Skip to main content
Log in

Multi-functionality of proteins involved in GPCR and G protein signaling: making sense of structure–function continuum with intrinsic disorder-based proteoforms

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

GPCR–G protein signaling system recognizes a multitude of extracellular ligands and triggers a variety of intracellular signaling cascades in response. In humans, this system includes more than 800 various GPCRs and a large set of heterotrimeric G proteins. Complexity of this system goes far beyond a multitude of pair-wise ligand–GPCR and GPCR–G protein interactions. In fact, one GPCR can recognize more than one extracellular signal and interact with more than one G protein. Furthermore, one ligand can activate more than one GPCR, and multiple GPCRs can couple to the same G protein. This defines an intricate multifunctionality of this important signaling system. Here, we show that the multifunctionality of GPCR–G protein system represents an illustrative example of the protein structure–function continuum, where structures of the involved proteins represent a complex mosaic of differently folded regions (foldons, non-foldons, unfoldons, semi-foldons, and inducible foldons). The functionality of resulting highly dynamic conformational ensembles is fine-tuned by various post-translational modifications and alternative splicing, and such ensembles can undergo dramatic changes at interaction with their specific partners. In other words, GPCRs and G proteins exist as sets of conformational/basic, inducible/modified, and functioning proteoforms characterized by a broad spectrum of structural features and possessing various functional potentials.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10

Similar content being viewed by others

References

  1. Bjarnadottir TK, Gloriam DE, Hellstrand SH, Kristiansson H, Fredriksson R, Schioth HB (2006) Comprehensive repertoire and phylogenetic analysis of the G protein-coupled receptors in human and mouse. Genomics 88(3):263–273. https://doi.org/10.1016/j.ygeno.2006.04.001

    Article  CAS  PubMed  Google Scholar 

  2. Anantharaman V, Abhiman S, de Souza RF, Aravind L (2011) Comparative genomics uncovers novel structural and functional features of the heterotrimeric GTPase signaling system. Gene 475(2):63–78. https://doi.org/10.1016/j.gene.2010.12.001

    Article  CAS  PubMed  Google Scholar 

  3. Southan C, Sharman JL, Benson HE, Faccenda E, Pawson AJ, Alexander SP, Buneman OP, Davenport AP, McGrath JC, Peters JA, Spedding M, Catterall WA, Fabbro D, Davies JA, Nc I (2016) The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. Nucleic Acids Res 44(D1):D1054–D1068. https://doi.org/10.1093/nar/gkv1037

    Article  CAS  PubMed  Google Scholar 

  4. Fredriksson R, Lagerstrom MC, Lundin LG, Schioth HB (2003) The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol 63(6):1256–1272. https://doi.org/10.1124/mol.63.6.1256

    Article  CAS  PubMed  Google Scholar 

  5. Flock T, Hauser AS, Lund N, Gloriam DE, Balaji S, Babu MM (2017) Selectivity determinants of GPCR–G-protein binding. Nature 545(7654):317–322. https://doi.org/10.1038/nature22070

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Isberg V, de Graaf C, Bortolato A, Cherezov V, Katritch V, Marshall FH, Mordalski S, Pin JP, Stevens RC, Vriend G, Gloriam DE (2015) Generic GPCR residue numbers—aligning topology maps while minding the gaps. Trends Pharmacol Sci 36(1):22–31. https://doi.org/10.1016/j.tips.2014.11.001

    Article  CAS  PubMed  Google Scholar 

  7. Neves SR, Ram PT, Iyengar R (2002) G protein pathways. Science 296(5573):1636–1639. https://doi.org/10.1126/science.1071550

    Article  CAS  PubMed  Google Scholar 

  8. Marinissen MJ, Gutkind JS (2001) G-protein-coupled receptors and signaling networks: emerging paradigms. Trends Pharmacol Sci 22(7):368–376. https://doi.org/10.1016/S0165-6147(00)01678-3

    Article  CAS  PubMed  Google Scholar 

  9. Latorraca NR, Venkatakrishnan AJ, Dror RO (2017) GPCR dynamics: structures in motion. Chem Rev 117(1):139–155. https://doi.org/10.1021/acs.chemrev.6b00177

    Article  CAS  PubMed  Google Scholar 

  10. Flower DR (1999) Modelling G-protein-coupled receptors for drug design. Biochim Biophys Acta 1422(3):207–234. https://doi.org/10.1016/S0304-4157(99)00006-4

    Article  CAS  PubMed  Google Scholar 

  11. Fischer E (1894) Einfluss der configuration auf die wirkung der enzyme. Ber Dt Chem Ges 27:2985–2993

    Article  CAS  Google Scholar 

  12. Uversky VN (2002) Natively unfolded proteins: a point where biology waits for physics. Protein Sci 11(4):739–756. https://doi.org/10.1110/ps.4210102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Uversky VN (2002) What does it mean to be natively unfolded? Eur J Biochem 269(1):2–12. https://doi.org/10.1046/j.0014-2956.2001.02649.x

    Article  CAS  PubMed  Google Scholar 

  14. Dunker AK, Lawson JD, Brown CJ, Williams RM, Romero P, Oh JS, Oldfield CJ, Campen AM, Ratliff CM, Hipps KW, Ausio J, Nissen MS, Reeves R, Kang C-H, Kissinger CR, Bailey RW, Griswold MD, Chiu W, Garner EC, Obradovic Z (2001) Intrinsically disordered protein. J Mol Graph Model 19(1):26–59. https://doi.org/10.1016/S1093-3263(00)00138-8

    Article  CAS  PubMed  Google Scholar 

  15. Tompa P (2003) The functional benefits of protein disorder. J Mol Struct Theochem 666:361–371. https://doi.org/10.1016/j.theochem.2003.08.047

    Article  CAS  Google Scholar 

  16. Uversky VN, Gillespie JR, Fink AL (2000) Why are “natively unfolded” proteins unstructured under physiologic conditions? Proteins 41(3):415–427. https://doi.org/10.1002/1097-0134(20001115)41:3%3c415:AID-PROT130%3e3.0.CO;2-7

    Article  CAS  PubMed  Google Scholar 

  17. Wright PE, Dyson HJ (1999) Intrinsically unstructured proteins: re-assessing the protein structure–function paradigm. J Mol Biol 293(2):321–331. https://doi.org/10.1006/jmbi.1999.3110

    Article  CAS  PubMed  Google Scholar 

  18. Xue B, Williams RW, Oldfield CJ, Dunker AK, Uversky VN (2010) Archaic chaos: intrinsically disordered proteins in Archaea. BMC Syst Biol 4(Suppl 1):S1. https://doi.org/10.1186/1752-0509-4-S1-S1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. van der Lee R, Buljan M, Lang B, Weatheritt RJ, Daughdrill GW, Dunker AK, Fuxreiter M, Gough J, Gsponer J, Jones DT, Kim PM, Kriwacki RW, Oldfield CJ, Pappu RV, Tompa P, Uversky VN, Wright PE, Babu MM (2014) Classification of intrinsically disordered regions and proteins. Chem Rev 114(13):6589–6631. https://doi.org/10.1021/cr400525m

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Habchi J, Tompa P, Longhi S, Uversky VN (2014) Introducing protein intrinsic disorder. Chem Rev 114(13):6561–6588. https://doi.org/10.1021/cr400514h

    Article  CAS  PubMed  Google Scholar 

  21. Dunker AK, Garner E, Guilliot S, Romero P, Albrecht K, Hart J, Obradovic Z, Kissinger C, Villafranca JE (1998) Protein disorder and the evolution of molecular recognition: theory, predictions and observations. Pac Symp Biocomput 473–484

  22. Tompa P (2002) Intrinsically unstructured proteins. Trends Biochem Sci 27(10):527–533. https://doi.org/10.1016/S0968-0004(02)02169-2

    Article  CAS  PubMed  Google Scholar 

  23. Daughdrill GW, Pielak GJ, Uversky VN, Cortese MS, Dunker AK (2005) Natively disordered proteins. In: Buchner J, Kiefhaber T (eds) Handbook of protein folding. Wiley, Weinheim, pp 271–353

    Google Scholar 

  24. Uversky VN (2013) Unusual biophysics of intrinsically disordered proteins. Biochim Biophys Acta 1834(5):932–951. https://doi.org/10.1016/j.bbapap.2012.12.008

    Article  CAS  PubMed  Google Scholar 

  25. Uversky VN, Dunker AK (2013) The case for intrinsically disordered proteins playing contributory roles in molecular recognition without a stable 3D structure. F1000 Biol Rep 5:1. https://doi.org/10.3410/b5-1

    Article  PubMed  PubMed Central  Google Scholar 

  26. Peng Z, Yan J, Fan X, Mizianty MJ, Xue B, Wang K, Hu G, Uversky VN, Kurgan L (2015) Exceptionally abundant exceptions: comprehensive characterization of intrinsic disorder in all domains of life. Cell Mol Life Sci 72(1):137–151. https://doi.org/10.1007/s00018-014-1661-9

    Article  CAS  PubMed  Google Scholar 

  27. Na I, Redmon D, Kopa M, Qin Y, Xue B, Uversky VN (2013) Ordered disorder of the astrocytic dystrophin-associated protein complex in the norm and pathology. PLoS One 8(8):e73476. https://doi.org/10.1371/journal.pone.0073476

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ward JJ, Sodhi JS, McGuffin LJ, Buxton BF, Jones DT (2004) Prediction and functional analysis of native disorder in proteins from the three kingdoms of life. J Mol Biol 337(3):635–645. https://doi.org/10.1016/j.jmb.2004.02.002

