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:

Target protein deglycosylation in living cells by a nanobody-fused split O-GlcNAcase

Abstract

O-linked N-acetylglucosamine (O-GlcNAc) is an essential and dynamic post-translational modification that is presented on thousands of nucleocytoplasmic proteins. Interrogating the role of O-GlcNAc on a single target protein is crucial, yet challenging to perform in cells. Herein, we developed a nanobody-fused split O-GlcNAcase (OGA) as an O-GlcNAc eraser for selective deglycosylation of a target protein in cells. After systematic cellular optimization, we identified a split OGA with reduced inherent deglycosidase activity that selectively removed O-GlcNAc from the desired target protein when directed by a nanobody. We demonstrate the generality of the nanobody-fused split OGA using four nanobodies against five target proteins and use the system to study the impact of O-GlcNAc on the transcription factors c-Jun and c-Fos. The nanobody-directed O-GlcNAc eraser provides a new strategy for the functional evaluation and engineering of O-GlcNAc via the selective removal of O-GlcNAc from individual proteins directly in cells.

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: Design and development of a nanobody-directed split OGA for O-GlcNAc removal in a protein-selective manner.
Fig. 2: Design and optimization of nanobody-fused split OGA for protein-selective deglycosylation on GFP–Nup62.
Fig. 3: The nanobody-fused split OGA is generalizable for protein-selective deglycosylation on various O-GlcNAcylated proteins.
Fig. 4: Multiple nanobody–tag pairs are applicable for protein-selective deglycosylation.
Fig. 5: Nanobody-fused split OGA facilitates the functional attribution of O-GlcNAc on c-Jun to protein stability and on c-Fos to AP-1 transcriptional activity.

Similar content being viewed by others

Data availability

The main data generated or analyzed during this study are included in this published article and its Supplementary Information. The MS proteomics data were searched against the UniProt/SwissProt human (Homo sapiens) protein database (19 August 2016; http://www.uniprot.org/proteomes/UP000005640) and have been deposited to the ProteomeXchange Consortium via the PRIDE51 partner repository with the dataset identifiers PXD022347 (Fig. 3d,e) and PXD018914 (Fig. 3f,g). Source data are provided with this paper.

Code availability

Code used to analyze the MS proteomics data has been deposited in GitHub and is available for download at https://github.com/harvardinformatics/quantproteomics/tree/master/PEA.

References

  1. Yang, X. & Qian, K. Protein O-GlcNAcylation: emerging mechanisms and functions. Nat. Rev. Mol. Cell Biol. 18, 452–465 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Levine, Z. G. & Walker, S. The biochemistry of O-GlcNAc transferase: which functions make it essential in mammalian cells? Annu. Rev. Biochem. 85, 631–657 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Alonso, J., Schimpl, M. & van Aalten, D. M. O-GlcNAcase: promiscuous hexosaminidase or key regulator of O-GlcNAc signaling? J. Biol. Chem. 289, 34433–34439 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Yuzwa, S. A. & Vocadlo, D. J. O-GlcNAc and neurodegeneration: biochemical mechanisms and potential roles in Alzheimer’s disease and beyond. Chem. Soc. Rev. 43, 6839–6858 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Ma, J. & Hart, G. W. Protein O-GlcNAcylation in diabetes and diabetic complications. Expert Rev. Proteom. 10, 365–380 (2013).

    Article  CAS  Google Scholar 

  6. Slawson, C. & Hart, G. W. O-GlcNAc signalling: implications for cancer cell biology. Nat. Rev. Cancer 11, 678–684 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hart, G. W. Nutrient regulation of signaling and transcription. J. Biol. Chem. 294, 2211–2231 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gorelik, A. & van Aalten, D. M. F. Tools for functional dissection of site-specific O-GlcNAcylation. RSC Chem. Biol. 1, 98–109 (2020).

    Article  CAS  Google Scholar 

  9. Martin, S. E. S. et al. Structure-based evolution of low nanomolar O-GlcNAc transferase inhibitors. J. Am. Chem. Soc. 140, 13542–13545 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhang, Z., Tan, E. P., VandenHull, N. J., Peterson, K. R. & Slawson, C. O-GlcNAcase expression is sensitive to changes in O-GlcNAc homeostasis. Front. Endocrinol. 5, 206 (2014).

