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 and mechanism of the ER-based glucosyltransferase ALG6

Abstract

In eukaryotic protein N-glycosylation, a series of glycosyltransferases catalyse the biosynthesis of a dolichylpyrophosphate-linked oligosaccharide before its transfer onto acceptor proteins1. The final seven steps occur in the lumen of the endoplasmic reticulum (ER) and require dolichylphosphate-activated mannose and glucose as donor substrates2. The responsible enzymes—ALG3, ALG9, ALG12, ALG6, ALG8 and ALG10—are glycosyltransferases of the C-superfamily (GT-Cs), which are loosely defined as containing membrane-spanning helices and processing an isoprenoid-linked carbohydrate donor substrate3,4. Here we present the cryo-electron microscopy structure of yeast ALG6 at 3.0 Å resolution, which reveals a previously undescribed transmembrane protein fold. Comparison with reported GT-C structures suggests that GT-C enzymes contain a modular architecture with a conserved module and a variable module, each with distinct functional roles. We used synthetic analogues of dolichylphosphate-linked and dolichylpyrophosphate-linked sugars and enzymatic glycan extension to generate donor and acceptor substrates using purified enzymes of the ALG pathway to recapitulate the activity of ALG6 in vitro. A second cryo-electron microscopy structure of ALG6 bound to an analogue of dolichylphosphate-glucose at 3.9 Å resolution revealed the active site of the enzyme. Functional analysis of ALG6 variants identified a catalytic aspartate residue that probably acts as a general base. This residue is conserved in the GT-C superfamily. Our results define the architecture of ER-luminal GT-C enzymes and provide a structural basis for understanding their catalytic mechanisms.

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: Functional analysis of ALG6.
Fig. 2: ALG6–6AG9-Fab interactions.
Fig. 3: ALG6 structure and GT-C architecture.
Fig. 4: Donor substrate binding and catalytic site of ALG6.

Similar content being viewed by others

Data availability

Atomic coordinates of the apo-ALG6 and Dol25-P-Glc-bound ALG6 models were deposited in the RCSB PDB under accession number 6SNI for apo-ALG6 and 6SNH for Dol25-P-Glc-bound ALG6. The 3D cryo-EM maps were deposited in the Electron Microscopy Data Bank (EMDB) under accession numbers EMD-10258 for apo-ALG6 and EMD-10257 for Dol25-P-Glc-bound ALG6.

References

  1. Kornfeld, R. & Kornfeld, S. Assembly of asparagine-linked oligosaccharides. Annu. Rev. Biochem. 54, 631–664 (1985).

    CAS  PubMed  Google Scholar 

  2. Burda, P. & Aebi, M. The dolichol pathway of N-linked glycosylation. Biochim. Biophys. Acta 1426, 239–257 (1999).

    CAS  PubMed  Google Scholar 

  3. Liu, J. & Mushegian, A. Three monophyletic superfamilies account for the majority of the known glycosyltransferases. Protein Sci. 12, 1418–1431 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Moremen, K. W. & Haltiwanger, R. S. Emerging structural insights into glycosyltransferase-mediated synthesis of glycans. Nat. Chem. Biol. 15, 853–864 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Runge, K. W., Huffaker, T. C. & Robbins, P. W. Two yeast mutations in glucosylation steps of the asparagine glycosylation pathway. J. Biol. Chem. 259, 412–417 (1984).

    CAS  PubMed  Google Scholar 

  6. Reiss, G., te Heesen, S., Zimmerman, J., Robbins, P. W. & Aebi, M. Isolation of the ALG6 locus of Saccharomyces cerevisiae required for glucosylation in the N-linked glycosylation pathway. Glycobiology 6, 493–498 (1996).

    CAS  PubMed  Google Scholar 

  7. Haeuptle, M. A. & Hennet, T. Congenital disorders of glycosylation: an update on defects affecting the biosynthesis of dolichol-linked oligosaccharides. Hum. Mutat. 30, 1628–1641 (2009).

    CAS  PubMed  Google Scholar 

  8. Jaeken, J., Lefeber, D. & Matthijs, G. Clinical utility gene card for: ALG6 defective congenital disorder of glycosylation. Eur. J. Hum. Genet. 23, 1–3 (2015).

