. Proc. Natl Acad. Sci. USA 111, 12049–12054 (2014).

    Article  CAS  Google Scholar 

  48. Rogers, J. M., Wong, C. T. & Clarke, J. Coupled folding and binding of the disordered protein PUMA does not require particular residual structure. J. Am. Chem. Soc. 136, 5197–5200 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Dogan, J., Schmidt, T., Mu, X., Engström, Å. & Jemth, P. Fast association and slow transitions in the interaction between two intrinsically disordered protein domains. J. Biol. Chem. 287, 34316–34324 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Iešmantavicˇius, V., Dogan, J., Jemth, P., Teilum, K. & Kjaergaard, M. Helical propensity in an intrinsically disordered protein accelerates ligand binding. Angew. Chem. Int. Ed. Engl. (2014).

  51. Schafmeister, C. E., Po, J. & Verdine, G. L. An all-hydrocarbon cross-linking system for enhancing the helicity and metabolic stability of peptides. J. Am. Chem. Soc. 122, 5891–5892 (2000).

    Article  CAS  Google Scholar 

  52. Bienkiewicz, E. A., Adkins, J. N. & Lumb, K. J. Functional consequences of preorganized helical structure in the intrinsically disordered cell-cycle inhibitor p27Kip1. Biochemistry 41, 752–759 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Baek, S. et al. Structure of the stapled p53 peptide bound to Mdm2. J. Am. Chem. Soc. 134, 103–106 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Otieno, S. & Kriwacki, R. Probing the role of nascent helicity in p27 function as a cell cycle regulator. PLoS ONE 7, e47177 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Borcherds, W. et al. Disorder and residual helicity alter p53-Mdm2 binding affinity and signaling in cells. Nature Chem. Biol. 10, 1000–1002 (2014).

    Article  CAS  Google Scholar 

  56. Bertagna, A., Toptygin, D., Brand, L. & Barrick, D. The effects of conformational heterogeneity on the binding of the Notch intracellular domain to effector proteins: a case of biologically tuned disorder. Biochem. Soc. Trans. 36, 157–166 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. Wang, Y. et al. Intrinsic disorder mediates the diverse regulatory functions of the Cdk inhibitor p21. Nature Chem. Biol. 7, 214–221 (2011).

    Article  CAS  Google Scholar 

  58. Baker, J. M. R. et al. CFTR regulatory region interacts with NBD1 predominantly via multiple transient helices. Nature Struct. Mol. Biol. 14, 738–745 (2007).

    Article  CAS  Google Scholar 

  59. Mittag, T. et al. Structure/function implications in a dynamic complex of the intrinsically disordered Sic1 with the Cdc4 subunit of an SCF ubiquitin ligase. Structure 18, 494–506 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Tompa, P. & Fuxreiter, M. Fuzzy complexes: polymorphism and structural disorder in protein–protein interactions. Trends Biochem. Sci. 33, 2–8 (2008).

    Article  CAS  PubMed  Google Scholar 

  61. Ferreon, J. C., Martinez-Yamout, M. A., Dyson, H. J. & Wright, P. E. Structural basis for subversion of cellular control mechanisms by the adenoviral E1A oncoprotein. Proc. Natl Acad. Sci. USA 106, 13260–13265 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Ishiyama, N. et al. Dynamic and static interactions between p120 catenin and E-cadherin regulate the stability of cell–cell adhesion. Cell 141, 117–128 (2010).

    Article  CAS  PubMed  Google Scholar 

  63. Ferreon, A. C., Ferreon, J. C., Wright, P. E. & Deniz, A. A. Modulation of allostery by protein intrinsic disorder. Nature 498, 390–394 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Flock, T., Weatheritt, R. J., Latysheva, N. S. & Babu, M. M. Controlling entropy to tune the functions of intrinsically disordered regions. Curr. Opin. Struct. Biol. 26, 62–72 (2014).

    Article  CAS  PubMed  Google Scholar 

  65. Mukherjee, S. P. et al. Analysis of the RelA:CBP/p300 interaction reveals its involvement in NF κ B-driven transcription. PLoS Biol. 11, e1001647 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Pelka, P., Ablack, J. N. G., Fonseca, G. J., Yousef, A. F. & Mymryk, J. S. Intrinsic structural disorder in adenovirus E1A: a viral molecular hub linking multiple diverse processes. J. Virol. 82, 7252–7263 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Berk, A. J. Recent lessons in gene expression, cell cycle control, and cell biology from adenovirus. Oncogene 24, 7673–7685 (2005).