    Article  Google Scholar 

  11. Yuzwa, S. A. et al. A potent mechanism-inspired O-GlcNAcase inhibitor that blocks phosphorylation of tau in vivo. Nat. Chem. Biol. 4, 483–490 (2008).

    Article  CAS  PubMed  Google Scholar 

  12. Alfaro, J. F. et al. Tandem mass spectrometry identifies many mouse brain O-GlcNAcylated proteins including EGF domain-specific O-GlcNAc transferase targets. Proc. Natl Acad. Sci. USA 109, 7280–7285 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. Vosseller, K. et al. O-linked N-acetylglucosamine proteomics of postsynaptic density preparations using lectin weak affinity chromatography and mass spectrometry. Mol. Cell Proteom. 5, 923–934 (2006).

    Article  CAS  Google Scholar 

  14. Woo, C. M., Iavarone, A. T., Spiciarich, D. R., Palaniappan, K. K. & Bertozzi, C. R. Isotope-targeted glycoproteomics (IsoTaG): a mass-independent platform for intact N- and O-glycopeptide discovery and analysis. Nat. Methods 12, 561–567 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gorelik, A. et al. Genetic recoding to dissect the roles of site-specific protein O-GlcNAcylation. Nat. Struct. Mol. Biol. 26, 1071–1077 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hart, G. W., Slawson, C., Ramirez-Correa, G. & Lagerlof, O. Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease. Annu. Rev. Biochem. 80, 825–858 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ruan, H. B., Nie, Y. & Yang, X. Regulation of protein degradation by O-GlcNAcylation: crosstalk with ubiquitination. Mol. Cell Proteom. 12, 3489–3497 (2013).

    Article  CAS  Google Scholar 

  18. Ramirez, D. H. et al. Engineering a proximity-directed O-GlcNAc transferase for selective protein O-GlcNAcylation in cells. ACS Chem. Biol. 15, 1059–1066 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ingram, J. R., Schmidt, F. I. & Ploegh, H. L. Exploiting nanobodies’ singular traits. Annu. Rev. Immunol. 36, 695–715 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Li, B., Li, H., Lu, L. & Jiang, J. Structures of human O-GlcNAcase and its complexes reveal a new substrate recognition mode. Nat. Struct. Mol. Biol. 24, 362–369 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Roth, C. et al. Structural and functional insight into human O-GlcNAcase. Nat. Chem. Biol. 13, 610–612 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Elsen, N. L. et al. Insights into activity and inhibition from the crystal structure of human O-GlcNAcase. Nat. Chem. Biol. 13, 613–615 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Gao, Y., Wells, L., Comer, F. I., Parker, G. J. & Hart, G. W. Dynamic O-glycosylation of nuclear and cytosolic proteins: cloning and characterization of a neutral, cytosolic β-N-acetylglucosaminidase from human brain. J. Biol. Chem. 276, 9838–9845 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Rexach, J. E. et al. Quantification of O-glycosylation stoichiometry and dynamics using resolvable mass tags. Nat. Chem. Biol. 6, 645–651 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kirchhofer, A. et al. Modulation of protein properties in living cells using nanobodies. Nat. Struct. Mol. Biol. 17, 133–138 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Butkinaree, C. et al. Characterization of β-N-acetylglucosaminidase cleavage by caspase-3 during apoptosis. J. Biol. Chem. 283, 23557–23566 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cetinbas, N., Macauley, M. S., Stubbs, K. A., Drapala, R. & Vocadlo, D. J. Identification of Asp174 and Asp175 as the key catalytic residues of human O-GlcNAcaseby functional analysis of site-directed mutants. Biochemistry 45, 3835–3844 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Woo, C. M. et al. Mapping and quantification of over 2000 O-linked glycopeptides in activated human T cells with isotope-targeted glycoproteomics (Isotag). Mol. Cell Proteom. 17, 764–775 (2018).