    Google Scholar 

  9. Morava, E. et al. ALG6-CDG: a recognizable phenotype with epilepsy, proximal muscle weakness, ataxia and behavioral and limb anomalies. J. Inherit. Metab. Dis. 39, 713–723 (2016).

    CAS  PubMed  Google Scholar 

  10. Ramírez, A. S. et al. Chemo-enzymatic synthesis of lipid-linked GlcNAc2Man5 oligosaccharides using recombinant Alg1, Alg2 and Alg11 proteins. Glycobiology 27, 1–8 (2017).

    Google Scholar 

  11. Ramírez, A. S. et al. Characterization of the single-subunit oligosaccharyltransferase STT3A from Trypanosoma brucei using synthetic peptides and lipid-linked oligosaccharide analogs. Glycobiology 27, 525–535 (2017).

    PubMed  PubMed Central  Google Scholar 

  12. Wild, R. et al. Structure of the yeast oligosaccharyltransferase complex gives insight into eukaryotic N-glycosylation. Science 359, 545–550 (2018).

    ADS  CAS  PubMed  Google Scholar 

  13. Fellouse, F. A. et al. High-throughput generation of synthetic antibodies from highly functional minimalist phage-displayed libraries. J. Mol. Biol. 373, 924–940 (2007).

    CAS  PubMed  Google Scholar 

  14. Gouw, M. et al. The eukaryotic linear motif resource—2018 update. Nucleic Acids Res. 46, D428–D434 (2018).

    CAS  PubMed  Google Scholar 

  15. Lombard, V., Golaconda Ramulu, H., Drula, E., Coutinho, P. M. & Henrissat, B. The carbohydrate-active enzymes database (CAZy) in 2013. Nucleic Acids Res. 42, D490–D495 (2014).

    CAS  PubMed  Google Scholar 

  16. Lizak, C., Gerber, S., Numao, S., Aebi, M. & Locher, K. P. X-ray structure of a bacterial oligosaccharyltransferase. Nature 474, 350–355 (2011).

    CAS  PubMed  Google Scholar 

  17. Matsumoto, S. et al. Crystal structures of an archaeal oligosaccharyltransferase provide insights into the catalytic cycle of N-linked protein glycosylation. Proc. Natl Acad. Sci. USA 110, 17868–17873 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bai, L., Wang, T., Zhao, G., Kovach, A. & Li, H. The atomic structure of a eukaryotic oligosaccharyltransferase complex. Nature 555, 328–333 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Petrou, V. I. et al. Structures of aminoarabinose transferase ArnT suggest a molecular basis for lipid A glycosylation. Science 351, 608–612 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bai, L., Kovach, A., You, Q., Kenny, A. & Li, H. Structure of the eukaryotic protein O-mannosyltransferase Pmt1–Pmt2 complex. Nat. Struct. Mol. Biol. 26, 704–711 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Albuquerque-Wendt, A., Hütte, H. J., Buettner, F. F. R., Routier, F. H. & Bakker, H. Membrane topological model of glycosyltransferases of the GT-C superfamily. Int. J. Mol. Sci. 20, 4842 (2019).

    PubMed Central  Google Scholar 

  22. Imbach, T. et al. A mutation in the human ortholog of the Saccharomyces cerevisiae ALG6 gene causes carbohydrate-deficient glycoprotein syndrome type-Ic. Proc. Natl Acad. Sci. USA 96, 6982–6987 (1999).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Imbach, T. et al. Multi-allelic origin of congenital disorder of glycosylation (CDG)-Ic. Hum. Genet. 106, 538–545 (2000).

    CAS  PubMed  Google Scholar 

  24. Westphal, V., Schottstädt, C., Marquardt, T. & Freeze, H. H. Analysis of multiple mutations in the hALG6 gene in a patient with congenital disorder of glycosylation Ic. Mol. Genet. Metab. 70, 219–223 (2000).

    CAS  PubMed  Google Scholar 

  25. Dercksen, M. et al. ALG6-CDG in South Africa: genotype–phenotype description of five novel patients. JIMD Rep. 8, 17–23 (2013).

    CAS  PubMed  Google Scholar 

  26. Runge, K. W. & Robbins, P. W. A new yeast mutation in the glucosylation steps of the asparagine-linked glycosylation pathway. Formation of a novel asparagine-linked oligosaccharide containing two glucose residues. J. Biol. Chem. 261, 15582–15590 (1986).

