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:

Biotinylation by antibody recognition—a method for proximity labeling

An Addendum to this article was published on 27 July 2018

This article has been updated

Abstract

The high-throughput detection of organelle composition and proteomic mapping of protein environment directly from primary tissue as well as the identification of interactors of insoluble proteins that form higher-order structures have remained challenges in biological research. We report a proximity-based labeling approach that uses an antibody to a target antigen to guide biotin deposition onto adjacent proteins in fixed cells and primary tissues, which allows proteins in close proximity to the target antigen to be captured and identified by mass spectrometry. We demonstrated the specificity and sensitivity of our method by examining the well-studied mitochondrial matrix. We then used the method to profile the dynamic interactome of lamin A/C in multiple cell and tissue types under various treatment conditions. The ability to detect proximal proteins and putative interactors in intact tissues, and to quantify changes caused by different conditions or in the presence of disease mutations, can provide a window into cell biology and disease pathogenesis.

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: Biotinylation by antibody recognition.
Figure 2: Identification of NE proteins in primary human tissue.
Figure 3: Differential proteomics.

Similar content being viewed by others

Change history

  • 27 July 2018

    In the version of this article initially published, the introduction focused on methods for characterizing the nuclear envelope and did not include a comprehensive overview of proximity-based methods, some of which have similarly utilized antibody-conjugated peroxidase for proximity labeling by biotin deposition. The authors apologize for the omission of the following references: Kotani, N. et al., Proc. Natl. Acad. Sci. USA 105, 7405–7409 (2008); Hashimoto, N. et al., Proteomics 12, 3154–3163 (2012); Li, X. W. et al., J. Biol. Chem. 289, 14434–14447 (2014); and Rees, J. S. et al., Curr. Protoc. Protein Sci. 80, 19.27.1–19.27.18 (2015).

References

  1. Roux, K.J., Kim, D.I., Raida, M. & Burke, B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J. Cell Biol. 196, 801–810 (2012).

    Article  CAS  Google Scholar 

  2. Kubben, N. et al. Identification of differential protein interactors of lamin A and progerin. Nucleus 1, 513–525 (2010).

    Article  Google Scholar 

  3. Dittmer, T.A. et al. Systematic identification of pathological lamin A interactors. Mol. Biol. Cell 25, 1493–1510 (2014).

    Article  Google Scholar 

  4. Depreux, F.F. et al. Disruption of the lamin A and matrin-3 interaction by myopathic LMNA mutations. Hum. Mol. Genet. 24, 4284–4295 (2015).

    Article  CAS  Google Scholar 

  5. Fu, Y. et al. MacroH2A1 associates with nuclear lamina and maintains chromatin architecture in mouse liver cells. Sci. Rep. 5, 17186 (2015).

    Article  CAS  Google Scholar 

  6. Engelke, R. et al. The quantitative nuclear matrix proteome as a biochemical snapshot of nuclear organization. J. Proteome Res. 13, 3940–3956 (2014).

    Article  CAS  Google Scholar 

  7. Calvo, S.E., Clauser, K.R. & Mootha, V.K. MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins. Nucleic Acids Res. 44, D1251–D1257 (2016).

    Article  CAS  Google Scholar 

  8. Pagliarini, D.J. et al. A mitochondrial protein compendium elucidates complex I disease biology. Cell 134, 112–123 (2008).

    Article  CAS  Google Scholar 

  9. Smith, A.C. & Robinson, A.J. MitoMiner v3.1, an update on the mitochondrial proteomics database. Nucleic Acids Res. 44, D1258–D1261 (2016).

    Article  CAS  Google Scholar 

  10. Rhee, H.W. et al. Proteomic mapping of mitochondria in living cells via spatially restricted enzymatic tagging. Science 339, 1328–1331 (2013).

    Article  CAS  Google Scholar 

  11. Hung, V. et al. Proteomic mapping of the human mitochondrial intermembrane space in live cells via ratiometric APEX tagging. Mol. Cell 55, 332–341 (2014).

    Article  CAS  Google Scholar 

  12. Forner, F., Foster, L.J., Campanaro, S., Valle, G. & Mann, M. Quantitative proteomic comparison of rat mitochondria from muscle, heart, and liver. Mol. Cell. Proteomics 5, 608–619 (2006).

    Article  CAS  Google Scholar 

  13. Verstraeten, V.L. et al. Protein farnesylation inhibitors cause donut-shaped cell nuclei attributable to a centrosome separation defect. Proc. Natl. Acad. Sci. USA 108, 4997–5002 (2011).

    Article  CAS  Google Scholar 

  14. Worman, H.J. & Schirmer, E.C. Nuclear membrane diversity: underlying tissue-specific pathologies in disease? Curr. Opin. Cell Biol. 34, 101–112 (2015).

    Article  CAS  Google Scholar 

  15. Draviam, R.A., Shand, S.H. & Watkins, S.C. The beta-delta-core of sarcoglycan is essential for deposition at the plasma membrane. Muscle Nerve 34, 691–701 (2006).

