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:

Structural basis for therapeutic inhibition of complement C5

Abstract

Activation of complement C5 generates the potent anaphylatoxin C5a and leads to pathogen lysis, inflammation and cell damage. The therapeutic potential of C5 inhibition has been demonstrated by eculizumab, one of the world's most expensive drugs. However, the mechanism of C5 activation by C5 convertases remains elusive, thus limiting development of therapeutics. Here we identify and characterize a new protein family of tick-derived C5 inhibitors. Structures of C5 in complex with the new inhibitors, the phase I and phase II inhibitor OmCI, or an eculizumab Fab reveal three distinct binding sites on C5 that all prevent activation of C5. The positions of the inhibitor-binding sites and the ability of all three C5–inhibitor complexes to competitively inhibit the C5 convertase conflict with earlier steric-inhibition models, thus suggesting that a priming event is needed for activation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Complement inhibition by RaCI.
Figure 2: Crystal structures of C5–OmCI–RaCI complexes and binding mode of RaCI family and OmCI inhibitors.
Figure 3: Remodeling of the C5a domain in the inhibited complex.
Figure 4: The structurally conserved core of the RaCI family members is sufficient for activity.
Figure 5: Eculizumab inhibits C5 activation by binding to a site centered on the MG7 domain.
Figure 6: All the inhibited C5 complexes can themselves inhibit cleavage of C5 by the native convertase, but not all the inhibitors inhibit cleavage by a CVF convertase.
Figure 7: The structures of the inhibited C5 complexes and competition assay yield an altered view of the C5-activation pathway.

Similar content being viewed by others

Accession codes

Primary accessions

Biological Magnetic Resonance Data Bank

Electron Microscopy Data Bank

NCBI Reference Sequence

Protein Data Bank

Referenced accessions

GenBank/EMBL/DDBJ

Protein Data Bank

References

  1. Kemper, C. & Köhl, J. Novel roles for complement receptors in T cell regulation and beyond. Mol. Immunol. 56, 181–190 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Ricklin, D., Hajishengallis, G., Yang, K. & Lambris, J.D. Complement: a key system for immune surveillance and homeostasis. Nat. Immunol. 11, 785–797 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Law, S.K. & Dodds, A.W. The internal thioester and the covalent binding properties of the complement proteins C3 and C4. Protein Sci. 6, 263–274 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Coulthard, L.G. & Woodruff, T.M. Is the complement activation product C3a a proinflammatory molecule? Re-evaluating the evidence and the myth. J. Immunol. 194, 3542–3548 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Pangburn, M.K. & Rawal, N. Structure and function of complement C5 convertase enzymes. Biochem. Soc. Trans. 30, 1006–1010 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Rawal, N. & Pangburn, M. Formation of high-affinity C5 convertases of the alternative pathway of complement. J. Immunol. 166, 2635–2642 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Rawal, N. & Pangburn, M.K. Formation of high affinity C5 convertase of the classical pathway of complement. J. Biol. Chem. 278, 38476–38483 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Daha, M.R., Fearon, D.T. & Austen, K.F. C3 requirements for formation of alternative pathway C5 convertase. J. Immunol. 117, 630–634 (1976).

    CAS  PubMed  Google Scholar 

  9. Medicus, R.G., Schreiber, R.D., Götze, O. & Müller-Eberhard, H.J. A molecular concept of the properdin pathway. Proc. Natl. Acad. Sci. USA 73, 612–616 (1976).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Morgan, B.P. The membrane attack complex as an inflammatory trigger. Immunobiology doi:10.1016/j.imbio.2015.04.006 (30 April 2015).

  12. Markiewski, M.M. & Lambris, J.D. The role of complement in inflammatory diseases from behind the scenes into the spotlight. Am. J. Pathol. 171, 715–727 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ricklin, D. & Lambris, J.D. Complement in immune and inflammatory disorders: pathophysiological mechanisms. J. Immunol. 190, 3831–3838 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Mollnes, T.E. & Kirschfink, M. Strategies of therapeutic complement inhibition. Mol. Immunol. 43, 107–121 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Nunn, M.A. et al. Complement inhibitor of C5 activation from the soft tick Ornithodoros moubata. J. Immunol. 174, 2084–2091 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Nene, V. et al. Genes transcribed in the salivary glands of female Rhipicephalus appendiculatus ticks infected with Theileria parva. Insect Biochem. Mol. Biol. 34, 1117–1128 (2004).