    Article  CAS  PubMed  Google Scholar 

  68. Fax, P., Lipinski, K. S., Esche, H. & Brockmann, D. cAMP-independent activation of the adenovirus type 12 E2 promoter correlates with the recruitment of CREB-1/ATF-1, E1A12S, and CBP to the E2-CRE. J. Biol. Chem. 275, 8911–8920 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. Green, M., Panesar, N. K. & Loewenstein, P. M. The transcription-repression domain of the adenovirus E1A oncoprotein targets p300 at the promoter. Oncogene 27, 4446–4455 (2008).

    Article  CAS  PubMed  Google Scholar 

  70. Frye, J. J. et al. Electron microscopy structure of human APC/CCDH1–EMI1 reveals multimodal mechanism of E3 ligase shutdown. Nature Struct. Mol. Biol. 20, 827–835 (2013).

    Article  CAS  Google Scholar 

  71. Nussinov, R. & Tsai, C. J. Allostery in disease and in drug discovery. Cell 153, 293–305 (2013).

    Article  CAS  PubMed  Google Scholar 

  72. Motlagh, H. N., Wrabl, J. O., Li, J. & Hilser, V. J. The ensemble nature of allostery. Nature 508, 331–339 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hilser, V. J. & Thompson, E. B. Intrinsic disorder as a mechanism to optimize allosteric coupling in proteins. Proc. Natl Acad. Sci. USA 104, 8311–8315 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Garcia-Pino, A. et al. Allostery and intrinsic disorder mediate transcription regulation by conditional cooperativity. Cell 142, 101–111 (2010).

    Article  CAS  PubMed  Google Scholar 

  75. Li, J., Motlagh, H. N., Chakuroff, C., Thompson, E. B. & Hilser, V. J. Thermodynamic dissection of the intrinsically disordered N-terminal domain of human glucocorticoid receptor. J. Biol. Chem. 287, 26777–26787 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Krishnan, N. et al. Targeting the disordered C terminus of PTP1B with an allosteric inhibitor. Nature Chem. Biol. 10, 558–566 (2014).

    Article  CAS  Google Scholar 

  77. Pejaver, V. et al. The structural and functional signatures of proteins that undergo multiple events of post-translational modification. Protein Sci. 23, 1077–1093 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Iakoucheva, L. M. et al. The importance of intrinsic disorder for protein phosphorylation. Nucleic Acids Res. 32, 1037–1049 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Chrivia, J. C. et al. Phosphorylated CREB binds specifically to nuclear protein CBP. Nature 365, 855–859 (1993).

    Article  CAS  PubMed  Google Scholar 

  80. Radhakrishnan, I. et al. Solution structure of the KIX domain of CBP bound to the transactivation domain of CREB: A model for activator:coactivator interactions. Cell 91, 741–752 (1997).

    Article  CAS  PubMed  Google Scholar 

  81. Nash, P. et al. Multisite phosphorylation of a CDK inhibitor sets a threshold for the onset of DNA replication. Nature 414, 514–521 (2001).

    Article  CAS  PubMed  Google Scholar 

  82. Mittag, T. et al. Dynamic equilibrium engagement of a polyvalent ligand with a single-site receptor. Proc. Natl Acad. Sci. USA 105, 17772–17777 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Samaga, R. & Klamt, S. Modeling approaches for qualitative and semi-quantitative analysis of cellular signaling networks. Cell Commun. Signal. 11, 43 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Ferrell, J. E. Jr & Ha, S. H. Ultrasensitivity part II: multisite phosphorylation, stoichiometric inhibitors, and positive feedback. Trends Biochem. Sci. 39, 556–569 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Lyons, N. A., Fonslow, B. R., Diedrich, J. K., Yates, J. R. & Morgan, D. O. Sequential primed kinases create a damage-responsive phosphodegron on Eco1. Nature Struct. Mol. Biol. 20, 194–201 (2013).

    Article  CAS  Google Scholar 

  86. Meek, D. W. & Anderson, C. W. Posttranslational Modification of p53: Cooperative integrators of function. Cold Spring Harb. Perspect. Biol. 1, a000950 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kruse, J. P. & Gu, W. Modes of p53 regulation. Cell 137, 609–622 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Vousden, K. H. & Prives, C. Blinded by the light: The growing complexity of p53. Cell 137, 413–431 (2009).

