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:

Structure of the C9orf72 ARF GAP complex that is haploinsufficient in ALS and FTD

Abstract

Mutation of C9orf72 is the most prevalent defect associated with amyotrophic lateral sclerosis and frontotemporal degeneration1. Together with hexanucleotide-repeat expansion2,3, haploinsufficiency of C9orf72 contributes to neuronal dysfunction4,5,6. Here we determine the structure of the C9orf72–SMCR8–WDR41 complex by cryo-electron microscopy. C9orf72 and SMCR8 both contain longin and DENN (differentially expressed in normal and neoplastic cells) domains7, and WDR41 is a β-propeller protein that binds to SMCR8 such that the whole structure resembles an eye slip hook. Contacts between WDR41 and the DENN domain of SMCR8 drive the lysosomal localization of the complex in conditions of amino acid starvation. The structure suggested that C9orf72–SMCR8 is a GTPase-activating protein (GAP), and we found that C9orf72–SMCR8–WDR41 acts as a GAP for the ARF family of small GTPases. These data shed light on the function of C9orf72 in normal physiology, and in amyotrophic lateral sclerosis and frontotemporal degeneration.

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: Cryo-EM structure of the C9orf72–SMCR8–WDR41 complex.
Fig. 2: HDX-MS of C9orf72–SMCR8 in the absence of WDR41.
Fig. 3: SMCR8 mutants do not localize on lysosomes.
Fig. 4: C9orf72–SMCR8 is a GAP for ARF proteins.

Similar content being viewed by others

Data availability

The electron microscopy density map has been deposited in the Electron Microscopy Data Bank with accession number EMD-21048. Atomic coordinates for C9orf72–SMCR8–WDR41 have been deposited in the PDB with accession number 6V4USource data are provided with this paper.

References

  1. Majounie, E. et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol. 11, 323–330 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. DeJesus-Hernandez, M. et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Renton, A. E. et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72, 257–268 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. O’Rourke, J. G. et al. C9orf72 is required for proper macrophage and microglial function in mice. Science 351, 1324–1329 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  5. Sivadasan, R. et al. C9ORF72 interaction with cofilin modulates actin dynamics in motor neurons. Nat. Neurosci. 19, 1610–1618 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Shi, Y. et al. Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nat. Med. 24, 313–325 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhang, D., Iyer, L. M., He, F. & Aravind, L. Discovery of novel DENN proteins: implications for the evolution of eukaryotic intracellular membrane structures and human disease. Front. Genet. 3, 283 (2012).

    PubMed  PubMed Central  Google Scholar 

  8. Sellier, C. et al. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J. 35, 1276–1297 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sullivan, P. M. et al. The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol. Commun. 4, 51 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Amick, J., Roczniak-Ferguson, A. & Ferguson, S. M. C9orf72 binds SMCR8, localizes to lysosomes, and regulates mTORC1 signaling. Mol. Biol. Cell 27, 3040–3051 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ugolino, J. et al. Loss of C9orf72 enhances autophagic activity via deregulated mTOR and TFEB signaling. PLoS Genet. 12, e1006443 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Yang, M. et al. A C9ORF72/SMCR8-containing complex regulates ULK1 and plays a dual role in autophagy. Sci. Adv. 2, e1601167 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  13. Jung, J. et al. Multiplex image-based autophagy RNAi screening identifies SMCR8 as ULK1 kinase activity and gene expression regulator. eLife 6, e23063 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Amick, J., Tharkeshwar, A. K., Amaya, C. & Ferguson, S. M. WDR41 supports lysosomal response to changes in amino acid availability. Mol. Biol. Cell 29, 2213–2227 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Amick, J., Tharkeshwar, A. K., Talaia, G. & Ferguson, S. M. PQLC2 recruits the C9orf72 complex to lysosomes in response to cationic amino acid starvation. J. Cell Biol. 219, e20190676 (2020).