    Article  CAS  Google Scholar 

  29. De Genst, E. J. et al. Structure and properties of a complex of α-synuclein and a single-domain camelid antibody. J. Mol. Biol. 402, 326–343 (2010).

    Article  PubMed  Google Scholar 

  30. Ling, J. et al. A nanobody that recognizes a 14-residue peptide epitope in the E2 ubiquitin-conjugating enzyme UBC6e modulates its activity. Mol. Immunol. 114, 513–523 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Traenkle, B. et al. Monitoring interactions and dynamics of endogenous β-catenin with intracellular nanobodies in living cells. Mol. Cell Proteom. 14, 707–723 (2015).

    Article  CAS  Google Scholar 

  32. Hess, J., Angel, P. & Schorpp-Kistner, M. AP-1 subunits: quarrel and harmony among siblings. J. Cell Sci. 117, 5965–5973 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Tai, H. C., Khidekel, N., Ficarro, S. B., Peters, E. C. & Hsieh-Wilson, L. C. Parallel identification of O-GlcNAc-modified proteins from cell lysates. J. Am. Chem. Soc. 126, 10500–10501 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Kim, S., Maynard, J. C., Strickland, A., Burlingame, A. L. & Milbrandt, J. Schwann cell O-GlcNAcylation promotes peripheral nerve remyelination via attenuation of the AP-1 transcription factor JUN. Proc. Natl Acad. Sci. USA 115, 8019–8024 (2018).

    Article  PubMed  Google Scholar 

  35. Qiao, Y. et al. High glucose stimulates tumorigenesis in hepatocellular carcinoma cells through AGER-dependent O-GlcNAcylation of c-Jun. Diabetes 65, 619–632 (2016).

    Article  CAS  PubMed  Google Scholar 

  36. Zhang, F. et al. O-GlcNAc modification is an endogenous inhibitor of the proteasome. Cell 115, 715–725 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Ranuncolo, S. M., Ghosh, S., Hanover, J. A., Hart, G. W. & Lewis, B. A. Evidence of the involvement of O-GlcNAc-modified human RNA polymerase II CTD in transcription in vitro and in vivo. J. Biol. Chem. 287, 23549–23561 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hardiville, S. et al. TATA-box binding protein O-GlcNAcylation at T114 regulates formation of the B-TFIID complex and is critical for metabolic gene regulation. Mol. Cell 77, 1143–1152 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Wu, T. et al. Targeted protein degradation as a powerful research tool in basic biology and drug target discovery. Nat. Struct. Mol. Biol. 27, 605–614 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Siriwardena, S. U. et al. Phosphorylation-inducing chimeric small molecules. J. Am. Chem. Soc. 142, 14052–14057 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Dong, J. X. et al. A toolbox of nanobodies developed and validated for use as intrabodies and nanoscale immunolabels in mammalian brain neurons. eLife 8, e48750 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Koch, B. et al. Generation and validation of homozygous fluorescent knock-in cells using CRISPR–Cas9 genome editing. Nat. Protoc. 13, 1465–1487 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kuey, C., Larocque, G., Clarke, N. I. & Royle, S. J. Unintended perturbation of protein function using GFP nanobodies in human cells. J. Cell Sci. 132, jcs234955 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Fridy, P. C. et al. A robust pipeline for rapid production of versatile nanobody repertoires. Nat. Methods 11, 1253–1260 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Farrants, H. et al. Chemogenetic control of nanobodies. Nat. Methods 17, 279–282 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Gil, A. A. et al. Optogenetic control of protein binding using light-switchable nanobodies. Nat. Commun. 11, 4044 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhu, Y. et al. O-GlcNAc occurs cotranslationally to stabilize nascent polypeptide chains. Nat. Chem. Biol. 11, 319–325 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Thompson, J. W., Griffin, M. E. & Hsieh-Wilson, L. C. Methods for the detection, study, and dynamic profiling of O-GlcNAc glycosylation. Methods Enzymol. 598, 101–135 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Woo, C. M. & Bertozzi, C. R. Isotope targeted glycoproteomics (IsoTaG) to characterize intact, metabolically labeled glycopeptides from complex proteomes. Curr. Protoc. Chem. Biol. 8, 59–82 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Huber, W., von Heydebreck, A., Sultmann, H., Poustka, A. & Vingron, M. Variance stabilization applied to microarray data calibration and to the quantification of differential expression. Bioinformatics 18, S96–S104 (2002).

