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.

  • Article
  • Published:

Orthosteric and allosteric action of the C5a receptor antagonists

Abstract

The C5a receptor (C5aR) is a G-protein-coupled receptor (GPCR) that can induce strong inflammatory response to the anaphylatoxin C5a. Targeting C5aR has emerged as a novel anti-inflammatory therapeutic method. However, developing potent C5aR antagonists as drugs has proven difficult. Here, we report two crystal structures of human C5aR in ternary complexes with the peptide antagonist PMX53 and a non-peptide antagonist, either avacopan or NDT9513727. The structures, together with other biophysical, computational docking and cell-based signaling data, reveal the orthosteric action of PMX53 and its effect of stabilizing the C5aR structure, as well as the allosteric action of chemically diverse non-peptide C5aR antagonists with different binding poses. Structural comparison analysis suggests the presence of similar allosteric sites in other GPCRs. We also discuss critical structural features of C5aR in activation, including a novel conformation of helix 8. On the basis of our results, we suggest novel strategies for developing C5aR-targeting drugs.

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

Fig. 1: Concurrent binding of PMX53 and NDT9513727 to C5aR and overall structures.
Fig. 2: Orthosteric binding site for PMX53.
Fig. 3: Allosteric binding sites for NDT9513727 and avacopan.
Fig. 4: Different binding poses of chemically diverse C5aR allosteric antagonists and the potential allosteric site in P2Y1 receptor.
Fig. 5: Structural motifs of C5aR in receptor activation.
Fig. 6: HDX-MS results of C5aR with two ligands.
Fig. 7: Unique conformation of helix 8.

Similar content being viewed by others

References

  1. Klos, A. et al. The role of the anaphylatoxins in health and disease. Mol. Immunol. 46, 2753–2766 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Klos, A., Wende, E., Wareham, K. J. & Monk, P. N. International union of basic and clinical pharmacology. [corrected]. LXXXVII. Complement peptide C5a, C4a, and C3a receptors. Pharmacol. Rev. 65, 500–543 (2013).

    Article  PubMed  CAS  Google Scholar 

  3. Bjarnadóttir, T. K. et al. Comprehensive repertoire and phylogenetic analysis of the G protein-coupled receptors in human and mouse. Genomics 88, 263–273 (2006).

    Article  PubMed  CAS  Google Scholar 

  4. Guo, R. F. & Ward, P. A. Role of C5a in inflammatory responses. Annu. Rev. Immunol. 23, 821–852 (2005).

    Article  PubMed  CAS  Google Scholar 

  5. Fonseca, M. I. et al. Contribution of complement activation pathways to neuropathology differs among mouse models of Alzheimer’s disease. J. Neuroinflammation 8, 4 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Landlinger, C. et al. Active immunization against complement factor C5a: a new therapeutic approach for Alzheimer’s disease. J. Neuroinflammation 12, 150 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Quadros, A. U. & Cunha, T. M. C5a and pain development: An old molecule, a new target. Pharmacol. Res. 112, 58–67 (2016).

    Article  PubMed  CAS  Google Scholar 

  8. Pandey, M. K. et al. Complement drives glucosylceramide accumulation and tissue inflammation in Gaucher disease. Nature 543, 108–112 (2017).

    Article  PubMed  CAS  Google Scholar 

  9. Markiewski, M. M. et al. Modulation of the antitumor immune response by complement. Nat. Immunol. 9, 1225–1235 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Nitta, H. et al. Enhancement of human cancer cell motility and invasiveness by anaphylatoxin C5a via aberrantly expressed C5a receptor (CD88). Clin. Cancer Res. 19, 2004–2013 (2013).

    Article  PubMed  CAS  Google Scholar 

  11. Vadrevu, S. K. et al. Complement c5a receptor facilitates cancer metastasis by altering T-cell responses in the metastatic niche. Cancer Res. 74, 3454–3465 (2014).

    Article  PubMed  CAS  Google Scholar 

  12. Darling, V. R., Hauke, R. J., Tarantolo, S. & Agrawal, D. K. Immunological effects and therapeutic role of C5a in cancer. Expert Rev. Clin. Immunol. 11, 255–263 (2015).

    Article  PubMed  CAS  Google Scholar 

  13. Woodruff, T. M., Nandakumar, K. S. & Tedesco, F. Inhibiting the C5-C5a receptor axis. Mol. Immunol. 48, 1631–1642 (2011).