    CAS  PubMed  Google Scholar 

  27. Lee, B. C. et al. Gating mechanism of the extracellular entry to the lipid pathway in a TMEM16 scramblase. Nat. Commun. 9, 3251 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  28. Lairson, L. L., Henrissat, B., Davies, G. J. & Withers, S. G. Glycosyltransferases: structures, functions, and mechanisms. Annu. Rev. Biochem. 77, 521–555 (2008).

    CAS  PubMed  Google Scholar 

  29. Albesa-Jové, D., Cifuente, J. O., Trastoy, B. & Guerin, M. E. Quick-soaking of crystals reveals unprecedented insights into the catalytic mechanism of glycosyltransferases. Methods Enzymol. 621, 261–279 (2019).

    PubMed  Google Scholar 

  30. Notenboom, V. et al. Insights into transition state stabilization of the β-1,4-glycosidase Cex by covalent intermediate accumulation in active site mutants. Nat. Struct. Biol. 5, 812–818 (1998).

    CAS  PubMed  Google Scholar 

  31. Chang, A., Singh, S., Phillips, G. N. Jr & Thorson, J. S. Glycosyltransferase structural biology and its role in the design of catalysts for glycosylation. Curr. Opin. Biotechnol. 22, 800–808 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Sharma, C. B., Knauer, R. & Lehle, L. Biosynthesis of lipid-linked oligosaccharides in yeast: the ALG3 gene encodes the Dol-P-Man:Man5GlcNAc2-PP-Dol mannosyltransferase. Biol. Chem. 382, 321–328 (2001).

    CAS  PubMed  Google Scholar 

  33. Napiórkowska, M. et al. Molecular basis of lipid-linked oligosaccharide recognition and processing by bacterial oligosaccharyltransferase. Nat. Struct. Mol. Biol. 24, 1100–1106 (2017).

    PubMed  Google Scholar 

  34. Davies, G. J., Planas, A. & Rovira, C. Conformational analyses of the reaction coordinate of glycosidases. Acc. Chem. Res. 45, 308–316 (2012).

    CAS  PubMed  Google Scholar 

  35. Ardèvol, A. & Rovira, C. Reaction mechanisms in carbohydrate-active enzymes: glycoside hydrolases and glycosyltransferases. Insights from ab initio quantum mechanics/molecular mechanics dynamic simulations. J. Am. Chem. Soc. 137, 7528–7547 (2015).

    PubMed  Google Scholar 

  36. Aebi, M. N-linked protein glycosylation in the ER. Biochim. Biophys. Acta 1833, 2430–2437 (2013).

    CAS  PubMed  Google Scholar 

  37. Denisov, I. G., Grinkova, Y. V., Lazarides, A. A. & Sligar, S. G. Directed self-assembly of monodisperse phospholipid bilayer nanodiscs with controlled size. J. Am. Chem. Soc. 126, 3477–3487 (2004).

    CAS  PubMed  Google Scholar 

  38. Fellouse, F. A., Wiesmann, C. & Sidhu, S. S. Synthetic antibodies from a four-amino-acid code: a dominant role for tyrosine in antigen recognition. Proc. Natl Acad. Sci. USA 101, 12467–12472 (2004).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dominik, P. K. & Kossiakoff, A. A. Phage display selections for affinity reagents to membrane proteins in nanodiscs. Methods Enzymol. 557, 219–245 (2015).

    CAS  PubMed  Google Scholar 

  40. Dominik, P. K. et al. Conformational chaperones for structural studies of membrane proteins using antibody phage display with nanodiscs. Structure 24, 300–309 (2016).

    CAS  PubMed  Google Scholar 

  41. Hornsby, M. et al. A high through-put platform for recombinant antibodies to folded proteins. Mol. Cell. Proteomics 14, 2833–2847 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Borowska, M. T., Dominik, P. K., Anghel, S. A., Kossiakoff, A. A. & Keenan, R. J. A YidC-like protein in the archaeal plasma membrane. Structure 23, 1715–1724 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Hattori, M., Hibbs, R. E. & Gouaux, E. A fluorescence-detection size-exclusion chromatography-based thermostability assay for membrane protein precrystallization screening. Structure 20, 1293–1299 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Scheres, S. H. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol. 180, 519–530 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Moriarty, N. W., Grosse-Kunstleve, R. W. & Adams, P. D. Electronic Ligand Builder and Optimization Workbench (eLBOW): a tool for ligand coordinate and restraint generation. Acta Crystallogr. D Biol. Crystallogr. 65, 1074–1080 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Afonine, P. V. et al. New tools for the analysis and validation of cryo-EM maps and atomic models. Acta Crystallogr. D Struct. Biol. 74, 814–840 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Kucukelbir, A., Sigworth, F. J. & Tagare, H. D. Quantifying the local resolution of cryo-EM density maps. Nat. Methods 11, 63–65 (2014).