    Article  CAS  Google Scholar 

  16. Farg, M. A. et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Hum. Mol. Genet. 23, 3579–3595 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Webster, C. P. et al. The C9orf72 protein interacts with Rab1a and the ULK1 complex to regulate initiation of autophagy. EMBO J. 35, 1656–1676 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lan, Y., Sullivan, P. M. & Hu, F. SMCR8 negatively regulates AKT and MTORC1 signaling to modulate lysosome biogenesis and tissue homeostasis. Autophagy 15, 871–885 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Shen, K. et al. Architecture of the human GATOR1 and GATOR1–Rag GTPases complexes. Nature 556, 64–69 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lawrence, R. E. et al. Structural mechanism of a Rag GTPase activation checkpoint by the lysosomal folliculin complex. Science 366, 971–977 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Shen, K. et al. Cryo-EM structure of the human FLCN–FNIP2–Rag–Ragulator complex. Cell 179, 1319–1329 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bar-Peled, L. et al. A tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 340, 1100–1106 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tsun, Z. Y. et al. The folliculin tumor suppressor is a GAP for the RagC/D GTPases that signal amino acid levels to mTORC1. Mol. Cell 52, 495–505 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Shen, K., Valenstein, M. L., Gu, X. & Sabatini, D. M. Arg78 of Nprl2 catalyzes GATOR1-stimulated GTP hydrolysis by the Rag GTPases. J. Biol. Chem. 294, 2970–2975 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sztul, E. et al. ARF GTPases and their GEFs and GAPs: concepts and challenges. Mol. Biol. Cell 30, 1249–1271 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Iyer, S., Subramanian, V. & Acharya, K. R. C9orf72, a protein associated with amyotrophic lateral sclerosis (ALS) is a guanine nucleotide exchange factor. PeerJ 6, e5815 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Wu, X. et al. Insights regarding guanine nucleotide exchange from the structure of a DENN-domain protein complexed with its Rab GTPase substrate. Proc. Natl Acad. Sci. USA 108, 18672–18677 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Corrionero, A. & Horvitz, H. R. A C9orf72 ALS/FTD ortholog acts in endolysosomal degradation and lysosomal homeostasis. Curr. Biol. 28, 1522–1535 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Jewell, J. L. et al. Differential regulation of mTORC1 by leucine and glutamine. Science 347, 194–198 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Nixon, R. A. The role of autophagy in neurodegenerative disease. Nat. Med. 19, 983–997 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Tang, D. et al. Cryo-EM structure of C9ORF72–SMCR8–WDR41 reveals the role as a GAP for Rab8a and Rab11a. Proc. Natl Acad. Sci. USA 117, 9876–9883 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Biyani, N. et al. Focus: the interface between data collection and data processing in cryo-EM. J. Struct. Biol. 198, 124–133 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhang, K. Gctf: real-time CTF determination and correction. J. Struct. Biol. 193, 1–12 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).

    PubMed  PubMed Central  Google Scholar 

  36. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    CAS  PubMed  Google Scholar 

  37. Zhang, Y. I-TASSER server for protein 3D structure prediction. BMC Bioinformatics 9, 40 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Webb, B. & Sali, A. Protein structure modeling with MODELLER. Methods Mol. Biol. 1137, 1–15 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Peng, J. & Xu, J. RaptorX: exploiting structure information for protein alignment by statistical inference. Proteins 79 (Suppl 10), 161–171 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N. & Sternberg, M. J. The Phyre2 web portal for protein modeling, prediction and analysis. Nat. Protocols 10, 845–858 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. McGuffin, L. J., Bryson, K. & Jones, D. T. The PSIPRED protein structure prediction server. Bioinformatics 16, 404–405 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  44. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D 74, 531–544 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Toso, K. L. Morris, V. Kasinath and P. Tobias for cryo-EM advice and support; X. Shi for HDX support; C. Behrends and G. Stjepanovic for suggestions and contributions to the early stages of the project; R. Lawrence and M. Lehmer for cell imaging advice; and X. Ren for assistance with cloning. This work was supported by NIH grants R01GM111730 (J.H.H.) and R01GM130995 (R.Z.), Department of Defense Peer Reviewed Medical Research Program Discovery Award W81XWH2010086 (J.H.H.), the Bakar Fellows program (J.H.H.), the Pew-Stewart Scholarship for Cancer Research and Damon Runyon-Rachleff Innovation Award (R.Z.), a postdoctoral fellowship from the Association for Frontotemporal Degeneration (M.-Y.S.) and an EMBO Long-Term Fellowship (S.A.F.).