    Article  CAS  PubMed  Google Scholar 

  29. Dunker AK, Obradovic Z, Romero P, Garner EC, Brown CJ (2000) Intrinsic protein disorder in complete genomes. Genome Inform Ser Workshop Genome Inform 11:161–171

    CAS  PubMed  Google Scholar 

  30. Hu G, Wang K, Song J, Uversky VN, Kurgan L (2018) Taxonomic landscape of the dark proteomes: whole-proteome scale interplay between structural darkness, intrinsic disorder, and crystallization propensity. Proteomics 18(21–22):e1800243. https://doi.org/10.1002/pmic.201800243

    Article  CAS  PubMed  Google Scholar 

  31. Dunker AK, Brown CJ, Lawson JD, Iakoucheva LM, Obradovic Z (2002) Intrinsic disorder and protein function. Biochemistry 41(21):6573–6582. https://doi.org/10.1021/bi012159+

    Article  CAS  PubMed  Google Scholar 

  32. Dunker AK, Brown CJ, Obradovic Z (2002) Identification and functions of usefully disordered proteins. Adv Protein Chem 62:25–49. https://doi.org/10.1016/S0065-3233(02)62004-2

    Article  CAS  PubMed  Google Scholar 

  33. Uversky VN (2003) Protein folding revisited. A polypeptide chain at the folding-misfolding-nonfolding cross-roads: which way to go? Cell Mol Life Sci 60(9):1852–1871. https://doi.org/10.1007/s00018-003-3096-6

    Article  CAS  PubMed  Google Scholar 

  34. Uversky VN (2013) A decade and a half of protein intrinsic disorder: biology still waits for physics. Protein Sci 22(6):693–724. https://doi.org/10.1002/pro.2261

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dyson HJ, Wright PE (2002) Coupling of folding and binding for unstructured proteins. Curr Opin Struct Biol 12(1):54–60. https://doi.org/10.1016/S0959-440X(02)00289-0

    Article  CAS  PubMed  Google Scholar 

  36. Dyson HJ, Wright PE (2005) Intrinsically unstructured proteins and their functions. Nat Rev Mol Cell Biol 6(3):197–208. https://doi.org/10.1038/nrm1589

    Article  CAS  PubMed  Google Scholar 

  37. Xie H, Vucetic S, Iakoucheva LM, Oldfield CJ, Dunker AK, Uversky VN, Obradovic Z (2007) Functional anthology of intrinsic disorder. 1. Biological processes and functions of proteins with long disordered regions. J Proteome Res 6(5):1882–1898. https://doi.org/10.1021/pr060392u

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dunker AK, Silman I, Uversky VN, Sussman JL (2008) Function and structure of inherently disordered proteins. Curr Opin Struct Biol 18(6):756–764. https://doi.org/10.1016/j.sbi.2008.10.002

    Article  CAS  PubMed  Google Scholar 

  39. Dunker AK, Uversky VN (2008) Signal transduction via unstructured protein conduits. Nat Chem Biol 4(4):229–230. https://doi.org/10.1038/nchembio0408-229

    Article  CAS  PubMed  Google Scholar 

  40. Oldfield CJ, Meng J, Yang JY, Yang MQ, Uversky VN, Dunker AK (2008) Flexible nets: disorder and induced fit in the associations of p53 and 14-3-3 with their partners. BMC Genom 9(Suppl 1):S1. https://doi.org/10.1186/1471-2164-9-S1-S1

    Article  CAS  Google Scholar 

  41. Dunker AK, Cortese MS, Romero P, Iakoucheva LM, Uversky VN (2005) Flexible nets. The roles of intrinsic disorder in protein interaction networks. FEBS J 272(20):5129–5148. https://doi.org/10.1111/j.1742-4658.2005.04948.x

    Article  CAS  PubMed  Google Scholar 

  42. Iakoucheva LM, Brown CJ, Lawson JD, Obradovic Z, Dunker AK (2002) Intrinsic disorder in cell-signaling and cancer-associated proteins. J Mol Biol 323(3):573–584. https://doi.org/10.1016/S0022-2836(02)00969-5

    Article  CAS  PubMed  Google Scholar 

  43. Uversky VN, Oldfield CJ, Dunker AK (2008) Intrinsically disordered proteins in human diseases: introducing the D2 concept. Annu Rev Biophys 37:215–246. https://doi.org/10.1146/annurev.biophys.37.032807.125924

    Article  CAS  PubMed  Google Scholar 

  44. Uversky VN, Dave V, Iakoucheva LM, Malaney P, Metallo SJ, Pathak RR, Joerger AC (2014) Pathological unfoldomics of uncontrolled chaos: intrinsically disordered proteins and human diseases. Chem Rev 114(13):6844–6879. https://doi.org/10.1021/cr400713r

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Dunker AK, Obradovic Z (2001) The protein trinity–linking function and disorder. Nat Biotechnol 19(9):805–806. https://doi.org/10.1038/nbt0901-805

    Article  CAS  PubMed  Google Scholar 

  46. Romero PR, Zaidi S, Fang YY, Uversky VN, Radivojac P, Oldfield CJ, Cortese MS, Sickmeier M, LeGall T, Obradovic Z, Dunker AK (2006) Alternative splicing in concert with protein intrinsic disorder enables increased functional diversity in multicellular organisms. Proc Natl Acad Sci USA 103(22):8390–8395. https://doi.org/10.1073/pnas.0507916103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jakob U, Kriwacki R, Uversky VN (2014) Conditionally and transiently disordered proteins: awakening cryptic disorder to regulate protein function. Chem Rev 114(13):6779–6805. https://doi.org/10.1021/cr400459c

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Uversky VN (2016) p53 proteoforms and intrinsic disorder: an illustration of the protein structure-function continuum concept. Int J Mol Sci 17(11):1874. https://doi.org/10.3390/ijms17111874

    Article  CAS  PubMed Central  Google Scholar 

  49. Schluter H, Apweiler R, Holzhutter HG, Jungblut PR (2009) Finding one’s way in proteomics: a protein species nomenclature. Chem Cent J 3:11. https://doi.org/10.1186/1752-153X-3-11

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Uhlen M, Bjorling E, Agaton C, Szigyarto CA, Amini B, Andersen E, Andersson AC, Angelidou P, Asplund A, Asplund C, Berglund L, Bergstrom K, Brumer H, Cerjan D, Ekstrom M, Elobeid A, Eriksson C, Fagerberg L, Falk R, Fall J, Forsberg M, Bjorklund MG, Gumbel K, Halimi A, Hallin I, Hamsten C, Hansson M, Hedhammar M, Hercules G, Kampf C, Larsson K, Linskog M, Lodewyckx W, Lund J, Lundeberg J, Magnusson K, Malm E, Nilsson P, Odling J, Oksvold P, Olsson I, Oster E, Ottosson J, Paavilainen L, Persson A, Rimini R, Rockberg J, Runeson M, Sivertsson A, Skollermo A, Steen J, Stenvall M, Sterky F, Stromberg S, Sundberg M, Tegel H, Tourle S, Wahlund E, Walden A, Wan JH, Wernerus H, Westberg J, Wester K, Wrethagen U, Xu LL, Hober S, Ponten F (2005) A human protein atlas for normal and cancer tissues based on antibody proteomics. Mol Cell Proteomics 4(12):1920–1932. https://doi.org/10.1074/mcp.M500279-MCP200

    Article  CAS  PubMed  Google Scholar 

  51. Farrah T, Deutsch EW, Omenn GS, Sun Z, Watts JD, Yamamoto T, Shteynberg D, Harris MM, Moritz RL (2014) State of the human proteome in 2013 as viewed through PeptideAtlas: comparing the kidney, urine, and plasma proteomes for the biology- and disease-driven human proteome project. J Proteome Res 13(1):60–75. https://doi.org/10.1021/Pr4010037

    Article  CAS  PubMed  Google Scholar 

  52. Farrah T, Deutsch EW, Hoopmann MR, Hallows JL, Sun Z, Huang CY, Moritz RL (2013) The State of the human proteome in 2012 as viewed through PeptideAtlas. J Proteome Res 12(1):162–171. https://doi.org/10.1021/Pr301012j

    Article  CAS  PubMed  Google Scholar 

  53. Reddy PJ, Ray S, Srivastava S (2015) The quest of the human proteome and the missing proteins: digging deeper. Omics-a J Integr Biol 19(5):276–282. https://doi.org/10.1089/omi.2015.0035

    Article  CAS  Google Scholar 

  54. Kim MS, Pinto SM, Getnet D, Nirujogi RS, Manda SS, Chaerkady R, Madugundu AK, Kelkar DS, Isserlin R, Jain S, Thomas JK, Muthusamy B, Leal-Rojas P, Kumar P, Sahasrabuddhe NA, Balakrishnan L, Advani J, George B, Renuse S, Selvan LDN, Patil AH, Nanjappa V, Radhakrishnan A, Prasad S, Subbannayya T, Raju R, Kumar M, Sreenivasamurthy SK, Marimuthu A, Sathe GJ, Chavan S, Datta KK, Subbannayya Y, Sahu A, Yelamanchi SD, Jayaram S, Rajagopalan P, Sharma J, Murthy KR, Syed N, Goel R, Khan AA, Ahmad S, Dey G, Mudgal K, Chatterjee A, Huang TC, Zhong J, Wu XY, Shaw PG, Freed D, Zahari MS, Mukherjee KK, Shankar S, Mahadevan A, Lam H, Mitchell CJ, Shankar SK, Satishchandra P, Schroeder JT, Sirdeshmukh R, Maitra A, Leach SD, Drake CG, Halushka MK, Prasad TSK, Hruban RH, Kerr CL, Bader GD, Iacobuzio-Donahue CA, Gowda H, Pandey A (2014) A draft map of the human proteome. Nature 509(7502):575. https://doi.org/10.1038/nature13302