    Article  PubMed  CAS  Google Scholar 

  14. Finch, A. M. et al. Low-molecular-weight peptidic and cyclic antagonists of the receptor for the complement factor C5a. J. Med. Chem. 42, 1965–1974 (1999).

    Article  PubMed  CAS  Google Scholar 

  15. March, D. R. et al. Potent cyclic antagonists of the complement C5a receptor on human polymorphonuclear leukocytes. Relationships between structures and activity. Mol. Pharmacol. 65, 868–879 (2004).

    Article  PubMed  CAS  Google Scholar 

  16. Monk, P. N., Scola, A. M., Madala, P. & Fairlie, D. P. Function, structure and therapeutic potential of complement C5a receptors. Br. J. Pharmacol. 152, 429–448 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Sumichika, H. et al. Identification of a potent and orally active non-peptide C5a receptor antagonist. J. Biol. Chem. 277, 49403–49407 (2002).

    Article  PubMed  CAS  Google Scholar 

  18. Brodbeck, R. M. et al. Identification and characterization of NDT 9513727 [N,N-bis(1,3-benzodioxol-5-ylmethyl)-1-butyl-2,4-diphenyl-1H-imidazole-5-methanamine], a novel, orally bioavailable C5a receptor inverse agonist. J. Pharmacol. Exp. Ther. 327, 898–909 (2008).

    Article  PubMed  CAS  Google Scholar 

  19. Bekker, P. et al. Characterization of pharmacologic and pharmacokinetic properties of CCX168, a potent and selective orally administered complement 5a receptor inhibitor, based on preclinical evaluation and randomized phase 1 clinical study. PLoS One 11, e0164646 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Seow, V. et al. Receptor residence time trumps drug-likeness and oral bioavailability in determining efficacy of complement C5a antagonists. Sci. Rep. 6, 24575 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Jayne, D. R. W. et al. Randomized trial of C5a receptor inhibitor avacopan in ANCA-associated vasculitis. J. Am. Soc. Nephrol. 28, 2756–2767 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Robertson, N. et al. Structure of the complement C5a receptor bound to the extra-helical antagonist NDT9513727. Nature 553, 111–114 (2018).

    Article  PubMed  CAS  Google Scholar 

  23. Lu, J. et al. Structural basis for the cooperative allosteric activation of the free fatty acid receptor GPR40. Nat. Struct. Mol. Biol. 24, 570–577 (2017).

    Article  PubMed  CAS  Google Scholar 

  24. Chun, E. et al. Fusion partner toolchest for the stabilization and crystallization of G protein-coupled receptors. Structure 20, 967–976 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Finch, A. M. et al. Biologically active conformer of the effector region of human C5a and modulatory effects of N-terminal receptor binding determinants on activity. J. Med. Chem. 40, 877–884 (1997).

    Article  PubMed  CAS  Google Scholar 

  26. Siciliano, S. J. et al. Two-site binding of C5a by its receptor: an alternative binding paradigm for G protein-coupled receptors. Proc. Natl. Acad. Sci. USA 91, 1214–1218 (1994).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Klco, J. M., Lassere, T. B. & Baranski, T. J. C5a receptor oligomerization. I. Disulfide trapping reveals oligomers and potential contact surfaces in a G protein-coupled receptor. J. Biol. Chem. 278, 35345–35353 (2003).

    Article  PubMed  CAS  Google Scholar 

  28. White, J. F. et al. Structure of the agonist-bound neurotensin receptor. Nature 490, 508–513 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Qin, L. et al. Structural biology. Crystal structure of the chemokine receptor CXCR4 in complex with a viral chemokine. Science 347, 1117–1122 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Shihoya, W. et al. Activation mechanism of endothelin ETB receptor by endothelin-1. Nature 537, 363–368 (2016).

    Article  PubMed  CAS  Google Scholar 

  31. Ma, Y. et al. Structural basis for apelin control of the human apelin receptor. Structure 25, 858–866.e854 (2017).

    Article  PubMed  CAS  Google Scholar 

  32. Cain, S. A., Coughlan, T. & Monk, P. N. Mapping the ligand-binding site on the C5a receptor: arginine74 of C5a contacts aspartate282 of the C5a receptor. Biochemistry 40, 14047–14052 (2001).