    CAS  PubMed  Google Scholar 

  53. The PyMOL Molecular Graphics System, Version 2.0 Schrödinger, LLC.

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

    CAS  PubMed  Google Scholar 

  55. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    CAS  PubMed  Google Scholar 

  56. Thompson, J. D., Higgins, D. G. & Gibson, T. J. CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res. 22, 4673–4680 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Sievers, F. & Higgins, D. G. Clustal Omega for making accurate alignments of many protein sequences. Protein Sci. 27, 135–145 (2018).

    CAS  PubMed  Google Scholar 

  58. Needleman, S. B. & Wunsch, C. D. A general method applicable to the search for similarities in the amino acid sequence of two proteins. J. Mol. Biol. 48, 443–453 (1970).

    CAS  PubMed  Google Scholar 

  59. Sabesan, S. & Neira, S. Synthesis of glycosyl phosphates and azides. Carbohydr. Res. 223, 169–185 (1992).

    CAS  Google Scholar 

  60. Maunier, V., Boullanger, P., Lafont, D. & Chevalier, Y. Synthesis and surface-active properties of amphiphilic 6-aminocarbonyl derivatives of d-glucose. Carbohydr. Res. 299, 49–57 (1997).

    CAS  Google Scholar 

  61. Williams, R. J. et al. Combined inhibitor free-energy landscape and structural analysis reports on the mannosidase conformational coordinate. Angew. Chem. Int. Edn Engl. 53, 1087–1091 (2014).

    CAS  Google Scholar 

  62. Malet, C. & Hindsgaul, O. Versatile functionalization of carbohydrate hydroxyl groups through their O-cyanomethyl ethers. J. Org. Chem. 61, 4649–4654 (1996).

    CAS  PubMed  Google Scholar 

  63. Li, T., Tikad, A., Pan, W. & Vincent, S. P. β-Stereoselective phosphorylations applied to the synthesis of ADP- and polyprenyl-β-mannopyranosides. Org. Lett. 16, 5628–5631 (2014).

    CAS  PubMed  Google Scholar 

  64. Li, S. T. et al. Reconstitution of the lipid-linked oligosaccharide pathway for assembly of high-mannose N-glycans. Nat. Commun. 10, 1813 (2019).

    ADS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by the Swiss National Science Foundation (SNF) Sinergia programmes TransGlyco (CRSII3_147632) and GlycoStart (CRSII5_173709) to M.A., J.-L.R. and K.P.L, SNF grant 310030B_166672 to K.P.L., as well as by the US National Institutes of Health grant GM117372 to A.A.K. Cryo-EM data were collected at the ScopeM facility at ETH Zürich. We thank the staff of ScopeM for technical support; J. Zürcher for technical support with protein expression and purification; J. Kowal and I. Manolaridis for help with EM data collection; and A. Ramirez and A. Alam for helpful discussions.

Author information

Authors and Affiliations

Authors

Contributions

K.P.L. and J.S.B. conceived the project. J.S.B. cloned, screened, expressed and purified proteins, reconstituted ALG6 into lipid nanodiscs, performed biochemical experiments and produced Dol25-PP-GlcNAc2Man9 using synthetic precursor substrates. G.P. synthesized Dol25-P-Glc and derivatives. J.B. synthesized Dol25-P-Man and Dol25-PP-GlcNAc2. K.N. and J.S.B. performed synthetic antibody generation. J.S.B. prepared the grids. R.N.I. collected cryo-EM data. J.S.B. assisted cryo-EM data collection, processed data, built the ALG6 models and refined the structures. T.D. and J.-L.R. supervised the synthesis of LLO substrates. A.A.K. provided the chaperone-assisted structure determination phage display pipeline and supervised synthetic antibody generation. M.A. analysed the data and contributed to writing the paper. J.S.B. and K.P.L. analysed the data and wrote the paper.