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Consortium TEP (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489(7414):57–74. https://doi.org/10.1038/nature11247

    Article  CAS  Google Scholar 

  56. Ponomarenko EA, Poverennaya EV, Ilgisonis EV, Pyatnitskiy MA, Kopylov AT, Zgoda VG, Lisitsa AV, Archakov AI (2016) The size of the human proteome: the width and depth. Int J Anal Chem 2016:7436849. https://doi.org/10.1155/2016/7436849

    Article  PubMed  PubMed Central  Google Scholar 

  57. Smith LM, Kelleher NL, Consortium for Top Down P (2013) Proteoform: a single term describing protein complexity. Nat Methods 10(3):186–187. https://doi.org/10.1038/nmeth.2369

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Iakoucheva LM, Radivojac P, Brown CJ, O’Connor TR, Sikes JG, Obradovic Z, Dunker AK (2004) The importance of intrinsic disorder for protein phosphorylation. Nucleic Acids Res 32(3):1037–1049. https://doi.org/10.1093/nar/gkh253

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Pejaver V, Hsu WL, Xin F, Dunker AK, Uversky VN, Radivojac P (2014) The structural and functional signatures of proteins that undergo multiple events of post-translational modification. Protein Sci 23(8):1077–1093. https://doi.org/10.1002/pro.2494

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Oldfield CJ, Cheng Y, Cortese MS, Romero P, Uversky VN, Dunker AK (2005) Coupled folding and binding with alpha-helix-forming molecular recognition elements. Biochemistry 44(37):12454–12470. https://doi.org/10.1021/bi050736e

    Article  CAS  PubMed  Google Scholar 

  61. Radivojac P, Iakoucheva LM, Oldfield CJ, Obradovic Z, Uversky VN, Dunker AK (2007) Intrinsic disorder and functional proteomics. Biophys J 92(5):1439–1456. https://doi.org/10.1529/biophysj.106.094045

    Article  CAS  PubMed  Google Scholar 

  62. Uversky VN, Dunker AK (2010) Understanding protein non-folding. Biochim Biophys Acta 1804(6):1231–1264. https://doi.org/10.1016/j.bbapap.2010.01.017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Uversky VN (2011) Multitude of binding modes attainable by intrinsically disordered proteins: a portrait gallery of disorder-based complexes. Chem Soc Rev 40(3):1623–1634. https://doi.org/10.1039/c0cs00057d

    Article  CAS  PubMed  Google Scholar 

  64. Uversky VN (2012) Disordered competitive recruiter: fast and foldable. J Mol Biol 418(5):267–268. https://doi.org/10.1016/j.jmb.2012.02.034

    Article  CAS  PubMed  Google Scholar 

  65. Mohan A, Oldfield CJ, Radivojac P, Vacic V, Cortese MS, Dunker AK, Uversky VN (2006) Analysis of molecular recognition features (MoRFs). J Mol Biol 362(5):1043–1059. https://doi.org/10.1016/j.jmb.2006.07.087

    Article  CAS  PubMed  Google Scholar 

  66. Vacic V, Oldfield CJ, Mohan A, Radivojac P, Cortese MS, Uversky VN, Dunker AK (2007) Characterization of molecular recognition features, MoRFs, and their binding partners. J Proteome Res 6(6):2351–2366. https://doi.org/10.1021/pr0701411

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Beadle GW, Tatum EL (1941) Genetic control of biochemical reactions in neurospora. Proc Natl Acad Sci USA 27(11):499–506

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Uversky VN (2015) Functional roles of transiently and intrinsically disordered regions within proteins. FEBS J 282(7):1182–1189. https://doi.org/10.1111/febs.13202

    Article  CAS  PubMed  Google Scholar 

  69. Uversky VN (2018) Functions of short lifetime biological structures at large: the case of intrinsically disordered proteins. Brief Funct Genomics. https://doi.org/10.1093/bfgp/ely023

    Article  Google Scholar 

  70. Venkatakrishnan AJ, Flock T, Prado DE, Oates ME, Gough J, Madan Babu M (2014) Structured and disordered facets of the GPCR fold. Curr Opin Struct Biol 27:129–137. https://doi.org/10.1016/j.sbi.2014.08.002

    Article  CAS  PubMed  Google Scholar 

  71. Attwood TK, Findlay JB (1994) Fingerprinting G-protein-coupled receptors. Protein Eng 7(2):195–203. https://doi.org/10.1093/protein/7.2.195

    Article  CAS  PubMed  Google Scholar 

  72. Kolakowski LF Jr (1994) GCRDb: a G-protein-coupled receptor database. Recept Channels 2(1):1–7

    CAS  PubMed  Google Scholar 

  73. Wu H, Wang C, Gregory KJ, Han GW, Cho HP, Xia Y, Niswender CM, Katritch V, Meiler J, Cherezov V, Conn PJ, Stevens RC (2014) Structure of a class C GPCR metabotropic glutamate receptor 1 bound to an allosteric modulator. Science 344(6179):58–64. https://doi.org/10.1126/science.1249489

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Nomura R, Suzuki Y, Kakizuka A, Jingami H (2008) Direct detection of the interaction between recombinant soluble extracellular regions in the heterodimeric metabotropic gamma-aminobutyric acid receptor. J Biol Chem 283(8):4665–4673. https://doi.org/10.1074/jbc.M705202200

    Article  CAS  PubMed  Google Scholar 

  75. Geng Y, Bush M, Mosyak L, Wang F, Fan QR (2013) Structural mechanism of ligand activation in human GABA(B) receptor. Nature 504(7479):254–259. https://doi.org/10.1038/nature12725

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Burmakina S, Geng Y, Chen Y, Fan QR (2014) Heterodimeric coiled-coil interactions of human GABAB receptor. Proc Natl Acad Sci USA 111(19):6958–6963. https://doi.org/10.1073/pnas.1400081111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Margeta-Mitrovic M, Jan YN, Jan LY (2000) A trafficking checkpoint controls GABA(B) receptor heterodimerization. Neuron 27(1):97–106. https://doi.org/10.1016/S0896-6273(00)00012-X

    Article  CAS  PubMed  Google Scholar 

  78. Pagano A, Rovelli G, Mosbacher J, Lohmann T, Duthey B, Stauffer D, Ristig D, Schuler V, Meigel I, Lampert C, Stein T, Prezeau L, Blahos J, Pin J, Froestl W, Kuhn R, Heid J, Kaupmann K, Bettler B (2001) C-terminal interaction is essential for surface trafficking but not for heteromeric assembly of GABA(b) receptors. J Neurosci 21(4):1189–1202. https://doi.org/10.1523/JNEUROSCI.21-04-01189.2001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kaupmann K, Malitschek B, Schuler V, Heid J, Froestl W, Beck P, Mosbacher J, Bischoff S, Kulik A, Shigemoto R, Karschin A, Bettler B (1998) GABA(B)-receptor subtypes assemble into functional heteromeric complexes. Nature 396(6712):683–687. https://doi.org/10.1038/25360

    Article  CAS  PubMed  Google Scholar 

  80. White JH, Wise A, Main MJ, Green A, Fraser NJ, Disney GH, Barnes AA, Emson P, Foord SM, Marshall FH (1998) Heterodimerization is required for the formation of a functional GABA(B) receptor. Nature 396(6712):679–682. https://doi.org/10.1038/25354

    Article  CAS  PubMed  Google Scholar 

  81. Galvez T, Duthey B, Kniazeff J, Blahos J, Rovelli G, Bettler B, Prezeau L, Pin JP (2001) Allosteric interactions between GB1 and GB2 subunits are required for optimal GABA(B) receptor function. EMBO J 20(9):2152–2159. https://doi.org/10.1093/emboj/20.9.2152

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Geng Y, Mosyak L, Kurinov I, Zuo H, Sturchler E, Cheng TC, Subramanyam P, Brown AP, Brennan SC, Mun HC, Bush M, Chen Y, Nguyen TX, Cao B, Chang DD, Quick M, Conigrave AD, Colecraft HM, McDonald P, Fan QR (2016) Structural mechanism of ligand activation in human calcium-sensing receptor. Elife. https://doi.org/10.7554/elife.13662

    Article  PubMed  PubMed Central  Google Scholar 

  83. Riccardi D, Brennan SC, Chang W (2013) The extracellular calcium-sensing receptor, CaSR, in fetal development. Best Pract Res Clin Endocrinol Metab 27(3):443–453. https://doi.org/10.1016/j.beem.2013.02.010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Ruat M, Traiffort E (2013) Roles of the calcium sensing receptor in the central nervous system. Best Pract Res Clin Endocrinol Metab 27(3):429–442. https://doi.org/10.1016/j.beem.2013.03.001

    Article  CAS  PubMed  Google Scholar 

  85. Bandyopadhyay S, Tfelt-Hansen J, Chattopadhyay N (2010) Diverse roles of extracellular calcium-sensing receptor in the central nervous system. J Neurosci Res 88(10):2073–2082. https://doi.org/10.1002/jnr.22391