    Article  PubMed  CAS  Google Scholar 

  33. Baranski, T. J. et al. C5a receptor activation. Genetic identification of critical residues in four transmembrane helices. J. Biol. Chem. 274, 15757–15765 (1999).

    Article  PubMed  CAS  Google Scholar 

  34. Higginbottom, A. et al. Comparative agonist/antagonist responses in mutant human C5a receptors define the ligand binding site. J. Biol. Chem. 280, 17831–17840 (2005).

    Article  PubMed  CAS  Google Scholar 

  35. Nikiforovich, G. V., Marshall, G. R. & Baranski, T. J. Modeling molecular mechanisms of binding of the anaphylatoxin C5a to the C5a receptor. Biochemistry 47, 3117–3130 (2008).

    Article  PubMed  CAS  Google Scholar 

  36. Qu, H., Ricklin, D. & Lambris, J. D. Recent developments in low molecular weight complement inhibitors. Mol. Immunol. 47, 185–195 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Waters, S. M. et al. Molecular characterization of the gerbil C5a receptor and identification of a transmembrane domain V amino acid that is crucial for small molecule antagonist interaction. J. Biol. Chem. 280, 40617–40623 (2005).

    Article  PubMed  CAS  Google Scholar 

  38. Zhang, D. et al. Two disparate ligand-binding sites in the human P2Y1 receptor. Nature 520, 317–321 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Gerber, B. O., Meng, E. C., Dotsch, V., Baranski, T. J. & Bourne, H. R. An activation switch in the ligand binding pocket of the C5a receptor. J. Biol. Chem. 276, 3394–3400 (2001).

    Article  PubMed  CAS  Google Scholar 

  40. Wu, B. et al. Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists. Science 330, 1066–1071 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Konteatis, Z. D. et al. Development of C5a receptor antagonists. Differential loss of functional responses. J. Immunol. 153, 4200–4205 (1994).

    PubMed  CAS  Google Scholar 

  42. Jia, N. et al. Pivotal advance: Interconversion between pure chemotactic ligands and chemoattractant/secretagogue ligands of neutrophil C5a receptor by a single amino acid substitution. J. Leukoc. Biol. 87, 965–975 (2010).

    Article  PubMed  CAS  Google Scholar 

  43. Rasmussen, S. G. et al. Structure of a nanobody-stabilized active state of the β(2) adrenoceptor. Nature 469, 175–180 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Huang, W. et al. Structural insights into µ-opioid receptor activation. Nature 524, 315–321 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Whistler, J. L. et al. Constitutive activation and endocytosis of the complement factor 5a receptor: evidence for multiple activated conformations of a G protein-coupled receptor. Traffic 3, 866–877 (2002).

    Article  PubMed  CAS  Google Scholar 

  46. Angel, T. E., Chance, M. R. & Palczewski, K. Conserved waters mediate structural and functional activation of family A (rhodopsin-like) G protein-coupled receptors. Proc. Natl. Acad. Sci. USA 106, 8555–8560 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Fenalti, G. et al. Molecular control of δ-opioid receptor signalling. Nature 506, 191–196 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Liu, W. et al. Structural basis for allosteric regulation of GPCRs by sodium ions. Science 337, 232–236 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Zhang, C. et al. High-resolution crystal structure of human protease-activated receptor 1. Nature 492, 387–392 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Li, S., Lee, S. Y. & Chung, K. Y. Conformational analysis of g protein-coupled receptor signaling by hydrogen/deuterium exchange mass spectrometry. Methods Enzymol 557, 261–278 (2015).

    Article  PubMed  CAS  Google Scholar 

  51. Konermann, L., Tong, X. & Pan, Y. Protein structure and dynamics studied by mass spectrometry: H/D exchange, hydroxyl radical labeling, and related approaches. J. Mass Spectrom. 43, 1021–1036 (2008).

    Article  PubMed  CAS  Google Scholar 

  52. Suvorova, E. S., Gripentrog, J. M., Oppermann, M. & Miettinen, H. M. Role of the carboxyl terminal di-leucine in phosphorylation and internalization of C5a receptor. Biochim. Biophys. Acta 1783, 1261–1270 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Rasmussen, S. G. et al. Crystal structure of the β2 adrenergic receptor-Gs protein complex. Nature 477, 549–555 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Zhou, X. E. et al. Identification of phosphorylation codes for arrestin recruitment by G protein-coupled receptors. Cell 170, 457–469.e413 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  55. Gripentrog, J. M. & Miettinen, H. M. Formyl peptide receptor-mediated ERK1/2 activation occurs through G(i) and is not dependent on beta-arrestin1/2. Cell. Signal. 20, 424–431 (2008).