    Article  PubMed  Google Scholar 

  51. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank P. Schwein and Z. Lin for helpful discussions and B. Budnik (principal scientist of the Harvard University Proteomics Facility). Support from the National Institutes of Health (U01CA242098-01, C.M.W.), the Burroughs Wellcome Fund, a Career Award at the Scientific Interface (C.M.W.), the Sloan Foundation (C.M.W.), the Merck Fellowship Fund and Harvard University is gratefully acknowledged.

Author information

Authors and Affiliations

Authors

Contributions

Y.G. and C.M.W. conceived the project. Y.G., D.H.R. and C.M.W. designed the experiments. Y.G. and S.W. performed the experiments. B.Y. and A.K.D. helped with quantitative MS analyses. C.A. contributed reagents and technical advice on confocal imaging analysis. Y.G. and C.M.W. analyzed the data and wrote the paper with input from all authors.

Corresponding author

Correspondence to Christina M. Woo.

Ethics declarations

Competing interests

Harvard University has filed a patent application (US Provisional Application No. 63/087,773, filed 5 October 2020) including work described herein. C.M.W., Y.G. and D.H.R. are inventors of this patent. All other 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.

Peer review information Nature Chemical Biology thanks Jing Li and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Extended data

Extended Data Fig. 1 Schematic representation of OGA and target protein constructs used in this study.

a, Schematic of the structures of human OGA and other truncations. Catalytic domain, stalk domain, HAT domain, and intrinsic disordered regions are shown in pink, cyan, orange and white, respectively. GS linker represents a 15-residue glycine and serine linker. b, Depiction of the strategy to fuse the nanobody on OGAs to achieve protein specificity. nGFP, nanobody against GFP. c, Design of GFP-fused, Ubc tag-fused and BC2 tag-fused proteins of interest used in this study. For GFP-fused proteins, GFP and a Flag tag are placed on the N-terminus and the EPEA tag is in the C-terminus unless otherwise noted. For Ubc tag-fused proteins, the 14-residue peptide tag, Flag tag and EPEA tag are sequentially placed in the C-terminus unless otherwise noted. For BC2 tag-fused proteins, the 12-residue peptide tag is placed on the N-terminus and Flag, EPEA tag are in the C-terminus. Peptide sequences of Ubc and BC2 were shown. d, Symbols used in this manuscript to represent the indicated split OGA constructs.

Source data

Extended Data Fig. 2 Identification of the minimal OGA for nanobody-directed deglycosylation on the target protein.

a, Enzymatic activities of OGA and its truncations are evaluated on GFP-Nup62. GFP-Nup62 was coexpressed with indicated constructs, enriched by anti-EPEA beads, and analyzed by immunoblotting to visualize the protein level and O-GlcNAc modification level, respectively. b, Evaluation of enzymatic activities of nGFP-OGA fusion proteins on GFP-Nup62. Expression levels of the indicated proteins and degree of O-GlcNAc modification were quantified by immunoblotting. The ratio equals to the intensity of anti-O-GlcNAc immunoblot normalized by the intensity of anti-Flag immunoblot. WCL, whole-cell lysate. The data are representative of two biological replicates.

Source data

Extended Data Fig. 3 nGFP-OGA(GS-∆HAT) has limited target protein selectivity and can alter subcellular localization of the target protein.

a, nGFP-OGA(GS-∆HAT) removes O-GlcNAc from Nup62 without GFP tag similar to the full length OGA (fl-OGA). HEK 293T whole cell lysates (WCL) and immunoprecipitation samples were analyzed by immunoblotting assays using the indicated antibodies. Results are representative of two biological replicates. b, nGFP colocalizes with nuclear transcription factor Sp1 with GFP and does not change the subcellular localization of GFP-Sp1 by immunofluorescence imaging. c, nGFP-OGA(GS-∆HAT) alters the subcellular localization of GFP-Sp1, but coexpression with fl-OGA does not change nuclear localization of GFP-Sp1 by immunofluorescence imaging. Channels are annotated on the top. Scale bar: 10 µm. Right: merged channel. Proteins coexpressed in each sample were labeled on the left side. Images are representative of at least three randomly selected frames.