    Article  CAS  PubMed  Google Scholar 

  86. Conigrave AD, Hampson DR (2006) Broad-spectrum L-amino acid sensing by class 3 G-protein-coupled receptors. Trends Endocrinol Metab 17(10):398–407. https://doi.org/10.1016/j.tem.2006.10.012

    Article  CAS  PubMed  Google Scholar 

  87. Zhang D, Gao ZG, Zhang K, Kiselev E, Crane S, Wang J, Paoletta S, Yi C, Ma L, Zhang W, Han GW, Liu H, Cherezov V, Katritch V, Jiang H, Stevens RC, Jacobson KA, Zhao Q, Wu B (2015) Two disparate ligand-binding sites in the human P2Y1 receptor. Nature 520(7547):317–321. https://doi.org/10.1038/nature14287

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Park D, Ravichandran KS (2010) Emerging roles of brain-specific angiogenesis inhibitor 1. Adv Exp Med Biol 706:167–178. https://doi.org/10.1007/978-1-4419-7913-1_15

    Article  CAS  PubMed  Google Scholar 

  89. Das S, Sarkar A, Ryan KA, Fox S, Berger AH, Juncadella IJ, Bimczok D, Smythies LE, Harris PR, Ravichandran KS, Crowe SE, Smith PD, Ernst PB (2014) Brain angiogenesis inhibitor 1 is expressed by gastric phagocytes during infection with Helicobacter pylori and mediates the recognition and engulfment of human apoptotic gastric epithelial cells. FASEB J 28(5):2214–2224. https://doi.org/10.1096/fj.13-243238

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Duda DG, Sunamura M, Lozonschi L, Yokoyama T, Yatsuoka T, Motoi F, Horii A, Tani K, Asano S, Nakamura Y, Matsuno S (2002) Overexpression of the p53-inducible brain-specific angiogenesis inhibitor 1 suppresses efficiently tumour angiogenesis. Br J Cancer 86(3):490–496. https://doi.org/10.1038/sj.bjc.6600067

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Stephenson JR, Purcell RH, Hall RA (2014) The BAI subfamily of adhesion GPCRs: synaptic regulation and beyond. Trends Pharmacol Sci 35(4):208–215. https://doi.org/10.1016/j.tips.2014.02.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Kaur B, Brat DJ, Devi NS, Van Meir EG (2005) Vasculostatin, a proteolytic fragment of brain angiogenesis inhibitor 1, is an antiangiogenic and antitumorigenic factor. Oncogene 24(22):3632–3642. https://doi.org/10.1038/sj.onc.1208317

    Article  CAS  PubMed  Google Scholar 

  93. Cork SM, Kaur B, Devi NS, Cooper L, Saltz JH, Sandberg EM, Kaluz S, Van Meir EG (2012) A proprotein convertase/MMP-14 proteolytic cascade releases a novel 40 kDa vasculostatin from tumor suppressor BAI1. Oncogene 31(50):5144–5152. https://doi.org/10.1038/onc.2012.1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Cork SM, Van Meir EG (2011) Emerging roles for the BAI1 protein family in the regulation of phagocytosis, synaptogenesis, neurovasculature, and tumor development. J Mol Med (Berl) 89(8):743–752. https://doi.org/10.1007/s00109-011-0759-x

    Article  CAS  Google Scholar 

  95. Oda K, Shiratsuchi T, Nishimori H, Inazawa J, Yoshikawa H, Taketani Y, Nakamura Y, Tokino T (1999) Identification of BAIAP2 (BAI-associated protein 2), a novel human homologue of hamster IRSp53, whose SH3 domain interacts with the cytoplasmic domain of BAI1. Cytogenet Cell Genet 84(1–2):75–82. https://doi.org/10.1159/000015219

    Article  CAS  PubMed  Google Scholar 

  96. Shiratsuchi T, Futamura M, Oda K, Nishimori H, Nakamura Y, Tokino T (1998) Cloning and characterization of BAI-associated protein 1: a PDZ domain-containing protein that interacts with BAI1. Biochem Biophys Res Commun 247(3):597–604. https://doi.org/10.1006/bbrc.1998.8603

    Article  CAS  PubMed  Google Scholar 

  97. Lu YC, Nazarko OV, Sando R 3rd, Salzman GS, Li NS, Sudhof TC, Arac D (2015) Structural basis of latrophilin-FLRT-UNC5 interaction in cell adhesion. Structure 23(9):1678–1691. https://doi.org/10.1016/j.str.2015.06.024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Abbott RJ, Spendlove I, Roversi P, Fitzgibbon H, Knott V, Teriete P, McDonnell JM, Handford PA, Lea SM (2007) Structural and functional characterization of a novel T cell receptor co-regulatory protein complex, CD97-CD55. J Biol Chem 282(30):22023–22032. https://doi.org/10.1074/jbc.M702588200

    Article  CAS  PubMed  Google Scholar 

  99. Paavola KJ, Stephenson JR, Ritter SL, Alter SP, Hall RA (2011) The N terminus of the adhesion G protein-coupled receptor GPR56 controls receptor signaling activity. J Biol Chem 286(33):28914–28921. https://doi.org/10.1074/jbc.M111.247973

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Stoveken HM, Hajduczok AG, Xu L, Tall GG (2015) Adhesion G protein-coupled receptors are activated by exposure of a cryptic tethered agonist. Proc Natl Acad Sci USA 112(19):6194–6199. https://doi.org/10.1073/pnas.1421785112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Gazit A, Yaniv A, Bafico A, Pramila T, Igarashi M, Kitajewski J, Aaronson SA (1999) Human frizzled 1 interacts with transforming Wnts to transduce a TCF dependent transcriptional response. Oncogene 18(44):5959–5966. https://doi.org/10.1038/sj.onc.1202985

    Article  CAS  PubMed  Google Scholar 

  102. Van Camp JK, Beckers S, Zegers D, Van Hul W (2014) Wnt signaling and the control of human stem cell fate. Stem Cell Rev 10(2):207–229. https://doi.org/10.1007/s12015-013-9486-8

    Article  CAS  Google Scholar 

  103. Marchetti B, L’Episcopo F, Morale MC, Tirolo C, Testa N, Caniglia S, Serapide MF, Pluchino S (2013) Uncovering novel actors in astrocyte-neuron crosstalk in Parkinson’s disease: the Wnt/beta-catenin signaling cascade as the common final pathway for neuroprotection and self-repair. Eur J Neurosci 37(10):1550–1563. https://doi.org/10.1111/ejn.12166

    Article  PubMed  PubMed Central  Google Scholar 

  104. Planutis K, Planutiene M, Nguyen AV, Moyer MP, Holcombe RF (2013) Invasive colon cancer, but not non-invasive adenomas induce a gradient effect of Wnt pathway receptor frizzled 1 (Fz1) expression in the tumor microenvironment. J Transl Med 11:50. https://doi.org/10.1186/1479-5876-11-50

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Clevers H, Nusse R (2012) Wnt/beta-catenin signaling and disease. Cell 149(6):1192–1205. https://doi.org/10.1016/j.cell.2012.05.012

    Article  CAS  PubMed  Google Scholar 

  106. Gay A, Towler DA (2017) Wnt signaling in cardiovascular disease: opportunities and challenges. Curr Opin Lipidol 28(5):387–396. https://doi.org/10.1097/MOL.0000000000000445

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Nusse R, Clevers H (2017) Wnt/beta-catenin signaling, disease, and emerging therapeutic modalities. Cell 169(6):985–999. https://doi.org/10.1016/j.cell.2017.05.016

    Article  CAS  PubMed  Google Scholar 

  108. Masckauchan TN, Kitajewski J (2006) Wnt/Frizzled signaling in the vasculature: new angiogenic factors in sight. Physiology (Bethesda) 21:181–188. https://doi.org/10.1152/physiol.00058.2005

    Article  Google Scholar 

  109. Zhang B, Ma JX (2010) Wnt pathway antagonists and angiogenesis. Protein Cell 1(10):898–906. https://doi.org/10.1007/s13238-010-0112-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Clevers H (2006) Wnt/beta-catenin signaling in development and disease. Cell 127(3):469–480. https://doi.org/10.1016/j.cell.2006.10.018

    Article  CAS  PubMed  Google Scholar 

  111. Gorojankina T (2016) Hedgehog signaling pathway: a novel model and molecular mechanisms of signal transduction. Cell Mol Life Sci 73(7):1317–1332. https://doi.org/10.1007/s00018-015-2127-4

    Article  CAS  PubMed  Google Scholar 

  112. Gong X, Qian H, Cao P, Zhao X, Zhou Q, Lei J, Yan N (2018) Structural basis for the recognition of Sonic Hedgehog by human Patched1. Science. https://doi.org/10.1126/science.aas8935

    Article  PubMed  Google Scholar 

  113. Ingham PW, Nakano Y, Seger C (2011) Mechanisms and functions of Hedgehog signalling across the metazoa. Nat Rev Genet 12(6):393–406. https://doi.org/10.1038/nrg2984

    Article  CAS  PubMed  Google Scholar 

  114. Briscoe J, Therond PP (2013) The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol 14(7):416–429. https://doi.org/10.1038/nrm3598

    Article  CAS  PubMed  Google Scholar 

  115. McMahon AP, Ingham PW, Tabin CJ (2003) Developmental roles and clinical significance of hedgehog signaling. Curr Top Dev Biol 53:1–114. https://doi.org/10.1016/S0070-2153(03)53002-2