    Article  PubMed  CAS  Google Scholar 

  56. Moriconi, A. et al. Targeting the minor pocket of C5aR for the rational design of an oral allosteric inhibitor for inflammatory and neuropathic pain relief. Proc. Natl. Acad. Sci. USA 111, 16937–16942 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Zeng, Q. et al. Discovery of an irreversible HCV NS5B polymerase inhibitor. Bioorg. Med. Chem. Lett. 23, 6585–6587 (2013).

    Article  PubMed  CAS  Google Scholar 

  58. Li, R., Coulthard, L. G., Wu, M. C., Taylor, S. M. & Woodruff, T. M. C5L2: a controversial receptor of complement anaphylatoxin, C5a. FASEB J. 27, 855–864 (2013).

    Article  PubMed  CAS  Google Scholar 

  59. Van Lith, L. H., Oosterom, J., Van Elsas, A. & Zaman, G. J. C5a-stimulated recruitment of beta-arrestin2 to the nonsignaling 7-transmembrane decoy receptor C5L2. J. Biomol. Screen. 14, 1067–1075 (2009).

    Article  PubMed  CAS  Google Scholar 

  60. Zhang, H. et al. Structural basis for selectivity and diversity in angiotensin II receptors. Nature 544, 327–332 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Caffrey, M. Crystallizing membrane proteins for structure determination: use of lipidic mesophases. Annu. Rev. Biophys. 38, 29–51 (2009).

    Article  PubMed  CAS  Google Scholar 

  62. Otwinowski, Z. & Minor, W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997).

    Article  CAS  PubMed  Google Scholar 

  63. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  PubMed  CAS  Google Scholar 

  66. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D Biol. Crystallogr. 66, 12–21 (2010).

    Article  PubMed  CAS  Google Scholar 

  67. Williams, C. J. et al. MolProbity: More and better reference data for improved all-atom structure validation. Protein Sci. 27, 293–315 (2018).

    Article  PubMed  CAS  Google Scholar 

  68. Mysinger, M. M. & Shoichet, B. K. Rapid context-dependent ligand desolvation in molecular docking. J. Chem. Inf. Model. 50, 1561–1573 (2010).

    Article  PubMed  CAS  Google Scholar 

  69. Duc, N. M. et al. Effective application of bicelles for conformational analysis of G protein-coupled receptors by hydrogen/deuterium exchange mass spectrometry. J. Am. Soc. Mass Spectrom. 26, 808–817 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank the staff at the GM/CA @ APS of Argonne National Laboratory at Chicago for their assistance with X-ray diffraction data collection. We acknowledge support from the University of Pittsburgh by the UPMC Competitive Medical Research Fund (CMRF) (C.Z.), the National Research Foundation of Korea funded by the Korean government (NFR-2015R1A1A1A05027473) (K.Y.C.), the funding support from the Biomedical Research Council, A*STAR (R.N.V.K.D. and H.F.) and the funding support from National Natural Science Foundation of China (31770791 and 315707410) (Z.W.). Z.W. is also supported by the Recruitment Program of Global Youth Experts of China.

Author information

Authors and Affiliations

Authors

Contributions

H.L. and C.Z. designed the research. H.L. performed all of the experiments for protein expression, purification, crystallization and functional characterization. H.R.K. and K.Y.C. performed HDX-MS experiments and data analysis. R.N.V.K.D. and H.F. performed computational docking studies. L.W. assisted with X-ray diffraction data collection. Z.W. determined the crystal structures. C.Z. processed the X-ray diffraction data and supervised the protein expression, purification, crystallization and functional studies. Z.W., K.Y.C. and C.Z. prepared the manuscript with the assistance from H.F.

Corresponding author

Correspondence to Cheng Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated Supplementary Information

Supplementary Figure 1 Chemically diverse non-peptide C5aR antagonists and C5aR crystallization.