Corresponding author

Correspondence to Kaspar P. Locher.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Xiaochen Bai 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 ALG6 activity in distinct lipidic environments and chemo-enzymatic synthesis of LLO intermediates.

a, Tricine gel-based analysis of the ALG6-catalysed reaction using Dol25-PP-GlcNAc2Man9 and Dol25-P-Glc substrates. The reactions were carried out in distinct detergents or with ALG6 reconstituted in lipid nanodiscs, as indicated above the gel lanes. The glycans were analysed as described in Fig. 1b. b, Tricine gel-based analysis of ALG reaction cascade intermediates, with reactions analysed as described in Fig. 1b. The structures of glycopeptides are schematically shown above the lanes and respective glycosyltransferase enzymes are indicated above the arrows.

Extended Data Fig. 2 Data processing and structure determination of the substrate-free ALG6–6AG9-Fab complex in lipid nanodiscs.

a, Overview of the EM data processing and structure determination pipeline using RELION 3.046. b, Representative cryo-EM micrograph. c, Representative 2D classes. d, A representative 2D class with 6AG9-Fab and the nanodisc belt highlighted. e, Spatial distribution of particles in the final iteration of 3D refinement. f, Refined and B-factor-sharpened EM map, coloured by local resolution estimation as calculated in ResMap52. g, Resolution estimation of the final map via Fourier shell correlation (FSC), as calculated in RELION 3.0. h, The FSC between model and map, as calculated in PHENIX mtriage51.

Extended Data Fig. 3 Molecular interactions at the interface of ALG6 and 6AG9-Fab.

a, Surface representation of ALG6 in light brown and cartoon representation of the variable fragment of 6AG9-Fab coloured red (heavy chain) and black (light chain). The five complementarity-determining region (CDR) loops of 6AG9-Fab interacting with ALG6 are shown in sphere representation and make contact mostly with the ER-luminal ‘arch’ formed by EL4 of ALG6. b, Detailed view of the individual CDR:ALG6 interactions, with 6AG9-Fab CDRs and ALG6 in stick representation and coloured as in a. The CDR sequences are indicated above the panels. Note that CDR1 of the light chain (LC) was not randomized. HC, heavy chain.

Extended Data Fig. 4 Substrate and lipid interactions of ALG6.

a, Distinct lipid binding sites were identified in the structure of nanodisc-reconstituted ALG6, shown in light brown surface representation. Ordered phospholipids and cholesteryl-hemisuccinate molecules are shown in stick representation (green), and the EM density map was contoured at 5 r.m.s.d. and carved to 1.6 Å. b, Electrostatic surface potential of ALG6, with negative charges coloured in red, neutral charges in white, positive charges in blue and bound Dol25-P-Glc depicted in stick representation with carbons in green. The inset shows a zoomed-in view of the kink induced in the dolichol moiety. c, Electrostatic surface potential of ALG6 shown from four angles to indicate potential binding sites for lipid-linked substrates of ALG6 based on the presence of suitable surface cavities. The red solid arrow indicates the binding site of the observed donor substrate Dol25-P-Glc. The red dashed arrows indicate potential binding sites of the acceptor substrate Dol-PP-GlcNAc2Man9. These sites were chosen based on groove-like features in the transmembrane region that might bind dolichol and positively charged patches at the ER-luminal membrane boundary that could help to bind the pyrophosphate moiety of the acceptor LLO. Binding to these sites would allow the terminal mannose of the A-branch of the acceptor substrate to reach the active site. d, Membrane deformation by ALG6 shown from distinct views. The EM map of the apo-ALG6 complex with the 6AG9-Fab complex in a lipid nanodisc is coloured yellow for ALG6, red (heavy chain) and black (light chain) for 6AG9-Fab, and transparent grey for density indicating the lipid nanodisc. Note that the lipid bilayer of the nanodisc surrounding the ALG6 protein is twisted and thinner in the region opposite the arch formed by EL4.