    Article  CAS  PubMed  Google Scholar 

  116. Taipale J, Beachy PA (2001) The Hedgehog and Wnt signalling pathways in cancer. Nature 411(6835):349–354. https://doi.org/10.1038/35077219

    Article  CAS  PubMed  Google Scholar 

  117. Zardawi SJ, O’Toole SA, Sutherland RL, Musgrove EA (2009) Dysregulation of Hedgehog, Wnt and Notch signalling pathways in breast cancer. Histol Histopathol 24(3):385–398. https://doi.org/10.14670/HH-24.385

    Article  CAS  PubMed  Google Scholar 

  118. Toftgard R (2000) Hedgehog signalling in cancer. Cell Mol Life Sci 57(12):1720–1731. https://doi.org/10.1007/PL00000654

    Article  CAS  PubMed  Google Scholar 

  119. Sharpe HJ, Wang W, Hannoush RN, de Sauvage FJ (2015) Regulation of the oncoprotein Smoothened by small molecules. Nat Chem Biol 11(4):246–255. https://doi.org/10.1038/nchembio.1776

    Article  CAS  PubMed  Google Scholar 

  120. Wang C, Wu H, Evron T, Vardy E, Han GW, Huang XP, Hufeisen SJ, Mangano TJ, Urban DJ, Katritch V, Cherezov V, Caron MG, Roth BL, Stevens RC (2014) Structural basis for Smoothened receptor modulation and chemoresistance to anticancer drugs. Nat Commun 5:4355. https://doi.org/10.1038/ncomms5355

    Article  CAS  PubMed  Google Scholar 

  121. Byrne EFX, Sircar R, Miller PS, Hedger G, Luchetti G, Nachtergaele S, Tully MD, Mydock-McGrane L, Covey DF, Rambo RP, Sansom MSP, Newstead S, Rohatgi R, Siebold C (2016) Structural basis of Smoothened regulation by its extracellular domains. Nature 535(7613):517–522. https://doi.org/10.1038/nature18934

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Liang YL, Khoshouei M, Radjainia M, Zhang Y, Glukhova A, Tarrasch J, Thal DM, Furness SGB, Christopoulos G, Coudrat T, Danev R, Baumeister W, Miller LJ, Christopoulos A, Kobilka BK, Wootten D, Skiniotis G, Sexton PM (2017) Phase-plate cryo-EM structure of a class B GPCR–G-protein complex. Nature 546(7656):118–123. https://doi.org/10.1038/nature22327

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Kusano S, Kukimoto-Niino M, Hino N, Ohsawa N, Okuda K, Sakamoto K, Shirouzu M, Shindo T, Yokoyama S (2012) Structural basis for extracellular interactions between calcitonin receptor-like receptor and receptor activity-modifying protein 2 for adrenomedullin-specific binding. Protein Sci 21(2):199–210. https://doi.org/10.1002/pro.2003

    Article  CAS  PubMed  Google Scholar 

  124. Liang YL, Khoshouei M, Deganutti G, Glukhova A, Koole C, Peat TS, Radjainia M, Plitzko JM, Baumeister W, Miller LJ, Hay DL, Christopoulos A, Reynolds CA, Wootten D, Sexton PM (2018) Cryo-EM structure of the active, Gs-protein complexed, human CGRP receptor. Nature 561(7724):492–497. https://doi.org/10.1038/s41586-018-0535-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Tao J, Hildebrand ME, Liao P, Liang MC, Tan G, Li S, Snutch TP, Soong TW (2008) Activation of corticotropin-releasing factor receptor 1 selectively inhibits CaV3.2 T-type calcium channels. Mol Pharmacol 73(6):1596–1609. https://doi.org/10.1124/mol.107.043612

    Article  CAS  PubMed  Google Scholar 

  126. Grace CR, Perrin MH, Gulyas J, Rivier JE, Vale WW, Riek R (2010) NMR structure of the first extracellular domain of corticotropin-releasing factor receptor 1 (ECD1-CRF-R1) complexed with a high affinity agonist. J Biol Chem 285(49):38580–38589. https://doi.org/10.1074/jbc.M110.121897

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Gulyas J, Rivier C, Perrin M, Koerber SC, Sutton S, Corrigan A, Lahrichi SL, Craig AG, Vale W, Rivier J (1995) Potent, structurally constrained agonists and competitive antagonists of corticotropin-releasing factor. Proc Natl Acad Sci USA 92(23):10575–10579

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Gilman AG (1987) G proteins: transducers of receptor-generated signals. Annu Rev Biochem 56:615–649. https://doi.org/10.1146/annurev.bi.56.070187.003151

    Article  CAS  PubMed  Google Scholar 

  129. Engelhardt S, Rochais F (2007) G proteins: more than transducers of receptor-generated signals? Circ Res 100(8):1109–1111. https://doi.org/10.1161/01.RES.0000266971.15127.e8

    Article  CAS  PubMed  Google Scholar 

  130. Simon MI, Strathmann MP, Gautam N (1991) Diversity of G proteins in signal transduction. Science 252(5007):802–808. https://doi.org/10.1126/science.1902986

    Article  CAS  PubMed  Google Scholar 

  131. Syrovatkina V, Alegre KO, Dey R, Huang XY (2016) Regulation, signaling, and physiological functions of G-proteins. J Mol Biol 428(19):3850–3868. https://doi.org/10.1016/j.jmb.2016.08.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Bourne HR, Sanders DA, McCormick F (1990) The GTPase superfamily: a conserved switch for diverse cell functions. Nature 348(6297):125–132. https://doi.org/10.1038/348125a0

    Article  CAS  PubMed  Google Scholar 

  133. Brand CS, Sadana R, Malik S, Smrcka AV, Dessauer CW (2015) Adenylyl cyclase 5 regulation by gbetagamma involves isoform-specific use of multiple interaction sites. Mol Pharmacol 88(4):758–767. https://doi.org/10.1124/mol.115.099556

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Farfel Z, Iiri T, Shapira H, Roitman A, Mouallem M, Bourne HR (1996) Pseudohypoparathyroidism, a novel mutation in the betagamma-contact region of Gsalpha impairs receptor stimulation. J Biol Chem 271(33):19653–19655. https://doi.org/10.1074/jbc.271.33.19653

    Article  CAS  PubMed  Google Scholar 

  135. Plagge A, Isles AR, Gordon E, Humby T, Dean W, Gritsch S, Fischer-Colbrie R, Wilkinson LS, Kelsey G (2005) Imprinted Nesp55 influences behavioral reactivity to novel environments. Mol Cell Biol 25(8):3019–3026. https://doi.org/10.1128/MCB.25.8.3019-3026.2005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Hu Q, Shokat KM (2018) Disease-causing mutations in the G protein Galphas subvert the roles of GDP and GTP. Cell 173(5):1254-1264 e1211. https://doi.org/10.1016/j.cell.2018.03.018

    Article  CAS  Google Scholar 

  137. Goricanec D, Stehle R, Egloff P, Grigoriu S, Pluckthun A, Wagner G, Hagn F (2016) Conformational dynamics of a G-protein alpha subunit is tightly regulated by nucleotide binding. Proc Natl Acad Sci USA 113(26):E3629–E3638. https://doi.org/10.1073/pnas.1604125113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Oldham WM, Van Eps N, Preininger AM, Hubbell WL, Hamm HE (2007) Mapping allosteric connections from the receptor to the nucleotide-binding pocket of heterotrimeric G proteins. Proc Natl Acad Sci USA 104(19):7927–7932. https://doi.org/10.1073/pnas.0702623104

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Mixon MB, Lee E, Coleman DE, Berghuis AM, Gilman AG, Sprang SR (1995) Tertiary and quaternary structural changes in Gi alpha 1 induced by GTP hydrolysis. Science 270(5238):954–960. https://doi.org/10.1126/science.270.5238.954

    Article  CAS  PubMed  Google Scholar 

  140. Coleman DE, Berghuis AM, Lee E, Linder ME, Gilman AG, Sprang SR (1994) Structures of active conformations of Gi alpha 1 and the mechanism of GTP hydrolysis. Science 265(5177):1405–1412. https://doi.org/10.1126/science.8073283

    Article  CAS  PubMed  Google Scholar 

  141. Chen CK, Chan NL, Wang AH (2011) The many blades of the beta-propeller proteins: conserved but versatile. Trends Biochem Sci 36(10):553–561. https://doi.org/10.1016/j.tibs.2011.07.004

    Article  CAS  PubMed  Google Scholar 

  142. Anurag M, Singh GP, Dash D (2012) Location of disorder in coiled coil proteins is influenced by its biological role and subcellular localization: a GO-based study on human proteome. Mol BioSyst 8(1):346–352. https://doi.org/10.1039/c1mb05210a

    Article  CAS  PubMed  Google Scholar 

  143. Gazi AD, Bastaki M, Charova SN, Gkougkoulia EA, Kapellios EA, Panopoulos NJ, Kokkinidis M (2008) Evidence for a coiled-coil interaction mode of disordered proteins from bacterial type III secretion systems. J Biol Chem 283(49):34062–34068. https://doi.org/10.1074/jbc.M803408200

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Gunasekaran K, Tsai CJ, Nussinov R (2004) Analysis of ordered and disordered protein complexes reveals structural features discriminating between stable and unstable monomers. J Mol Biol 341(5):1327–1341. https://doi.org/10.1016/j.jmb.2004.07.002

    Article  CAS  PubMed  Google Scholar 

  145. Wu Z, Hu G, Yang J, Peng Z, Uversky VN, Kurgan L (2015) In various protein complexes, disordered protomers have large per-residue surface areas and area of protein-, DNA- and RNA-binding interfaces. FEBS Lett 589(19 Pt A):2561–2569. https://doi.org/10.1016/j.febslet.2015.08.014