(a) Chemical structures of four C5aR non-peptide antagonists: avacopan (CCX168), NDT9513727, W54011 and CP-447697. The central tertiary nitrogen group is circled. (b) To aid the crystallization, we engineered human wtC5aR by replacing the N-terminal residues 1-29 with cytochrome b262 RIL (BRIL). We also introduced a TEV protease site between 331 and 332 and cut off the C-terminal segment 332-350 before crystallization by TEV protease. The construct is referred as C5aR-BRIL. Crystals of C5aR with PMX53 and avacopan in lipidic cubic phase are shown in the upper right corner. (c) The Fo-Fc omit electron-density maps for PMX53 (green), NDT9513727 (cyan) and avacopan (orange) contoured at 2σ are shown as purple mesh.

Supplementary Figure 2 Structural comparison with thermostable C5aR, crystal packing and ligand binding at the intracellular surface.

(a) Left: Superimposition of the C5aR-PMX53-NDT9513727 structure with the structure of thermostable C5aR-NDT95132727. Right: The surfaces of these two structures viewed from the extracellular side. The large differences are observed for ECL2 and the extracellular end of TM4. The unique conformation of helix 8 is observed in both structures. (b) Top: Lattice packing in C5aR-PMX53-avacopan crystals. Receptor is shown in blue and BRIL is shown in purple. PMX53 and avacopan are shown in green and orange respectively. In any two C5aR molecules that form crystal-packing interactions, the allosteric sites are far away from each other with a distance of ~27 Å (measured between two W255 residues in the sites). Bottom: In each layer of the crystal lattice, one receptor packs against the surrounding receptor molecules in an anti-parallel manner, excluding the possibility of forming physiological C5aR dimers in the crystals. (c) The malonate molecule (yellow) bound at the cytoplasmic surface of C5aR-PMX53-avacopan (blue). An Fo-Fc omit electron-density map for the malonate contoured at 2σ is shown as purple mesh. Residues N712.39, R1343.50 and T2406.33 that form hydrogen-bonding interactions with the malonate are shown as sticks.

Supplementary Figure 3 Comparison of C5aR structure to the structures of other GPCRs with peptide ligands.

Crystal structures of GPCRs bound to the peptide ligands: C5aR with PMX53; CXCR4 with CVX15 (PDB ID 3OE0); CXCR4 with mVIP-II (PDB ID 4RWS); neurotensin receptor 1 (NTSR) with neurotensin (8-13) (PDB ID 4GRV); endothelin receptor type-B (ETBR) with endothelin-1 (PDB ID 5GLH); apelin receptor (Apelin R) with an apelin mimetic peptide (PDB ID 5VBL). C5aR is colored in blue and other GPCRs are colored in grey. PMX53 is colored in green and other peptide ligands are colored in red. PMX53 interacts with ECL2 but not N-terminus of C5aR, while other peptide ligands all interact with the N-termini and ECL2s of their receptors.

Supplementary Figure 4 Allosteric site for NDT9513727.

(a) Allosteric binding site for NDT9513727 (cyan) in our structure. All the residues that interact with NDT9513727 are in the same conformation as observed in the structure of thermostable C5aR. However, the residue A1564.45 was mutated to leucine in the thermostable C5aR, which may provide additional hydrophobic interactions for the binding of NDT9513727. (b) The effects of 6 mutations V159A, V159H, L163W, L209W, W213L and W213G on the antagonistic action of NDT9513727 measured by an agonist YR10-induced Ca2 + release assay. The cellular calcium level was determined by measuring the fluorescence of fluo-4 in HEK293T cells stably expressing wtC5aR. 10 μM YR10 was used to stimulate the receptor. Data points are presented as the mean values + /- SEM, n = 3.

Supplementary Figure 5 Conservation of the allosteric site in other GPCRs.

Crystal structures of GPCRs with small compounds bound to the similar site as identified in C5aR and GPR40 for the allosteric modulators (PDB IDs shown in the brackets): (a) C5aR with avacopan; (b) δ-opioid receptor with 1-oleoyl-R-glycerol (DOR, 4N6H); (c) μ-opioid receptor with a PEG400 molecule (MOR, 5C1M); (d) N/OFQ opioid receptor with 1-oleoyl-R-glycerol (NOR, 4EA3); (e) viral GPCR US28 with a succinic acid molecule (US28, 4XT1); (f) A2a adenosine receptor with an oleic acid molecule (A2AR, 5K2D); (g) chemokine receptor CCR9 with an oleic acid molecule (CCR9, 5LWE); (i) cannabinoid receptor CB1 with an oleic acid molecule (CB1, 5TGZ). The cysteine residue at 5.57 position C5.57 is conserved in all of these GPCRs except A2AR and CB1.