    Article  PubMed  Google Scholar 

  17. Barratt-Due, A. et al. Ornithodoros moubata complement inhibitor is an equally effective C5 inhibitor in pigs and humans. J. Immunol. 187, 4913–4919 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Hepburn, N.J., Chamberlain-Banoub, J.L., Williams, A.S., Morgan, B.P. & Harris, C.L. Prevention of experimental autoimmune myasthenia gravis by rat Crry-Ig: a model agent for long-term complement inhibition in vivo. Mol. Immunol. 45, 395–405 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Fredslund, F. et al. Structure of and influence of a tick complement inhibitor on human complement component 5. Nat. Immunol. 9, 753–760 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Laursen, N.S. et al. Substrate recognition by complement convertases revealed in the C5-cobra venom factor complex. EMBO J. 30, 606–616 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Laursen, N.S. et al. Structural basis for inhibition of complement C5 by the SSL7 protein from Staphylococcus aureus. Proc. Natl. Acad. Sci. USA 107, 3681–3686 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhang, X., Boyar, W., Galakatos, N. & Gonnella, N.C. Solution structure of a unique C5a semi-synthetic antagonist: implications in receptor binding. Protein Sci. 6, 65–72 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kini, R.M. & Doley, R. Structure, function and evolution of three-finger toxins: mini proteins with multiple targets. Toxicon 56, 855–867 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Mans, B.J. & Ribeiro, J.M. Function, mechanism and evolution of the moubatin-clade of soft tick lipocalins. Insect Biochem. Mol. Biol. 38, 841–852 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Roversi, P. et al. Bifunctional lipocalin ameliorates murine immune complex-induced acute lung injury. J. Biol. Chem. 288, 18789–18802 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Roversi, P. et al. The structure of OMCI, a novel lipocalin inhibitor of the complement system. J. Mol. Biol. 369, 784–793 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nishimura, J. et al. Genetic variants in C5 and poor response to eculizumab. N. Engl. J. Med. 370, 632–639 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Thomas, T.C. et al. Inhibition of complement activity by humanized anti-C5 antibody and single-chain Fv. Mol. Immunol. 33, 1389–1401 (1996).

    Article  CAS  PubMed  Google Scholar 

  29. Elmlund, D. & Elmlund, H. Cryogenic electron microscopy and single-particle analysis. Annu. Rev. Biochem. 84, 499–517 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Janssen, B.J. et al. Insights into complement convertase formation based on the structure of the factor B-cobra venom factor complex. EMBO J. 28, 2469–2478 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mortensen, S. et al. Structural basis for the function of complement component C4 within the classical and lectin pathways of complement. J. Immunol. 194, 5488–5496 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Rooijakkers, S.H. et al. Structural and functional implications of the alternative complement pathway C3 convertase stabilized by a staphylococcal inhibitor. Nat. Immunol. 10, 721–727 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Janssen, B.J., Christodoulidou, A., McCarthy, A., Lambris, J.D. & Gros, P. Structure of C3b reveals conformational changes that underlie complement activity. Nature 444, 213–216 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Reis, E.S. et al. Applying complement therapeutics to rare diseases. Clin. Immunol. 161, 225–240 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Vogt, W., Schmidt, G., Von Buttlar, B. & Dieminger, L. A new function of the activated third component of complement: binding to C5, an essential step for C5 activation. Immunology 34, 29–40 (1978).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Kinoshita, T. et al. C5 convertase of the alternative complement pathway: covalent linkage between two C3b molecules within the trimolecular complex enzyme. J. Immunol. 141, 3895–3901 (1988).

    CAS  PubMed  Google Scholar 

  37. Takata, Y. et al. Covalent association of C3b with C4b within C5 convertase of the classical complement pathway. J. Exp. Med. 165, 1494–1507 (1987).

    Article  CAS  PubMed  Google Scholar 

  38. Berends, E.T. et al. Molecular insights into the surface-specific arrangement of complement C5 convertase enzymes. BMC Biol. 13, 93 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Rawal, N. & Pangburn, M.K. Functional role of the noncatalytic subunit of complement C5 convertase. J. Immunol. 164, 1379–1385 (2000).

    Article  CAS  PubMed  Google Scholar 

  40. Sandoval, A., Ai, R., Ostresh, J.M. & Ogata, R.T. Distal recognition site for classical pathway convertase located in the C345C/netrin module of complement component C5. J. Immunol. 165, 1066–1073 (2000).

    Article  CAS  PubMed  Google Scholar 

  41. Grabherr, M.G. et al. Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat. Biotechnol. 29, 644–652 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Roos, A. et al. Functional characterization of the lectin pathway of complement in human serum. Mol. Immunol. 39, 655–668 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Delaglio, F. et al. NMRPipe: a multidimensional spectral processing system based on UNIX pipes. J. Biomol. NMR 6, 277–293 (1995).