    Article  CAS  PubMed  Google Scholar 

  146. Bock A, Kostenis E, Trankle C, Lohse MJ, Mohr K (2014) Pilot the pulse: controlling the multiplicity of receptor dynamics. Trends Pharmacol Sci 35(12):630–638. https://doi.org/10.1016/j.tips.2014.10.002

    Article  CAS  PubMed  Google Scholar 

  147. Van Eps N, Caro LN, Morizumi T, Kusnetzow AK, Szczepek M, Hofmann KP, Bayburt TH, Sligar SG, Ernst OP, Hubbell WL (2017) Conformational equilibria of light-activated rhodopsin in nanodiscs. Proc Natl Acad Sci USA 114(16):E3268–E3275. https://doi.org/10.1073/pnas.1620405114

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Manglik A, Kim TH, Masureel M, Altenbach C, Yang Z, Hilger D, Lerch MT, Kobilka TS, Thian FS, Hubbell WL, Prosser RS, Kobilka BK (2015) Structural insights into the dynamic process of beta2-adrenergic receptor signaling. Cell 161(5):1101–1111. https://doi.org/10.1016/j.cell.2015.04.043

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Nygaard R, Zou Y, Dror RO, Mildorf TJ, Arlow DH, Manglik A, Pan AC, Liu CW, Fung JJ, Bokoch MP, Thian FS, Kobilka TS, Shaw DE, Mueller L, Prosser RS, Kobilka BK (2013) The dynamic process of beta(2)-adrenergic receptor activation. Cell 152(3):532–542. https://doi.org/10.1016/j.cell.2013.01.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Kofuku Y, Ueda T, Okude J, Shiraishi Y, Kondo K, Maeda M, Tsujishita H, Shimada I (2012) Efficacy of the beta(2)-adrenergic receptor is determined by conformational equilibrium in the transmembrane region. Nat Commun 3:1045. https://doi.org/10.1038/ncomms2046

    Article  CAS  PubMed  Google Scholar 

  151. Violin JD, Crombie AL, Soergel DG, Lark MW (2014) Biased ligands at G-protein-coupled receptors: promise and progress. Trends Pharmacol Sci 35(7):308–316. https://doi.org/10.1016/j.tips.2014.04.007

    Article  CAS  PubMed  Google Scholar 

  152. Rajagopal S, Rajagopal K, Lefkowitz RJ (2010) Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nat Rev Drug Discov 9(5):373–386. https://doi.org/10.1038/nrd3024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Strachan RT, Sun JP, Rominger DH, Violin JD, Ahn S, Rojas Bie Thomsen A, Zhu X, Kleist A, Costa T, Lefkowitz RJ (2014) Divergent transducer-specific molecular efficacies generate biased agonism at a G protein-coupled receptor (GPCR). J Biol Chem 289(20):14211–14224. https://doi.org/10.1074/jbc.M114.548131

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Wacker D, Wang C, Katritch V, Han GW, Huang XP, Vardy E, McCorvy JD, Jiang Y, Chu M, Siu FY, Liu W, Xu HE, Cherezov V, Roth BL, Stevens RC (2013) Structural features for functional selectivity at serotonin receptors. Science 340(6132):615–619. https://doi.org/10.1126/science.1232808

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Wootten D, Simms J, Miller LJ, Christopoulos A, Sexton PM (2013) Polar transmembrane interactions drive formation of ligand-specific and signal pathway-biased family B G protein-coupled receptor conformations. Proc Natl Acad Sci USA 110(13):5211–5216. https://doi.org/10.1073/pnas.1221585110

    Article  PubMed  PubMed Central  Google Scholar 

  156. Katritch V, Cherezov V, Stevens RC (2013) Structure-function of the G protein-coupled receptor superfamily. Annu Rev Pharmacol Toxicol 53:531–556. https://doi.org/10.1146/annurev-pharmtox-032112-135923

    Article  CAS  PubMed  Google Scholar 

  157. Mary S, Damian M, Louet M, Floquet N, Fehrentz JA, Marie J, Martinez J, Baneres JL (2012) Ligands and signaling proteins govern the conformational landscape explored by a G protein-coupled receptor. Proc Natl Acad Sci USA 109(21):8304–8309. https://doi.org/10.1073/pnas.1119881109

    Article  PubMed  PubMed Central  Google Scholar 

  158. Rahmeh R, Damian M, Cottet M, Orcel H, Mendre C, Durroux T, Sharma KS, Durand G, Pucci B, Trinquet E, Zwier JM, Deupi X, Bron P, Baneres JL, Mouillac B, Granier S (2012) Structural insights into biased G protein-coupled receptor signaling revealed by fluorescence spectroscopy. Proc Natl Acad Sci USA 109(17):6733–6738. https://doi.org/10.1073/pnas.1201093109

    Article  PubMed  PubMed Central  Google Scholar 

  159. Liu JJ, Horst R, Katritch V, Stevens RC, Wuthrich K (2012) Biased signaling pathways in beta2-adrenergic receptor characterized by 19F-NMR. Science 335(6072):1106–1110. https://doi.org/10.1126/science.1215802

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Reiter E, Ahn S, Shukla AK, Lefkowitz RJ (2012) Molecular mechanism of beta-arrestin-biased agonism at seven-transmembrane receptors. Annu Rev Pharmacol Toxicol 52:179–197. https://doi.org/10.1146/annurev.pharmtox.010909.105800

    Article  CAS  PubMed  Google Scholar 

  161. Kahsai AW, Xiao K, Rajagopal S, Ahn S, Shukla AK, Sun J, Oas TG, Lefkowitz RJ (2011) Multiple ligand-specific conformations of the beta2-adrenergic receptor. Nat Chem Biol 7(10):692–700. https://doi.org/10.1038/nchembio.634

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Rajagopal S, Ahn S, Rominger DH, Gowen-MacDonald W, Lam CM, Dewire SM, Violin JD, Lefkowitz RJ (2011) Quantifying ligand bias at seven-transmembrane receptors. Mol Pharmacol 80(3):367–377. https://doi.org/10.1124/mol.111.072801

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Kenakin T (2011) Functional selectivity and biased receptor signaling. J Pharmacol Exp Ther 336(2):296–302. https://doi.org/10.1124/jpet.110.173948

    Article  CAS  PubMed  Google Scholar 

  164. Kobilka BK, Deupi X (2007) Conformational complexity of G-protein-coupled receptors. Trends Pharmacol Sci 28(8):397–406. https://doi.org/10.1016/j.tips.2007.06.003

    Article  CAS  PubMed  Google Scholar 

  165. Yao X, Parnot C, Deupi X, Ratnala VR, Swaminath G, Farrens D, Kobilka B (2006) Coupling ligand structure to specific conformational switches in the beta2-adrenoceptor. Nat Chem Biol 2(8):417–422. https://doi.org/10.1038/nchembio801

    Article  CAS  PubMed  Google Scholar 

  166. Pierce KL, Premont RT, Lefkowitz RJ (2002) Seven-transmembrane receptors. Nat Rev Mol Cell Biol 3(9):639–650. https://doi.org/10.1038/nrm908

    Article  CAS  PubMed  Google Scholar 

  167. Luttrell LM, Maudsley S, Gesty-Palmer D (2018) Translating in vitro ligand bias into in vivo efficacy. Cell Signal 41:46–55. https://doi.org/10.1016/j.cellsig.2017.05.002

    Article  CAS  PubMed  Google Scholar 

  168. Maudsley S, Martin B, Luttrell LM (2005) The origins of diversity and specificity in g protein-coupled receptor signaling. J Pharmacol Exp Ther 314(2):485–494. https://doi.org/10.1124/jpet.105.083121

    Article  CAS  PubMed  Google Scholar 

  169. McLatchie LM, Fraser NJ, Main MJ, Wise A, Brown J, Thompson N, Solari R, Lee MG, Foord SM (1998) RAMPs regulate the transport and ligand specificity of the calcitonin-receptor-like receptor. Nature 393(6683):333–339. https://doi.org/10.1038/30666

    Article  CAS  PubMed  Google Scholar 

  170. Evans BN, Rosenblatt MI, Mnayer LO, Oliver KR, Dickerson IM (2000) CGRP-RCP, a novel protein required for signal transduction at calcitonin gene-related peptide and adrenomedullin receptors. J Biol Chem 275(40):31438–31443. https://doi.org/10.1074/jbc.M005604200

    Article  CAS  PubMed  Google Scholar 

  171. Luttrell LM, Ferguson SS, Daaka Y, Miller WE, Maudsley S, Della Rocca GJ, Lin F, Kawakatsu H, Owada K, Luttrell DK, Caron MG, Lefkowitz RJ (1999) Beta-arrestin-dependent formation of beta2 adrenergic receptor-Src protein kinase complexes. Science 283(5402):655–661

    Article  CAS  PubMed  Google Scholar 

  172. Maudsley S, Martin B, Janssens J, Etienne H, Jushaj A, van Gastel J, Willemsen A, Chen H, Gesty-Palmer D, Luttrell LM (2016) Informatic deconvolution of biased GPCR signaling mechanisms from in vivo pharmacological experimentation. Methods 92:51–63. https://doi.org/10.1016/j.ymeth.2015.05.013