Source data

Extended Data Fig. 4 Optimization of nGFP-fused split OGA constructs in living cells.

a, Coexpressing N- and C-fragments of OGA reconstitutes deglycosidase activity in HEK 293T cells. b, Split OGA fragments, N2 and C3, instead of C4, associate with each other when coexpressed in HEK 293T cells. The asterisk indicated IgG heavy chain from anti-c-Myc magnetic beads. c, Comparison of nGFP-fused N- and C-terminal OGA fragments on GFP-Nup62 in HEK 293T cells. The pair of N2 and nGFP-fused C3 (N2 + nGFP-C3) shows the best deglycosylation performance. In a and c, activities of fragments alone or pairs with/without nGFP were evaluated on GFP-Nup62, which was enriched by beads against EPEA tag and blotted with RL2 antibody to reveal O-GlcNAc modification level. D174N, a catalytically impaired mutation on OGA. Anti-myc and anti-HA blots detect expression of full-length (fl-OGA) or N-terminal fragment, and C-terminal fragment, respectively. WCL, whole-cell lysates. The data in a-c are representative of at least two biological replicates.

Source data

Extended Data Fig. 5 nGFP-splitOGA selectively deglycosylates the target protein without affecting the global O-GlcNAc proteome.

a, nGFP-splitOGA has little effect on endogenous glycoprotein CREB. HEK 293T cells coexpressing OGA constructs with GFP-Nup62 were subjected to mass shift assay. The intensities of O-GlcNAcylated and unmodified CREB were quantified. The ratios are shown below the anti-CREB blot. WCL, whole-cell lysates. b, Overexpression of selected split OGA constructs with target protein has little effect on global O-GlcNAcylation level. For OGT inhibition, cells were treated with 25 µM OSMI-4b for 30 h. Global O-GlcNAcylation level was evaluated by anti-O-GlcNAc (RL2) antibody. c, nGFP-splitOGA has minimal effect on protein levels of endogenous OGT, OGA and glycoprotein CREB. Anti-myc and anti-HA blots detect expression of N-terminal fragment, and C-terminal fragment, respectively. d, Comparison of overexpressed proteins with the corresponding endogenous proteins. The antibody against OGA recognizes both endogenous OGA and the overexpressed N-terminal fragment of split OGA. Endogenous Nup62(*) and OGA (**) are indicated. The data in a-d are representative of at least two biological replicates.

Source data

Extended Data Fig. 6 Confocal imaging of intracellular distributions of GFP-Sp1 and the split OGAs in HEK 293T cells.

a, GFP-Sp1 localized in nucleus. b, Intracellular distributions of N2 and nGFP-C3 fragments when coexpressed in HEK 293T cells. Two fragments of nGFP-splitOGA were distributed on both cytoplasm and nucleus. c, Subcellular localizations of GFP-Sp1, N fragment and C fragment when expressed simultaneously in HEK 293T cells. Two fragments of split OGA without nGFP (c, upper row) were distributed on both cytoplasm and nucleus. C-terminal fragment of nGFP-splitOGA (c, bottom row) reveals better colocalization with nuclear protein GFP-Sp1, showing the binding between nGFP and GFP. Split OGAs do not change the subcellular localization of GFP-Sp1. Channels are annotated on the top. Scale bar: 10 µm. Right: merged channel. Proteins coexpressed in each sample were labeled on the left side. Images are representative of at least three randomly selected frames.