Extended Data Fig. 5 Structural similarity of the conserved module of GT-C enzymes.

a, Cα traces of the first seven transmembrane helices and the first external loop EL1 of selected structures are shown following superposition. These following GT-C family members of known structure were used, with the GT family number indicated: S. cerevisiae (S.c.) PMT1 (GT39, PDB ID 6P25), S.c. ALG6 (GT57, this study), C. lari (C.l.) PglB (GT66, PDB ID 3RCE), A. fulgidus (A.f.) AglB (GT66, PDB ID 3WAJ), S.c. STT3 (GT66, PDB ID 6EZN) and C. metallidurans ArnT (GT83, PDB ID 5F15). b, Close-up view of EL-h1, EL-h2 and the connecting loop of GT-Cs where the catalytic role of the conserved aspartate, shown in stick representation, was experimentally demonstrated.

Extended Data Fig. 6 Residues involved in CDGs caused by ALG6 mutations.

The structure of yeast ALG6 is depicted as a Cα trace and as a transparent surface representation coloured in light brown. The surface is coloured green for residues conserved between yeast and human ALG6. Cα atoms of residues that have been shown to cause CDGs in the human homologue of ALG6 are shown in sphere representation. Residue numbers in brackets indicate the corresponding residues of human ALG6 according to pairwise protein sequence alignment with EMBOSS needle58.

Extended Data Fig. 7 Sequence alignment of ALG6 homologues and orthologues.

a, Alignment of amino acid sequences of yeast and human ALG6 and ALG8 proteins, generated with Clustal Omega57 (Uniprot identifiers: Q12001, Q9Y672, P40351 and Q9BVK2). Secondary structure elements of yeast ALG6 are depicted and labelled above the sequence. The dashed lines indicate regions that are disordered in the ALG6 structures. Cytosolic regions are labelled ‘cyto’ and ER-luminal regions are labelled ‘ER-lumen’. The red dot indicates the catalytic base Asp69, which is a structurally conserved aspartate residue that has been observed or proposed to be the catalytic base in all GT-C enzymes of known structures. The black dots indicate every tenth amino acid in the sequence of S. cerevisiae ALG6. b, Surface representation of yeast ALG6 (light brown), with residues identical to human ALG6 highlighted in green (left) and residues identical to yeast ALG8 highlighted in blue (right).

Extended Data Fig. 8 Data processing and structure determination of the substrate-bound ALG6–6AG9-Fab complex in detergent solution.

a, Overview of the EM data processing and structure determination pipeline using RELION 3.046. b, A representative cryo-EM micrograph. c, Representative 2D classes. d, Spatial distribution of particles in the final iteration of 3D refinement. e, Resolution estimation of the final map via FSC, as calculated in RELION 3.0. f, The final refined and B-factor-sharpened map, coloured by local resolution estimation, as calculated in ResMap52. g, The FSC between model and map, as calculated in PHENIX mtriage51.

Extended Data Fig. 9 Mechanism of ALG6-catalysed glucosyl transfer.

a, Chemical structures of synthetic donor substrate analogues (left) and their functional analysis (right). Compound numbers are indicated in bold and in parentheses. Analysis of ALG6 activity as described in Fig. 1b but in the presence of different substrate analogues as indicated above the lanes. The lane labelled Man9-LLO is a control sample for size comparison. In one of the ALG6 reactions, a pre-incubation of ALG6 with 10 mM EDTA was used to remove any divalent ions from the solution. b, Proposed three-state mechanism of ALG6; intermediate states are numbered and encircled. State 1 represents the apo state of ALG6 based on our EM structure, with ALG6 in surface representation (brown) and clipped for better visualization of the substrate-binding pocket. State 2 represents ALG6 bound to a donor substrate based on our EM structure, with Dol25-P-Glc shown in sphere representation with carbon atoms coloured green and oxygen atoms coloured red. State 3 represents a putative ternary complex based on the apo structure of ALG6, with substrates drawn manually. The inset depicts the proposed reaction mechanism, with Asp69 acting as a general base that deprotonates the C3 hydroxyl group of the attacking mannose moiety.

Extended Data Table 1 Cryo-EM data collection, refinement and validation statisticsa as defined in PHENIX49

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-6 and Supplementary Methods

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bloch, J.S., Pesciullesi, G., Boilevin, J. et al. Structure and mechanism of the ER-based glucosyltransferase ALG6. Nature 579, 443–447 (2020). https://doi.org/10.1038/s41586-020-2044-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2044-z

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