Author information

Authors and Affiliations

Authors

Contributions

M.-Y.S. designed and carried out all experiments, except for GEF assays and lysosome colocalization experiments, and performed all data analysis. S.A.F. performed GEF assays. R.Z. carried out lysosome colocalization experiments. M.-Y.S. and J.H.H. conceptualized the project and wrote the first draft. All authors contributed to the editing of the manuscript.

Corresponding author

Correspondence to James H. Hurley.

Ethics declarations

Competing interests

J.H.H. is a scientific founder and receives research funding from Casma Therapeutics. R.Z. is a co-founder and stockholder in Frontier Medicines Corp.

Additional information

Peer review information Nature thanks Aaron Burberry, Sjors Scheres and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Purification of the C9orf72–SMCR8–WDR41 and C9orf72–SMCR8 complex, as well as the HDX data for the trimer.

a, The Superose-6 gel filtration elution profile for the C9orf72–SMCR8–WDR41 complex. b, The Superose-6 gel filtration elution profile for the C9orf72–SMCR8 complex. mAU, milli-absorbance units. c, The purified full-length C9orf72–SMCR8–WDR41 and C9orf72–SMCR8 complexes were analysed by SDS–PAGE. The proteins were purified at least five times with similar results (ac). df, Deuterium uptake data for the C9orf72–SMCR8–WDR41 complex at the 0.5-s time point, with error bars from triplicate technical measurements. Peptides with more than 50% deuterium uptake are the flexible regions. Y axis represents the average per cent deuteration. X axis demonstrates the midpoint of a single peptic peptide.

Source data

Extended Data Fig. 2 Cryo-EM data processing.

a, A representative cryo-EM micrograph of the C9orf72–SMCR8–WDR41 complex. b, Representative 2D classes. c, Orientation distribution of the aligned C9orf72–SMCR8–WDR41 particles. d, Image processing procedure.

Extended Data Fig. 3 Resolution estimation of the cryo-EM map, as well as model building and validation.

a, Comparison between FSC curves. b, Refined coordinate model fit of the indicated region in the cryo-EM density. c, Refinement and map-versus-model FSC. d, Cross-validation of test FSC curves to assess overfitting. The refinement target resolution (4.2 Å) is indicated. e. Different views of the final reconstruction.

Source data

Extended Data Fig. 4 Deuterium uptake of C9orf72–SMCR8–WDR41.

HDX-MS data are shown in heat map format, in which peptides are represented by rectangular strips above the protein sequence. Absolute deuterium uptake after 0.5, 5, 50, 500 and 50,000 s are indicated by a colour gradient below the protein sequence.

Extended Data Fig. 5 Deuterium uptake of C9orf72–SMCR8 complex.

HDX-MS data are shown in heat map format, in which peptides are represented by rectangular strips above the protein sequence. Absolute deuterium uptake after 0.5, 5, 50, 500 and 50,000 s are indicated by a colour gradient below the protein sequence.

Extended Data Fig. 6 Mapping the protected region from HDX results onto the SMCR8–C9orf72–WDR41 structure.

a, The HDX uptake difference at 0.5 s was mapped on C9orf72–SMCR8. bd, Close-up view of M1 (b) and M2–M5 (c) regions of SMCR8, and the M1 region of C9orf72 (d). e, Enlarged view of the WDR41 residues in the SMCR8–WDR41 interface.