    Article  CAS  Google Scholar 

  16. Uhlen, M. et al. Towards a knowledge-based Human Protein Atlas. Nat. Biotechnol. 28, 1248–1250 (2010).

    Article  CAS  Google Scholar 

  17. Beck, B.D. & Dynlacht, J.R. Heat-induced aggregation of XRCC5 (Ku80) in nontolerant and thermotolerant cells. Radiat. Res. 156, 767–774 (2001).

    Article  CAS  Google Scholar 

  18. Eriksson, M. et al. Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature 423, 293–298 (2003).

    Article  CAS  Google Scholar 

  19. Liu, G.H. et al. Recapitulation of premature ageing with iPSCs from Hutchinson–Gilford progeria syndrome. Nature 472, 221–225 (2011).

    Article  CAS  Google Scholar 

  20. de Haan, W. Lipodystrophy and muscular dystrophy caused by PTRF mutations. Clin. Genet. 77, 436–437 (2010).

    Article  CAS  Google Scholar 

  21. Taranum, S. et al. LINC complex alterations in DMD and EDMD/CMT fibroblasts. Eur. J. Cell Biol. 91, 614–628 (2012).

    Article  CAS  Google Scholar 

  22. Heydemann, A., Demonbreun, A., Hadhazy, M., Earley, J.U. & McNally, E.M. Nuclear sequestration of delta-sarcoglycan disrupts the nuclear localization of lamin A/C and emerin in cardiomyocytes. Hum. Mol. Genet. 16, 355–363 (2007).

    Article  CAS  Google Scholar 

  23. Kandert, S. et al. Nesprin-2 giant safeguards nuclear envelope architecture in LMNA S143F progeria cells. Hum. Mol. Genet. 16, 2944–2959 (2007).

    Article  CAS  Google Scholar 

  24. Lei, K. et al. Inner nuclear envelope proteins SUN1 and SUN2 play a prominent role in the DNA damage response. Curr. Biol. 22, 1609–1615 (2012).

    Article  CAS  Google Scholar 

  25. Starr, D.A. Laminopathies: too much SUN is a bad thing. Curr. Biol. 22, R678–R680 (2012).

    Article  CAS  Google Scholar 

  26. Datta, S., Snow, C.J. & Paschal, B.M. A pathway linking oxidative stress and the Ran GTPase system in progeria. Mol. Biol. Cell 25, 1202–1215 (2014).

    Article  Google Scholar 

  27. Nikolova-Krstevski, V. et al. Nesprin-1 and actin contribute to nuclear and cytoskeletal defects in lamin A/C-deficient cardiomyopathy. J. Mol. Cell. Cardiol. 50, 479–486 (2011).

    Article  CAS  Google Scholar 

  28. Swift, J. et al. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341, 1240104 (2013).

    Article  Google Scholar 

  29. Volonte, D. et al. Inhibition of nuclear factor-erythroid 2-related factor (Nrf2) by caveolin-1 promotes stress-induced premature senescence. Mol. Biol. Cell 24, 1852–1862 (2013).

    Article  CAS  Google Scholar 

  30. Strambio-De-Castillia, C., Niepel, M. & Rout, M.P. The nuclear pore complex: bridging nuclear transport and gene regulation. Nat. Rev. Mol. Cell Biol. 11, 490–501 (2010).

    Article  CAS  Google Scholar 

  31. Mi, H., Muruganujan, A., Casagrande, J.T. & Thomas, P.D. Large-scale gene function analysis with the PANTHER classification system. Nat. Protoc. 8, 1551–1566 (2013).

    Article  Google Scholar 

  32. Zwerger, M. et al. Altering lamina assembly reveals lamina-dependent and -independent functions for A-type lamins. J. Cell Sci. 128, 3607–3620 (2015).

    Article  CAS  Google Scholar 

  33. Britton, S., Coates, J. & Jackson, S.P. A new method for high-resolution imaging of Ku foci to decipher mechanisms of DNA double-strand break repair. J. Cell Biol. 202, 579–595 (2013).

    Article  CAS  Google Scholar 

  34. Cao, K., Capell, B.C., Erdos, M.R., Djabali, K. & Collins, F.S. A lamin A protein isoform overexpressed in Hutchinson–Gilford progeria syndrome interferes with mitosis in progeria and normal cells. Proc. Natl. Acad. Sci. USA 104, 4949–4954 (2007).

    Article  CAS  Google Scholar 

  35. Bar, D.Z. & Collins, F.S. Biotinylation by antibody recognition. Protocol Exchange http://dx.doi.org/10.1038/protex.2017.153 (2017).