    Article  CAS  PubMed  Google Scholar 

  173. Maudsley S, Martin B, Gesty-Palmer D, Cheung H, Johnson C, Patel S, Becker KG, Wood WH 3rd, Zhang Y, Lehrmann E, Luttrell LM (2015) Delineation of a conserved arrestin-biased signaling repertoire in vivo. Mol Pharmacol 87(4):706–717. https://doi.org/10.1124/mol.114.095224

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Leysen H, van Gastel J, Hendrickx JO, Santos-Otte P, Martin B, Maudsley S (2018) G protein-coupled receptor systems as crucial regulators of DNA damage response processes. Int J Mol Sci. https://doi.org/10.3390/ijms19102919

    Article  PubMed  PubMed Central  Google Scholar 

  175. Ritter SL, Hall RA (2009) Fine-tuning of GPCR activity by receptor-interacting proteins. Nat Rev Mol Cell Biol 10(12):819–830. https://doi.org/10.1038/nrm2803

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Pak Y, Pham N, Rotin D (2002) Direct binding of the beta1 adrenergic receptor to the cyclic AMP-dependent guanine nucleotide exchange factor CNrasGEF leads to Ras activation. Mol Cell Biol 22(22):7942–7952

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Hall RA, Premont RT, Chow CW, Blitzer JT, Pitcher JA, Claing A, Stoffel RH, Barak LS, Shenolikar S, Weinman EJ, Grinstein S, Lefkowitz RJ (1998) The beta2-adrenergic receptor interacts with the Na+/H+-exchanger regulatory factor to control Na+/H+ exchange. Nature 392(6676):626–630. https://doi.org/10.1038/33458

    Article  CAS  PubMed  Google Scholar 

  178. Mahon MJ, Donowitz M, Yun CC, Segre GV (2002) Na(+)/H(+) exchanger regulatory factor 2 directs parathyroid hormone 1 receptor signalling. Nature 417(6891):858–861. https://doi.org/10.1038/nature00816

    Article  CAS  PubMed  Google Scholar 

  179. Balasubramanian S, Fam SR, Hall RA (2007) GABAB receptor association with the PDZ scaffold Mupp1 alters receptor stability and function. J Biol Chem 282(6):4162–4171. https://doi.org/10.1074/jbc.M607695200

    Article  CAS  PubMed  Google Scholar 

  180. Guillaume JL, Daulat AM, Maurice P, Levoye A, Migaud M, Brydon L, Malpaux B, Borg-Capra C, Jockers R (2008) The PDZ protein mupp1 promotes Gi coupling and signaling of the Mt1 melatonin receptor. J Biol Chem 283(24):16762–16771. https://doi.org/10.1074/jbc.M802069200

    Article  CAS  PubMed  Google Scholar 

  181. Yao R, Natsume Y, Noda T (2004) MAGI-3 is involved in the regulation of the JNK signaling pathway as a scaffold protein for frizzled and Ltap. Oncogene 23(36):6023–6030. https://doi.org/10.1038/sj.onc.1207817

    Article  CAS  PubMed  Google Scholar 

  182. Zhang H, Wang D, Sun H, Hall RA, Yun CC (2007) MAGI-3 regulates LPA-induced activation of Erk and RhoA. Cell Signal 19(2):261–268. https://doi.org/10.1016/j.cellsig.2006.06.008

    Article  CAS  PubMed  Google Scholar 

  183. Yamada T, Ohoka Y, Kogo M, Inagaki S (2005) Physical and functional interactions of the lysophosphatidic acid receptors with PDZ domain-containing Rho guanine nucleotide exchange factors (RhoGEFs). J Biol Chem 280(19):19358–19363. https://doi.org/10.1074/jbc.M414561200

    Article  CAS  PubMed  Google Scholar 

  184. Yun HM, Kim S, Kim HJ, Kostenis E, Kim JI, Seong JY, Baik JH, Rhim H (2007) The novel cellular mechanism of human 5-HT6 receptor through an interaction with Fyn. J Biol Chem 282(8):5496–5505. https://doi.org/10.1074/jbc.M606215200

    Article  CAS  PubMed  Google Scholar 

  185. Hall RA, Lefkowitz RJ (2002) Regulation of G protein-coupled receptor signaling by scaffold proteins. Circ Res 91(8):672–680

    Article  CAS  PubMed  Google Scholar 

  186. Marrero MB, Venema VJ, Ju H, Eaton DC, Venema RC (1998) Regulation of angiotensin II-induced JAK2 tyrosine phosphorylation: roles of SHP-1 and SHP-2. Am J Physiol 275(5 Pt 1):C1216–C1223

    Article  CAS  PubMed  Google Scholar 

  187. Godeny MD, Sayyah J, VonDerLinden D, Johns M, Ostrov DA, Caldwell-Busby J, Sayeski PP (2007) The N-terminal SH2 domain of the tyrosine phosphatase, SHP-2, is essential for Jak2-dependent signaling via the angiotensin II type AT1 receptor. Cell Signal 19(3):600–609. https://doi.org/10.1016/j.cellsig.2006.08.010

    Article  CAS  PubMed  Google Scholar 

  188. Lukashova V, Asselin C, Krolewski JJ, Rola-Pleszczynski M, Stankova J (2001) G-protein-independent activation of Tyk2 by the platelet-activating factor receptor. J Biol Chem 276(26):24113–24121. https://doi.org/10.1074/jbc.M100720200

    Article  CAS  PubMed  Google Scholar 

  189. Lukashova V, Chen Z, Duhe RJ, Rola-Pleszczynski M, Stankova J (2003) Janus kinase 2 activation by the platelet-activating factor receptor (PAFR): roles of Tyk2 and PAFR C terminus. J Immunol 171(7):3794–3800

    Article  CAS  PubMed  Google Scholar 

  190. Shih M, Lin F, Scott JD, Wang HY, Malbon CC (1999) Dynamic complexes of beta2-adrenergic receptors with protein kinases and phosphatases and the role of gravin. J Biol Chem 274(3):1588–1595

    Article  CAS  PubMed  Google Scholar 

  191. Fraser ID, Cong M, Kim J, Rollins EN, Daaka Y, Lefkowitz RJ, Scott JD (2000) Assembly of an A kinase-anchoring protein-beta(2)-adrenergic receptor complex facilitates receptor phosphorylation and signaling. Curr Biol 10(7):409–412

    Article  CAS  PubMed  Google Scholar 

  192. Fan G, Shumay E, Wang H, Malbon CC (2001) The scaffold protein gravin (cAMP-dependent protein kinase-anchoring protein 250) binds the beta 2-adrenergic receptor via the receptor cytoplasmic Arg-329 to Leu-413 domain and provides a mobile scaffold during desensitization. J Biol Chem 276(26):24005–24014. https://doi.org/10.1074/jbc.M011199200

    Article  CAS  PubMed  Google Scholar 

  193. Tao J, Wang HY, Malbon CC (2003) Protein kinase A regulates AKAP250 (gravin) scaffold binding to the beta2-adrenergic receptor. EMBO J 22(24):6419–6429. https://doi.org/10.1093/emboj/cdg628

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Gardner LA, Tavalin SJ, Goehring AS, Scott JD, Bahouth SW (2006) AKAP79-mediated targeting of the cyclic AMP-dependent protein kinase to the beta1-adrenergic receptor promotes recycling and functional resensitization of the receptor. J Biol Chem 281(44):33537–33553. https://doi.org/10.1074/jbc.M601809200

    Article  CAS  PubMed  Google Scholar 

  195. Brakeman PR, Lanahan AA, O’Brien R, Roche K, Barnes CA, Huganir RL, Worley PF (1997) Homer: a protein that selectively binds metabotropic glutamate receptors. Nature 386(6622):284–288. https://doi.org/10.1038/386284a0

    Article  CAS  PubMed  Google Scholar 

  196. Xiao B, Tu JC, Petralia RS, Yuan JP, Doan A, Breder CD, Ruggiero A, Lanahan AA, Wenthold RJ, Worley PF (1998) Homer regulates the association of group 1 metabotropic glutamate receptors with multivalent complexes of homer-related, synaptic proteins. Neuron 21(4):707–716

    Article  CAS  PubMed  Google Scholar 

  197. Tu JC, Xiao B, Yuan JP, Lanahan AA, Leoffert K, Li M, Linden DJ, Worley PF (1998) Homer binds a novel proline-rich motif and links group 1 metabotropic glutamate receptors with IP3 receptors. Neuron 21(4):717–726

    Article  CAS  PubMed  Google Scholar 

  198. Kato A, Ozawa F, Saitoh Y, Fukazawa Y, Sugiyama H, Inokuchi K (1998) Novel members of the Vesl/Homer family of PDZ proteins that bind metabotropic glutamate receptors. J Biol Chem 273(37):23969–23975

    Article  CAS  PubMed  Google Scholar 

  199. Kammermeier PJ, Worley PF (2007) Homer 1a uncouples metabotropic glutamate receptor 5 from postsynaptic effectors. Proc Natl Acad Sci USA 104(14):6055–6060. https://doi.org/10.1073/pnas.0608991104

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Kammermeier PJ (2008) Endogenous homer proteins regulate metabotropic glutamate receptor signaling in neurons. J Neurosci 28(34):8560–8567. https://doi.org/10.1523/JNEUROSCI.1830-08.2008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Sokolina K, Kittanakom S, Snider J, Kotlyar M, Maurice P, Gandia J, Benleulmi-Chaachoua A, Tadagaki K, Oishi A, Wong V, Malty RH, Deineko V, Aoki H, Amin S, Yao Z, Morato X, Otasek D, Kobayashi H, Menendez J, Auerbach D, Angers S, Przulj N, Bouvier M, Babu M, Ciruela F, Jockers R, Jurisica I, Stagljar I (2017) Systematic protein-protein interaction mapping for clinically relevant human GPCRs. Mol Syst Biol 13(3):918. https://doi.org/10.15252/msb.20167430