    Article  CAS  PubMed  Google Scholar 

  44. Güntert, P. Automated NMR structure calculation with CYANA. Methods Mol. Biol. 278, 353–378 (2004).

    PubMed  Google Scholar 

  45. Tian, Y., Opella, S.J. & Marassi, F.M. Improved chemical shift prediction by Rosetta conformational sampling. J. Biomol. NMR 54, 237–243 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Winn, M.D. et al. Overview of the CCP4 suite and current developments. Acta Crystallogr. D Biol. Crystallogr. 67, 235–242 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Emsley, P., Lohkamp, B., Scott, W.G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Adams, P.D. et al. Advances, interactions, and future developments in the CNS, Phenix, and Rosetta structural biology software systems. Annu. Rev. Biophys. 42, 265–287 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Krissinel, E. & Henrick, K. Inference of macromolecular assemblies from crystalline state. J. Mol. Biol. 372, 774–797 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Tang, G. et al. EMAN2: an extensible image processing suite for electron microscopy. J. Struct. Biol. 157, 38–46 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Elmlund, H., Elmlund, D. & Bengio, S. PRIME: probabilistic initial 3D model generation for single-particle cryo-electron microscopy. Structure 21, 1299–1306 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Elmlund, D. & Elmlund, H. SIMPLE: software for ab initio reconstruction of heterogeneous single-particles. J. Struct. Biol. 180, 420–427 (2012).

    Article  PubMed  Google Scholar 

  54. Yang, Z. et al. UCSF Chimera, MODELLER, and IMP: an integrated modeling system. J. Struct. Biol. 179, 269–278 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank N. Darvill, T. Tang and L. Britton for experimental contributions, S. Jensen for technical support, M. Slovak (Institute of Zoology, Bratislava, Slovakia) for providing salivary glands, D.R. Greaves (University of Oxford) for mouse serum and T.E. Mollnes (Oslo University Hospital) for pig serum. We thank the High-Throughput Genomics Group at the Wellcome Trust Centre for Human Genetics (funded by Wellcome Trust grant reference 090532/Z/09/Z and Medical Research Council Hub grant G0900747 91070) for generating the sequencing data. This work was financially supported by a Rubicon grant from the Netherlands Organisation for Scientific Research to M.M.J. (825.11.030) and a Wellcome Investigator Award to S.M.L. (100298). We thank the staff at the Clive and Vera Ramaciotti Centre for Structural Cryo-Electron Microscopy at Monash University for assistance with EM image acquisition. We acknowledge the Diamond Light Source for time on I02 under proposal MX9306. EM calculations were performed within the Multi-modal Australian Sciences Imaging and Visualisation Environment (MASSIVE).

Author information

Authors and Affiliations

Authors

Contributions

S.M.L. and M.A.N. initially conceived the work. S.M.L. and M.M.J. designed the biochemical experiments, and M.M.J. performed them with assistance from N.M.B. M.M.J. crystallized the complexes, and the structures were solved by S.M.L. and S.J. M.M.J., S.M.L. and S.J. analyzed the structures. D.S. designed, collected and solved the NMR structure. H.E. designed the EM experiments and, together with S.M.L., collected and analyzed the data. Y.I.L. processed the HiSeq data and assembled the transcriptome. All authors were involved in discussion of results and preparation of the manuscript.

Corresponding authors

Correspondence to Hans Elmlund or Susan M Lea.

Ethics declarations

Competing interests

M.M.J., M.A.N. and S.M.L. hold intellectual property in the area of use of recombinant tick complement inhibitors as therapeutic agents.

Integrated supplementary information

Supplementary Figure 1 Complement inhibition by salivary gland extract (SGE) and RaCI homologs.

(A) SGE from both male and female ticks inhibit the classical pathway in a haemolysis assay. SGE equalling 2 glands (female) or 1 gland (male) were added to the haemolysis assay. Error bars, s.e.m. (n = 3 technical replicates). (B) Inhibition of classical pathway in a haemolysis assay with supernatants from stable transfected Drosophila melanogaster S2 cell lines. Individual values of two technical replicates are shown. (C) Classical and alternative pathway inhibition by RaCI family members and OmCI using an ELISA based activation assay, similar to the Wieslab assay used in Fig. 1b. The difference between the IC50 values for CP and AP inhibition likely reflects the difference in C5 concentrations used in the assays (1% serum in CP assay versus 10% serum in the AP assay). Error bars, s.e.m. (n = 3 technical replicates). (D) Cross-species reactivity of RaCI homologues and OmCI in a haemolysis assay. Error bars, s.e.m. (n = 3 technical replicates).