  36. Vizcaíno, J.A. et al. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 44, D447–D456 (2016).

    Article  Google Scholar 

Download references

Acknowledgements

We thank S. Patel and M. Gucek of the NHLBI Proteomics Core for operating the mass spectrometer and for technical assistance in sample preparation; S. Wincovitch for microscopy assistance; and J. Fekecs for preparing Figure 1a. We thank L. Bonnycastle and A. Dubose for critical reading of the manuscript, useful suggestions and comments.

Author information

Authors and Affiliations

Authors

Contributions

D.Z.B. and F.S.C. designed the experiments with input from Y.G. D.Z.B. and K.A. performed the experiments with assistance from M.R.E. U.T. performed the mouse work, and D.Z.B. analyzed the data. D.Z.B. and F.S.C. wrote the manuscript, and K.A and Y.G. provided comments and edits.

Corresponding author

Correspondence to Francis S Collins.

Ethics declarations

Competing interests

The National Institutes of Health has filed for a patent application, number 62211160, covering some parts of the information contained in this article.

Integrated supplementary information

Supplementary Figure 1 BAR identifies known nuclear envelope proteins by Western blot and mass-spectrometry

A. Aligned signal distribution of lamin A/C (MAB3211; Cy3) and biotin (FITC-Avidin), derived from Fig 1B.

B. Western blot using Streptavidin-HRP showing antibody guided efficient biotin labeling and recovery of proteins. Beads – post-elution beads were boiled in Laemmli sample buffer to validate effective elution from the beads.

C. LAP2, a known lamin A/C binding protein, is pulled down by BAR, as evident from a Western blot.

D. A scaffold (Proteome Software, Portland, Oregon) screenshot showing multiple known NE and lamin A/C interacting proteins, and their peptide counts in the antibody sample and control.

Supplementary Figure 2 Effect of labeling time and antibody target on BAR

A. Protein level signal intensity (i.e. area) of 1 minute in-gel digestion vs 7 minutes on bead digestion. Proteins ordered (x-axis) in descending 7 minute intensity.

B. Heat-map showing the fraction of each dataset covered by any other dataset.

C. Heavy (lamin A/C MAB3211 antibody) to light (lamin A/C N-18 antibody) ratios of lamins, LAP2β and nuclear pore complex proteins.

D. Heavy (lamin A/C) to light (lamin B1) ratios of lamins, LAP2β and nuclear pore complex proteins.

Supplementary Figure 3 Additional protein staining in mouse and human tissues reproduce original findings

A. Primary mouse skeletal muscle stained for lamin A/C. BF - brightfield.

B. Primary human adipose tissue showing two doughnut shaped nuclei.

C. Primary human skeletal muscle tissue showing SGCA staining at the nuclear periphery.

Scale bar - 10 μm.

Supplementary Figure 4 SILAC controls for differential proteomics show minimal variations.

A. Scatter plot showing signal intensity of BAR extracted proteins from heavy vs. light labeled untreated HeLa cells.

B. Heavy/Light ratio of HeLa NE proteins in untreated cells. Proteins are ordered by peptide count. Proteins passing the filtering criteria in the LMNA-Unbound dataset were considered NE proteins.

Supplementary Figure 5 Ku70 and Ku80 interact with lamin A/C, as evident by FRET

Förster resonance energy transfer (FRET) in HeLa cells show that for Ku70 and Ku80, but not DNA-PKcs, fluorescence intensity increases after bleaching of lamin A/C adjacent fluorophores. FRET efficiency was 0 for DNA-PKcs, 0.34 for Ku70 and 0.15 for Ku80. We note that negative FRET results, as is the case for DNA-PKcs, cannot be interpreted as a lack of interaction.

Supplementary Figure 6 Heat shock affects the localization but not abundance of Ku70, Ku80 and HSPA8

A. Immunofluorescence of Ku80 and HSPA8 before and after heat shock shows nuclear envelope enrichment of Ku80 following heat shock, nuclear envelope localization of HSPA8 prior to heat shock and cytoplasmic depletion and nuclear envelope aggregation of HSPA8 following heat shock. Scale bar - 10 μm.

B. Western blot of lamin A/C, Ku70 and Ku80 showing no changes in amount of protein following heat shock. Cells were counted and equal amount of cells was loaded to gel.

Supplementary Figure 7 CAV1 localization in primary fibroblasts and skeletal muscle

A. HGPS fibroblasts stained for CAV1 and merged with brightfield.

B. Human skeletal muscle stained with CAV1 and DAPI.

Scale bar - 10 μm.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 908 kb)

Life Sciences Reporting Summary (PDF 181 kb)

Supplementary Protocol

Supplementary Protocol (PDF 101 kb)

Supplementary Table 1

Antibodies, plasmids and cell lines used. (XLSX 48 kb)

Supplementary Data

Mass spectrometry data and analysis. (XLSX 3679 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bar, D., Atkatsh, K., Tavarez, U. et al. Biotinylation by antibody recognition—a method for proximity labeling. Nat Methods 15, 127–133 (2018). https://doi.org/10.1038/nmeth.4533

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.4533

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