Extended Data Fig. 7 Co-expression and pulldown validation of C9orf72–SMCR8 and SMCR8–WDR41 interface.

a, Close-up view of the residues that mediate the DENN– DENN dimerization between C9orf72 and SMCR8. b, Co-expression and pulldown experiment of Strep-tagged SMCR8 with GST–C9orf72 mutants and WDR41. c, Pulldown experiment of GST–WDR41 mutants with C9orf72–SMCR8. The pulldown experiments were carried out at least twice with similar results (b, c).

Extended Data Fig. 8 Structural comparison between C9orf72–SMCR8 and FNIP2–FLCN.

Left, structural alignment of full-length FNIP2–FLCN and C9orf72–SMCR8. Right, comparison between the longin dimers (top), DENN dimers (middle) and SMCR8 DENN with C9orf72 DENN domain (bottom).

Extended Data Fig. 9 GTPase assay for different small GTPases with C9orf72–SMCR8–WDR41.

a, SDS–PAGE of GAP protein complex (top) and GTPase proteins (bottom) used in the experiments. b, Tryptophan fluorescence GTPase signal was measured for purified RAGA–RAGC, ARL8A, ARL8B, RAB1A and RAB7A before and after addition of C9orf72–SMCR8–WDR41. The fluorescence signal upon GAP addition was normalized to 1 for each experiment. The experiments were carried out in triplicate and one representative plot is plotted. c, Tryptophan fluorescence GTPase signal was measured for purified ARF6, ARF(Q67L) or ARF5, before and after addition of C9orf72–SMCR8–WDR41, C9orf72–SMCR8(R147A)–WDR41 or C9orf72–SMCR8. d, HPLC-based GTPase assay with ARF6, ARF5, RAB1A, RAB7A, ARL8A, ARL8B and RAGA–RAGC proteins in the absence and addition of C9orf72–SMCR8–WDR41 complex, as indicated. The experiments were carried out in triplicate and one representative plot is shown. All experiments were carried out at least three times independently with similar results (ad).

Source data

Extended Data Fig. 10 HPLC-based GTPase assay with ARF1 or ARF1(Q71L) proteins.

Assays were performed in the absence and addition of GAP complex, as indicated. The experiments were carried out in triplicate and one representative plot is shown. All experiments were carried out at least three times independently with similar results.

Extended Data Fig. 11 GEF assay for different small GTPases with C9orf72–SMCR8–WDR41.

a, SDS–PAGE of C9orf72–SMCR8–WDR41 complex and GTPase proteins used in the experiments. b, GEF assay with mantGDP-reloaded RAB1A, RAB5A, RAB7A, RAB8A and RAB39B proteins in the absence and addition of C9orf72–SMCR8–WDR41 complex, as indicated. RAB5A treated with RABEX5 was used as a positive-control reaction. Data were baseline-subtracted and normalized to the signal immediately after GTP addition, which also is the 0-s time point in the plots. Plots are mean ± s.d. of each technical triplicate experiment. All experiments were carried out at least twice independently with similar results (a, b). c, Structural alignment of C9orf72–SMCR8–WDR41 with DENND1B–RAB35 (PDB 3TW8).

Source data

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics

Supplementary information

Supplementary Information

This file contains Supplementary Fig.1 presenting the uncropped gels for Fig. 2 and Extended Data Figs. 1, 7, 9, and 11.

Reporting Summary

Supplementary Data

This file contains Supplementary Dataset 1 presenting the HDX data summary.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Su, MY., Fromm, S.A., Zoncu, R. et al. Structure of the C9orf72 ARF GAP complex that is haploinsufficient in ALS and FTD. Nature 585, 251–255 (2020). https://doi.org/10.1038/s41586-020-2633-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2633-x

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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