Supplementary Figure 2 The custom-made Fab (EcuFab), based on the sequence of Eculizumab, is a fully active complement inhibitor.

The custom-made Fab (EcuFab), based on the sequence of Eculizumab, is a fully active complement inhibitor. Classical (CP) and alternative (AP) pathway inhibition by the Fab fragment using an ELISA based activation assay show that the IC50 values are similar to RaCIs and OmCI in Supplementary Figure 1. Error bars, s.e.m. (n = 3 technical replicates).

Supplementary Figure 3 Different location of C345c domain in our new structures of inhibited C5 compared with apo-C5.

Different location of C345c domain with respect to the rest of C5 in our new structures of inhibited C5 (wheat) compared to the earlier structures of apo-C5 (orange; Fredslund et al., Nat. Immunol. 9, 753-760, 2011). The new location is consistent with that previously seen in the CVF-C5 complex (olive; Laursen et al., EMBO J. 9,606-616, 2011). The CVF component of the C5-CVF complex is shown in a salmon ribbon representation highlighting the clash with the C345c domain in the apo-C5 like position. The residues that will form the anaphylatoxin C5a are coloured in shades of red and the overall view is the same as in the close-up of this region shown in Figure 3. The structures are overlaid by superposition of the C5a domain in each structure.

Supplementary Figure 4 Overviews of the ternary complex.

(A) Front view of C5-OmCI-RaCI3 complex. (B) Cartoon showing domain organisation of C5 and interaction sites for both tick-inhibitors (yellow stars)

Supplementary Figure 5 RaCI contacts mapped onto cross-species sequence alignment of the MG1 and MG2 domains of C5.

C5 residues that make contact with one or more RaCIs in the crystal structures are highlighted in black (van der Waals interaction) and red (salt or hydrogen bonds).

Supplementary Figure 6 RaCI contacts mapped onto cross-species sequence alignment of the C5d domain of C5.

C5 residues that make contact with one or more RaCIs in the crystal structures are highlighted in black (van der Waals interaction) and red (salt or hydrogen bonds). The large deletion in the Canis lupus sequence is likely due to sequencing or assembly errors and may not represent the actual sequence.

Supplementary Figure 7 OmCI contacts mapped onto cross-species sequence alignment of the CUB and C5d domains of C5.

C5 residues that make contact with OmCI in the crystal structures are highlighted in black (van der Waals interaction) and red (salt or hydrogen bonds). The large deletion in the Canis lupus sequence is likely due to sequencing or assembly errors and may not represent the actual sequence.

Supplementary Figure 8 OmCI contacts mapped onto cross-species sequence alignment of the C345c domain of C5.

C5 residues that make contact with OmCI in the crystal structures are highlighted in black (van der Waals interaction) and red (salt or hydrogen bonds).

Supplementary Figure 9 OmCI can bind C5 and LTB4 simultaneously.

An overlay of the structure of OmCI (blue cartoon) in complex with LTB4 (VDW spheres, carbon-green, oxygen-red) (PDB ID 3zuo; Roversi et al., J. Biol. Chem 288, 18789-18802, 2013) onto OmCI (cyan cartoon) in complex with C5 (grey cartoon) demonstrates that LTB4 binding and exchange are both compatible with C5 binding. Two views related by a rotation of 180 degrees are shown with the view on the right hand side being equivalent to the views of the complex shown in Fig. 2.

Supplementary Figure 10 The binary inhibited C5 complexes are competitive inhibitors of further C5 cleavage by the native convertase.

Purified binary C5 complexes with any of OmCI, RaCI or EcuFab compete with C5 at an initial stage of the activation pathway. –ve control is histamine-binding protein 2. Error bars, s.e.m. (n = 6; 2 independent experiments with 3 technical replicates each).

Supplementary Figure 11 Eculizumab sterically hinders the binding of CVF to C5.

Crystal structure of C5-OmCI-RaCI fitted in the EM envelope as in Figure 5E. Superposition of C5-CVF complex (Laursen et al., EMBO J. 9,606-616, 2011) on the C5-OmCI-RaCI structure results in a steric clash between CVF (salmon ribbon) and the EM volume where the Fab is positioned.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 and Supplementary Tables 1 and 2 (PDF 6806 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jore, M., Johnson, S., Sheppard, D. et al. Structural basis for therapeutic inhibition of complement C5. Nat Struct Mol Biol 23, 378–386 (2016). https://doi.org/10.1038/nsmb.3196

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3196

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