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Benleulmi-Chaachoua A, Chen L, Sokolina K, Wong V, Jurisica I, Emerit MB, Darmon M, Espin A, Stagljar I, Tafelmeyer P, Zamponi GW, Delagrange P, Maurice P, Jockers R (2016) Protein interactome mining defines melatonin MT1 receptors as integral component of presynaptic protein complexes of neurons. J Pineal Res 60(1):95–108. https://doi.org/10.1111/jpi.12294

    Article  CAS  PubMed  Google Scholar 

  203. Gutierrez-Rodriguez M, Herranz R (2015) From multiple PAR1 receptor/protein interactions to their multiple therapeutic implications. Curr Top Med Chem 15(20):2080–2114

    Article  CAS  PubMed  Google Scholar 

  204. Dunn HA, Patil DN, Cao Y, Orlandi C, Martemyanov KA (2018) Synaptic adhesion protein ELFN1 is a selective allosteric modulator of group III metabotropic glutamate receptors in trans. Proc Natl Acad Sci USA 115(19):5022–5027. https://doi.org/10.1073/pnas.1722498115

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Iyer K, Burkle L, Auerbach D, Thaminy S, Dinkel M, Engels K (2005) Stagljar I (2005) Utilizing the split-ubiquitin membrane yeast two-hybrid system to identify protein–protein interactions of integral membrane proteins. Sci STKE 275:pl3. https://doi.org/10.1126/stke.2752005pl3

    Article  Google Scholar 

  206. Snider J, Kittanakom S, Damjanovic D, Curak J, Wong V, Stagljar I (2010) Detecting interactions with membrane proteins using a membrane two-hybrid assay in yeast. Nat Protoc 5(7):1281–1293. https://doi.org/10.1038/nprot.2010.83

    Article  CAS  PubMed  Google Scholar 

  207. Asseck LY, Grefen C (2018) Detecting interactions of membrane proteins: the split-ubiquitin system. Methods Mol Biol 1794:49–60. https://doi.org/10.1007/978-1-4939-7871-7_4

    Article  CAS  PubMed  Google Scholar 

  208. Petschnigg J, Wong V, Snider J, Stagljar I (2012) Investigation of membrane protein interactions using the split-ubiquitin membrane yeast two-hybrid system. Methods Mol Biol 812:225–244. https://doi.org/10.1007/978-1-61779-455-1_13

    Article  CAS  PubMed  Google Scholar 

  209. Thaminy S, Miller J, Stagljar I (2004) The split-ubiquitin membrane-based yeast two-hybrid system. Methods Mol Biol 261:297–312. https://doi.org/10.1385/1-59259-762-9:297

    Article  CAS  PubMed  Google Scholar 

  210. Stagljar I, Korostensky C, Johnsson N, te Heesen S (1998) A genetic system based on split-ubiquitin for the analysis of interactions between membrane proteins in vivo. Proc Natl Acad Sci USA 95(9):5187–5192

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Downes GB, Gautam N (1999) The G protein subunit gene families. Genomics 62(3):544–552. https://doi.org/10.1006/geno.1999.5992

    Article  CAS  PubMed  Google Scholar 

  212. Temple BR, Jones CD, Jones AM (2010) Evolution of a signaling nexus constrained by protein interfaces and conformational states. PLoS Comput Biol 6(10):e1000962. https://doi.org/10.1371/journal.pcbi.1000962

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Jones AM, Assmann SM (2004) Plants: the latest model system for G-protein research. EMBO Rep 5(6):572–578. https://doi.org/10.1038/sj.embor.7400174

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Pandey S, Chen JG, Jones AM, Assmann SM (2006) G-protein complex mutants are hypersensitive to abscisic acid regulation of germination and postgermination development. Plant Physiol 141(1):243–256. https://doi.org/10.1104/pp.106.079038

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Ding L, Pandey S, Assmann SM (2008) Arabidopsis extra-large G proteins (XLGs) regulate root morphogenesis. Plant J 53(2):248–263. https://doi.org/10.1111/j.1365-313X.2007.03335.x

    Article  CAS  PubMed  Google Scholar 

  216. Chakravorty D, Trusov Y, Zhang W, Acharya BR, Sheahan MB, McCurdy DW, Assmann SM, Botella JR (2011) An atypical heterotrimeric G-protein gamma-subunit is involved in guard cell K(+)-channel regulation and morphological development in Arabidopsis thaliana. Plant J 67(5):840–851. https://doi.org/10.1111/j.1365-313X.2011.04638.x

    Article  CAS  PubMed  Google Scholar 

  217. Urano D, Chen JG, Botella JR, Jones AM (2013) Heterotrimeric G protein signalling in the plant kingdom. Open Biol 3(3):120186. https://doi.org/10.1098/rsob.120186

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Wolfenstetter S, Chakravorty D, Kula R, Urano D, Trusov Y, Sheahan MB, McCurdy DW, Assmann SM, Jones AM, Botella JR (2015) Evidence for an unusual transmembrane configuration of AGG3, a class C Ggamma subunit of Arabidopsis. Plant J 81(3):388–398. https://doi.org/10.1111/tpj.12732

    Article  CAS  PubMed  Google Scholar 

  219. Urano D, Maruta N, Trusov Y, Stoian R, Wu Q, Liang Y, Jaiswal DK, Thung L, Jackson D, Botella JR, Jones AM (2016) Saltational evolution of the heterotrimeric G protein signaling mechanisms in the plant kingdom. Sci Signal 9(446):ra93. https://doi.org/10.1126/scisignal.aaf9558

    Article  CAS  PubMed  Google Scholar 

  220. Liang Y, Gao Y, Jones AM (2017) Extra large G-protein interactome reveals multiple stress response function and partner-dependent XLG subcellular localization. Front Plant Sci 8:1015. https://doi.org/10.3389/fpls.2017.01015

    Article  PubMed  PubMed Central  Google Scholar 

  221. Klopffleisch K, Phan N, Augustin K, Bayne RS, Booker KS, Botella JR, Carpita NC, Carr T, Chen JG, Cooke TR, Frick-Cheng A, Friedman EJ, Fulk B, Hahn MG, Jiang K, Jorda L, Kruppe L, Liu C, Lorek J, McCann MC, Molina A, Moriyama EN, Mukhtar MS, Mudgil Y, Pattathil S, Schwarz J, Seta S, Tan M, Temp U, Trusov Y, Urano D, Welter B, Yang J, Panstruga R, Uhrig JF, Jones AM (2011) Arabidopsis G-protein interactome reveals connections to cell wall carbohydrates and morphogenesis. Mol Syst Biol 7:532. https://doi.org/10.1038/msb.2011.66

    Article  PubMed  PubMed Central  Google Scholar 

  222. Fischer-Colbrie R, Eder S, Lovisetti-Scamihorn P, Becker A, Laslop A (2002) Neuroendocrine secretory protein 55: a novel marker for the constitutive secretory pathway. Ann N Y Acad Sci 971:317–322. https://doi.org/10.1111/j.1749-6632.2002.tb04486.x

    Article  CAS  PubMed  Google Scholar 

  223. Bastepe M (2007) The GNAS locus: quintessential complex gene encoding Gsalpha, XLalphas, and other imprinted transcripts. Curr Genomics 8(6):398–414. https://doi.org/10.2174/138920207783406488

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Freson K, Jaeken J, Van Helvoirt M, de Zegher F, Wittevrongel C, Thys C, Hoylaerts MF, Vermylen J, Van Geet C (2003) Functional polymorphisms in the paternally expressed XLalphas and its cofactor ALEX decrease their mutual interaction and enhance receptor-mediated cAMP formation. Hum Mol Genet 12(10):1121–1130. https://doi.org/10.1093/hmg/ddg130

    Article  CAS  PubMed  Google Scholar 

  225. Shatsky M, Nussinov R, Wolfson HJ (2004) A method for simultaneous alignment of multiple protein structures. Proteins 56(1):143–156. https://doi.org/10.1002/prot.10628

    Article  CAS  PubMed  Google Scholar 

  226. Waldschmidt HV, Homan KT, Cruz-Rodriguez O, Cato MC, Waninger-Saroni J, Larimore KM, Cannavo A, Song J, Cheung JY, Kirchhoff PD, Koch WJ, Tesmer JJ, Larsen SD (2016) Structure-based design, synthesis, and biological evaluation of highly selective and potent G protein-coupled receptor kinase 2 inhibitors. J Med Chem 59(8):3793–3807. https://doi.org/10.1021/acs.jmedchem.5b02000

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported in part by the President of the Russian Federation Scholarship SP-3665.2018.4 (A.V.F.), grants from Russian Science Foundation RSCF 18-75-10115 (A.V.F.) and RSCF 19-15-00107 (K.K.T.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Vladimir N. Uversky.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (PDF 18461 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fonin, A.V., Darling, A.L., Kuznetsova, I.M. et al. Multi-functionality of proteins involved in GPCR and G protein signaling: making sense of structure–function continuum with intrinsic disorder-based proteoforms. Cell. Mol. Life Sci. 76, 4461–4492 (2019). https://doi.org/10.1007/s00018-019-03276-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-019-03276-1

Keywords

Navigation