    Article  CAS  PubMed  Google Scholar 

  89. Grossman, S. R. et al. Polyubiquitination of p53 by a ubiquitin ligase activity of p300. Science 300, 342 (2003).

    Article  CAS  PubMed  Google Scholar 

  90. Ferreon, J. C. et al. Cooperative regulation of p53 by modulation of ternary complex formation with CBP/p300 and HDM2. Proc. Natl Acad. Sci. USA 106, 6591–6596 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Sakaguchi, K. et al. Damage-mediated phosphorylation of human p53 threonine 18 through a cascade mediated by a casein 1-like kinase. Effect on Mdm2 binding. J. Biol. Chem. 275, 9278–9283 (2000).

    Article  CAS  PubMed  Google Scholar 

  92. Schon, O., Friedler, A., Freund, S. & Fersht, A. R. Binding of p53-derived ligands to MDM2 induces a variety of long range conformational changes. J. Mol. Biol. 336, 197–202 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Müller-Späth, S. et al. Charge interactions can dominate the dimensions of intrinsically disordered proteins. Proc. Natl Acad. Sci. USA 107, 14609–14614 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  94. Querfurth, C. et al. Circadian conformational change of the Neurospora clock protein FREQUENCY triggered by clustered hyperphosphorylation of a basic domain. Mol. Cell 43, 713–722 (2011).

    Article  CAS  PubMed  Google Scholar 

  95. Baker, C. L., Kettenbach, A. N., Loros, J. J., Gerber, S. A. & Dunlap, J. C. Quantitative proteomics reveals a dynamic interactome and phase-specific phosphorylation in the Neurospora circadian clock. Mol. Cell 34, 354–363 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Tang, C. T. et al. Setting the pace of the Neurospora circadian clock by multiple independent FRQ phosphorylation events. Proc. Natl Acad. Sci. USA 106, 10722–10727 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Holt, L. J. et al. Global analysis of Cdk1 substrate phosphorylation sites provides insights into evolution. Science 325, 1682–1686 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kõivomägi, M. et al. Multisite phosphorylation networks as signal processors for Cdk1. Nature Struct. Mol. Biol. 20, 1415–1424 (2013).

    Article  CAS  Google Scholar 

  99. Pufall, M. A. & Graves, B. J. Autoinhibitory domains: modular effectors of cellular regulation. Annu. Rev. Cell Dev. Biol. 18, 421–462 (2002).

    Article  CAS  PubMed  Google Scholar 

  100. Trudeau, T. et al. Structure and intrinsic disorder in protein autoinhibition. Structure 21, 332–341 (2013).

    Article  CAS  PubMed  Google Scholar 

  101. Li, P., Martins, I. R. S., Amarasinghe, G. K. & Rosen, M. K. Internal dynamics control activation and activity of the autoinhibited Vav DH domain. Nature Struct. Mol. Biol. 15, 613–618 (2008).

    Article  CAS  Google Scholar 

  102. Yu, B. et al. Structural and energetic mechanisms of cooperative autoinhibition and activation of Vav1. Cell 140, 246–256 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Wu, H. Higher-order assemblies in a new paradigm of signal transduction. Cell 153, 287–292 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Weatheritt, R. J., Gibson, T. J. & Babu, M. M. Asymmetric mRNA localization contributes to fidelity and sensitivity of spatially localized systems. Nature Struct. Mol. Biol. 21, 833–839 (2014).

    Article  CAS  Google Scholar 

  105. Li, J. et al. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 150, 339–350 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Li, P. et al. Phase transitions in the assembly of multivalent signalling proteins. Nature 483, 336–340 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Kedersha, N., Ivanov, P. & Anderson, P. Stress granules and cell signaling: more than just a passing phase? Trends Biochem. Sci. 38, 494–506 (2013).

    Article  CAS  PubMed  Google Scholar 

  108. Wippich, F. et al. Dual specificity kinase DYRK3 couples stress granule condensation/dissolution to mTORC1 signaling. Cell 152, 791–805 (2013).

    Article  CAS  PubMed  Google Scholar 

  109. Michelitsch, M. D. & Weissman, J. S. A census of glutamine/asparagine-rich regions: Implications for their conserved function and the prediction of novel prions. Proc. Natl Acad. Sci. USA 97, 11910–11915 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Wang, E. T. et al. Alternative isoform regulation in human tissue transcriptomes. Nature 456, 470–476 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Buljan, M. et al. Tissue-specific splicing of disordered segments that embed binding motifs rewires protein interaction networks. Mol. Cell 46, 871–883 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Ellis, J. D. et al. Tissue-specific alternative splicing remodels protein–protein interaction networks. Mol. Cell 46, 884–892 (2012).