Supplementary Figure 6 Different sodium coordination sites in four GPCRs.

The sodium coordination sites in four GPCRs: C5aR, protease-activated receptor 1(PAR1, PDB ID 3VW7), A2A adenosine receptor (A2AR, PDB ID 4EIY) and δ-opioid receptor (δOR, PDB ID 4N6H) are shown. The sodium ion (shown as a purple sphere) interacts with different sets of residues at the cytoplasmic side of these GPCRs and water molecules (shown as red spheres).

Supplementary Figure 7 HDX-MS sequence coverage map and deuterium uptake plots.

(a) Bars indicated the peptic peptides in different regions of C5aR for HDX analysis. (b) Deuterium uptake plots of those regions in C5aR are represented. The results of three samples, C5aR bound to NDT9513727 (NDT), C5aR bound to PMX53 (PMX53) and C5aR bound to both ligands (NDT + PMX53), are compared. There is no significant difference between the results of PMX53 and NDT + PMX53. Results are means ± S.E. for at least three independent experiments. *p < 0.05: NDT-bound state vs. PMX53-bound state, #p < 0.05: NDT-bound state vs. NDT and PMX53-bound state

Supplementary Figure 8 Unique helix 8 in C5aR.

(a) Hydrophobic interactions that stabilize the conformation of helix 8. In both C5aR structures, residues L315, L319, V322 and L323 on helix 8 form extensive hydrophobic interactions with the surrounding residues L571.52, V581.53, V611.56, F752.43, V2436.36, Y3007.53 and V3017.54 from TM1, TM2, TM6 and TM7, respectively. (b) The conformation of helix 8 in four GPCRs: C5aR, angiotensin II receptor 2 (AT2R, PDB ID 5UNF), δ-opioid receptor (δOR, PDB ID 4N6H) and protease-activated receptor 1(PAR1, PDB ID 3VW7). In nearly all solved structures of rhodopsin-like GPCRs, either there is no well-folded helix 8 as seen in PAR1 or the helix 8 adopts a similar conformation as that in δOR with the C-terminus extending away from the helical bundle. In AT2R, the helix 8 adopts a unique conformation to interact with the cytoplasmic surface, which can sterically block the binding of signaling effectors and result in the lacking of signaling responses for AT2R. In C5aR, the helix 8 is in a reversed orientation compared to that in δOR and inserts between TM1 and TM7. Such conformation has not been observed in other GPCRs so far.

Supplementary Figure 9 Extracellular sites for potential non-peptide C5aR antagonist.

(a) Alignment of C5aR structure with the structures of two angiotensin II receptors AT1R (pink, PDB ID 4ZUD) and AT2R (light pink, PDB ID 5UNF). PMX53 is colored in green and the antagonists of AT1R and AT2R are colored in red. The binding sites for angiotensin antagonists overlap with the bottom region of the PMX53 binding site, suggesting the feasibility of developing non-peptide C5aR antagonists that target this region. (b) Extracellular allosteric antagonist DF2593A. Two residues, D2827.35 and N1193.35, were proposed to form direct polar interactions with DF2593A. However, there is no enough space in our structures around these two residues to accommodate DF2593A, even with the absence of PMX53 (green sticks). It is likely that DF2593A binding is associated with large conformational changes of the extracellular region, resulting in a distinct binding pocket compared to PMX53.

Supplementary Figure 10 Sequence alignment of C5aR and C5L2.

The purple arrows point to the residues in C5aR that are involved in the interactions with PMX53, including F44, L92, I96, P113, I116, R178, E199, R206, Y178, T261, D282 and V286. Except for F44, R178 and T261, all other residues are conserved in C5L2. However, the substitution of G189 in C5aR, as indicated by the blue star, by V187 in C5L2 may introduce steric clash with PMX53. The right panel shows the position of G189 relative to PMX53 (green sticks) in the structure of C5aR (blue). The distance between G189 and the Pro residue in PMX53 is too short to tolerate any substitution of G189. The orange star indicates the residue W213 in C5aR, which is critical for the binding of non-peptide antagonists. It is substituted by G211 in C5L2.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, H., Kim, H.R., Deepak, R.N.V.K. et al. Orthosteric and allosteric action of the C5a receptor antagonists. Nat Struct Mol Biol 25, 472–481 (2018). https://doi.org/10.1038/s41594-018-0067-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-018-0067-z

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