Source data

Extended Data Fig. 7 Mass spectrometry analysis on the activity and selectivity of nGFP-splitOGA on GFP-Nup62.

a, Schematic representation of the workflow of O-GlcNAcylated protein enrichment and mass spectrometry-based identification. Proteins with O-GlcNAc modification were labeled with GalNAz by GalT(Y289L)-mediated chemoenzymatic labeling, followed by a click reaction with an alkyne-biotin probe. Biotin-labeled proteome was enriched by streptavidin beads and digested by trypsin. Released peptides were labeled by TMT reagents and compiled into a single pool. Proteins were identified and quantified by LC-MS. bd, Reproducibility of the TMT experiments of O-GlcNAcylated proteome shown in Fig. 3d,e. The signal abundances of the corresponding TMT channels for each protein were extracted and were log10 transformed for full-length OGA treatment (b, fl-OGA), nGFP-splitOGA treatment (c) and its inactive form [N2(D174N) + nGFP-C3] treatment (d) groups (n = 2 independent biological replicates).

Source data

Extended Data Fig. 8 Peptide tag BC2 and its nanobody can be adapted to split OGA to achieve protein-selective deglycosylation.

a, Schematic of nanobodies against BC2 and EPEA tag adapted to split OGA. BC2 tag refers to a 12-residue peptide epitope, which is functional irrespective of its position on the target protein. b, nBC2-splitOGA is able to remove O-GlcNAc from Nup62 tagged with BC2 and EPEA in a similar manner to nEPEA-splitOGA. Symbols represent the corresponding OGA constructs as indicated in Extended Data Fig. 1. Anti-myc and anti-HA blots detect expression of full-length (fl-OGA) or N-terminal and C-terminal fragment, respectively. WCL, whole-cell lysate. The data are representative of two biological replicates.

Source data

Extended Data Fig. 9 Modulation of O-GlcNAc modification level and validation of its functional contribution on the stability of GFP-c-Jun.

a, O-GlcNAc level on GFP-c-Jun and endogenous CREB were evaluated by the mass-shift assay. GFP-c-Jun was coexpressed with indicated OGA constructs. The intensities of O-GlcNAcylated and unmodified c-Jun and CREB were quantified. Quantification is shown as mean ± s.d. of n = 3 independent biological experiments. All ratios were normalized by the Blank samples. Unpaired two-tailed Student’s t tests were used for statistical analysis. n.s., not significant. b, Whole cell lysates (WCL) were analyzed by immunoblotting assays using the indicated antibodies. Anti-myc and anti-HA blots detect expression of full-length (fl-OGA) or N-terminal fragment, and C-terminal fragment, respectively. The data in a and b are representative of at least three biological replicates. c, The stability of GFP-c-Jun was enhanced by OGA inhibition (Thiamet-G treatment) and impeded by OGT inhibition (Ac45SGlcNAc treatment). HEK 293T cells expressing GFP-c-Jun pre-treated with DMSO or Ac45SGlcNAc or Thiamet-G were incubated with 50 μM CHX for up to 12 h, during which the protein level of GFP-c-Jun and global O-GlcNAcylation level were monitored. Results in c are representative of two biological replicates.

Source data

Extended Data Fig. 10 Modulation of O-GlcNAc modification level with nUbc-splitOGA on c-Fos-Ubc in comparison to OGT inhibition.

Immunoblotting analysis of protein expression and O-GlcNAcylation status of c-Fos-Ubc and endogenous c-Jun under the indicated treatments corresponding to Fig. 5d,e. by either enrichment against EPEA-tag (a) or chemoenzymatic labeling followed with Biotin-IP (b). Endogenous c-Jun shows negligible changes on O-GlcNAcylation status with the coexpression of nUbc-splitOGA but shows reduced O-GlcNAc modification upon OGT inhibition with OSMI-4b. No detectable endogenous c-Fos was observed in HEK 293T cells. The data in a and b are representative of two biological replicates.

Supplementary information

Supplementary Information

Supplementary Figs. 1–20 and Table 1.

Reporting Summary

Supplementary Data

Statistical source data for Supplementary Fig. 4.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 6

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 7

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 8

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 9

Unprocessed western blots and/or gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ge, Y., Ramirez, D.H., Yang, B. et al. Target protein deglycosylation in living cells by a nanobody-fused split O-GlcNAcase. Nat Chem Biol 17, 593–600 (2021). https://doi.org/10.1038/s41589-021-00757-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00757-y

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