    Article  CAS  PubMed  Google Scholar 

  113. Korneta, I. & Bujnicki, J. M. Intrinsic disorder in the human spliceosomal proteome. PLoS Computat. Biol. 8, e1002641 (2012).

    Article  CAS  Google Scholar 

  114. Romero, P. R. et al. Alternative splicing in concert with protein intrinsic disorder enables increased functional diversity in multicellular organisms. Proc. Natl Acad. Sci. USA 103, 8390–8395 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Mermoud, J. E., Cohen, P. & Lamond, A. I. Ser/Thr-specific protein phosphatases are required for both catalytic steps of pre-mRNA splicing. Nucleic Acids Res. 20, 5263–5269 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Xiao, S. H. & Manley, J. L. Phosphorylation of the ASF/SF2 RS domain affects both protein–protein and protein–RNA interactions and is necessary for splicing. Genes Dev. 11, 334–344 (1997).

    Article  CAS  PubMed  Google Scholar 

  117. Xiang, S. et al. Phosphorylation drives a dynamic switch in serine/arginine-rich proteins. Structure 21, 2162–2174 (2013).

    Article  CAS  PubMed  Google Scholar 

  118. Kwon, I. et al. Poly-dipeptides encoded by the C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells. Science 345, 1139–1145 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Muñoz, M. J., de la Mata, M. & Kornblihtt, A. R. The carboxy terminal domain of RNA polymerase II and alternative splicing. Trends Biochem. Sci. 35, 497–504 (2010).

    Article  CAS  PubMed  Google Scholar 

  120. Hsin, J.-P. & Manley, J. L. The RNA polymerase II CTD coordinates transcription and RNA processing. Genes Dev. 26, 2119–2137 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. David, C. J., Boyne, A. R., Millhouse, S. R. & Manley, J. L. The RNA polymerase II C-terminal domain promotes splicing activation through recruitment of a U2AF65–Prp19 complex. Genes Dev. 25, 972–983 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Dye, M. J., Gromak, N. & Proudfoot, N. J. Exon tethering in transcription by RNA polymerase II. Mol. Cell 21, 849–859 (2006).

    Article  CAS  PubMed  Google Scholar 

  123. Theillet, F. X. et al. Physicochemical properties of cells and their effects on intrinsically disordered proteins (IDPs). Chem. Rev. 114, 6661–6714 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Sakon, J. J. & Weninger, K. R. Detecting the conformation of individual proteins in live cells. Nature Methods 7, 203–205 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Gebhardt, J. C. et al. Single-molecule imaging of transcription factor binding to DNA in live mammalian cells. Nature Methods 10, 421–426 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Wells, M. et al. Structure of tumor suppressor p53 and its intrinsically disordered N-terminal transactivation domain. Proc. Natl Acad. Sci. USA 105, 5762–5767 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  127. Cheng, Y. et al. Rational drug design via intrinsically disordered protein. Trends Biotechnol. 24, 435–442 (2006).

    Article  CAS  PubMed  Google Scholar 

  128. Mészáros, B., Tompa, P., Simon, I. & Dosztányi, Z. Molecular principles of the interactions of disordered proteins. J. Mol. Biol. 372, 549–561 (2007).

    Article  CAS  PubMed  Google Scholar 

  129. Lao, B. B. et al. Rational design of topographical helix mimics as potent inhibitors of protein–protein interactions. J. Am. Chem. Soc. 136, 7877–7888 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Lao, B. B. et al. In vivo modulation of hypoxia-inducible signaling by topographical helix mimetics. Proc. Natl Acad. Sci. USA 111, 7588–7593 (2014).

    Article  CAS  Google Scholar 

  131. Hammoudeh, D. I., Follis, A. V., Prochownik, E. V. & Metallo, S. J. Multiple independent binding sites for small-molecule inhibitors on the oncoprotein c-myc. J. Am. Chem. Soc. 131, 7390–7401 (2009).

    Article  CAS  PubMed  Google Scholar 

  132. Davey, N. E., Trave, G. & Gibson, T. J. How viruses hijack cell regulation. Trends Biochem. Sci. 36, 159–169 (2011).

    Article  CAS  PubMed  Google Scholar 

  133. Hagai, T., Azia, A., Babu, M. M. & Andino, R. Use of host-like peptide motifs in viral proteins is a prevalent strategy in host-virus interactions. Cell Rep. 7, 1729–1739 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Peng, K. et al. Optimizing intrinsic disorder predictors with protein evolutionary information. J. Bioinform. Comput. Biol. 3, 35–60 (2005).

    Article  CAS  PubMed  Google Scholar 

  135. Dosztányi, Z., Csizmok, V., Tompa, P. & Simon, I. The pairwise energy content estimated from amino acid composition discriminates between folded and intrinsically unstructured proteins. J. Mol. Biol. 347, 827–839 (2005).

    Article  CAS  PubMed  Google Scholar 

  136. Dosztányi, Z., Csizmok, V., Tompa, P. & Simon, I. IUPred: web server for the prediction of intrinsically unstructured regions of proteins based on estimated energy content. Bioinformatics 21, 3433–3434 (2005).

    Article  CAS  PubMed  Google Scholar 

  137. Dosztányi, Z., Mészáros, B. & Simon, I. ANCHOR: web server for predicting protein binding regions in disordered proteins. Bioinformatics 25, 2745–2746 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Mészáros, B., Simon, I. & Dosztányi, Z. Prediction of protein binding regions in disordered proteins. PLoS Comput. Biol. 5, e1000376 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Obradovic, Z. et al. Predicting intrinsic disorder from amino acid sequence. Proteins: Struct. Function Bioinformat. 53 (Suppl. 6), 566–572 (2003).

    Article  CAS  Google Scholar 

  140. Ishida, T. & Kinoshita, K. PrDOS: prediction of disordered protein regions from amino acid sequence. Nucleic Acids Res. 35, W460–W464 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  141. Walsh, I., Martin, A. J., Di Domenico, T. & Tosatto, S. C. ESpritz: accurate and fast prediction of protein disorder. Bioinformatics 28, 503–509 (2012).

    Article  CAS  PubMed  Google Scholar 

  142. Fukuchi, S., Homma, K., Minezaki, Y., Gojobori, T. & Nishikawa, K. Development of an accurate classification system of proteins into structured and unstructured regions that uncovers novel structural domains: its application to human transcription factors. BMC Struct. Biol. 9, 26 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Wojciak, J. M., Martinez-Yamout, M. A., Dyson, H. J. & Wright, P. E. Structural basis for recruitment of CBP/p300 coactivators by STAT1 and STAT2 transactivation domains. EMBO J. 28, 948–958 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Bösch, C., Bundi, A., Oppliger, M. & Wüthrich, K. 1H nuclear-magnetic-resonance studies of the molecular conformation of monomeric glucagon in aqueous solution. Eur. J. Biochem. 91, 209–214 (1978).

    Article  Google Scholar 

  145. Altarejos, J. Y. & Montminy, M. CREB and the CRTC co-activators: sensors for hormonal and metabolic signals. Nature Rev. Mol. Cell Biol. 12, 141–151 (2011).

    Article  CAS  Google Scholar 

  146. Sickmeier, M. et al. DisProt: the database of disordered proteins. Nucleic Acids Res. 35, D786–D793 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors apologize to the many colleagues whose work could not be cited owing to space limitations. Work in the authors' laboratories was supported by grants CA096865 (P.E.W.) and GM71862 (H.J.D.) from the US National Institutes of Health and by the Skaggs Institute for Chemical Biology (P.E.W.).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Peter E. Wright or H. Jane Dyson.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Related links

Related links

DATABASES

Database of Disordered Protein Prediction (D2P2)

DisProt database

Protein Ensemble Database (pE-DB)

RCSB Protein Data Bank

2LWW

Glossary

Association rates

The speeds with which complexes are created from their component parts. The speed of the reverse process, detachment of the components, is known as the dissociation rate. The binding affinity is determined by the relative magnitude of the association and dissociation rates; for example, a fast association rate ('on-rate') and a slow dissociation rate ('off-rate') is characteristic of a high-affinity complex.

Conformational ensembles

Structural descriptions of proteins that do not have a single, well-ordered three-dimensional structure. Conformational ensembles contain a multitude of different interconverting structures, which, when averaged, are consistent with observed parameters, such as nuclear magnetic resonance (NMR) spectra or small-angle X-ray scattering data.

Binding free energy

The difference in free energy (ΔG) between the free and bound states of a complex. If the complex is stable, the binding free energy is negative.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wright, P., Dyson, H. Intrinsically disordered proteins in cellular signalling and regulation. Nat Rev Mol Cell Biol 16, 18–29 (2015). https://doi.org/10.1038/nrm3920

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm3920

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