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.

  • Protocol
  • Published:

Quantitative mapping of fluorescently tagged cellular proteins using FCS-calibrated four-dimensional imaging

Abstract

The ability to tag a protein at its endogenous locus with a fluorescent protein (FP) enables quantitative understanding of protein dynamics at the physiological level. Genome-editing technology has now made this powerful approach routinely applicable to mammalian cells and many other model systems, thereby opening up the possibility to systematically and quantitatively map the cellular proteome in four dimensions. 3D time-lapse confocal microscopy (4D imaging) is an essential tool for investigating spatial and temporal protein dynamics; however, it lacks the required quantitative power to make the kind of absolute and comparable measurements required for systems analysis. In contrast, fluorescence correlation spectroscopy (FCS) provides quantitative proteomic and biophysical parameters such as protein concentration, hydrodynamic radius, and oligomerization but lacks the capability for high-throughput application in 4D spatial and temporal imaging. Here we present an automated experimental and computational workflow that integrates both methods and delivers quantitative 4D imaging data in high throughput. These data are processed to yield a calibration curve relating the fluorescence intensities (FIs) of image voxels to the absolute protein abundance. The calibration curve allows the conversion of the arbitrary FIs to protein amounts for all voxels of 4D imaging stacks. Using our workflow, users can acquire and analyze hundreds of FCS-calibrated image series to map their proteins of interest in four dimensions. Compared with other protocols, the current protocol does not require additional calibration standards and provides an automated acquisition pipeline for FCS and imaging data. The protocol can be completed in 1 d.

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: Principle of FCS for point confocal microscopes.
Figure 2: Workflow for FCS-calibrated imaging.
Figure 3: Example of image quantification using FCS calibration.

Similar content being viewed by others

References

  1. Cong, L., Ran, F., Cox, D., Lin, S. & Barretto, R. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Shen, B. et al. Efficient genome modification by CRISPR-Cas9 nickase with minimal off-target effects. Nat. Methods 11, 399–402 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Trevino, A.E. & Zhang, F. in Methods in Enzymology 546, 161–174 (Elsevier, 2014).

    Article  CAS  PubMed  Google Scholar 

  4. Koch, B. et al. Generation and validation of homozygous fluorescent knock-in cells using CRISPR–Cas9 genome editing. Nat. Protoc. http://dx.doi.org/10.1038/nprot.2018.042 (2018).

  5. Digman, M.A., Stakic, M. & Gratton, E. in Methods in Enzymology 518, 121–144 (Elsevier, 2013).

    Article  CAS  PubMed  Google Scholar 

  6. Krieger, J.W. et al. Imaging fluorescence (cross-) correlation spectroscopy in live cells and organisms. Nat. Protoc. 10, 1948–74 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Bacia, K., Kim, S.A. & Schwille, P. Fluorescence cross-correlation spectroscopy in living cells. Nat. Methods 3, 83–89 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. Bacia, K. & Schwille, P. Practical guidelines for dual-color fluorescence cross-correlation spectroscopy. Nat. Protoc. 2, 2842–2856 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Elson, E.L. Fluorescence correlation spectroscopy: past, present, future. Biophys. J. 101, 2855–2870 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Digman, M.A. & Gratton, E. Lessons in fluctuation correlation spectroscopy. Annu. Rev. Phys. Chem. 62, 645–668 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Mahen, R. et al. Comparative assessment of fluorescent transgene methods for quantitative imaging in human cells. Mol. Biol. Cell 25, 3610–3618 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Walther, N. et al. A quantitative map of human condensins provides new insights into mitotic chromosome architecture. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201801048 (2018).

  13. Cuylen, S. et al. Ki-67 acts as a biological surfactant to disperse mitotic chromosomes. Nature 535, 308–312 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Germier, T. et al. Real-time imaging of a single gene reveals transcription-initiated local confinement. Biophys. J. 113, 1383–1394 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cai, Y. et al. An experimental and computational framework to build a dynamic protein atlas of human cell division. bioRxiv http://dx.doi.org/10.1101/227751 (2017).

  16. Conrad, C. et al. Micropilot: automation of fluorescence microscopy-based imaging for systems biology. Nat. Methods 8, 246–249 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Tischer, C., Hilsenstein, V., Hanson, K. & Pepperkok, R. Adaptive fluorescence microscopy by online feedback image analysis. Methods Cell Biol. 123, 489–503 (2014).

    Article  PubMed  Google Scholar 

  18. Wachsmuth, M. et al. High-throughput fluorescence correlation spectroscopy enables analysis of proteome dynamics in living cells. Nat. Biotechnol. 33, 384–389 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Kremers, G.-J., Gilbert, S.G., Cranfill, P.J., Davidson, M.W. & Piston, D.W. Fluorescent proteins at a glance. J. Cell Sci. 124, 157–160 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Shaner, N.C. et al. A bright monomeric green fluorescent protein derived from Branchiostoma lanceolatum. Nat. Methods 10, 407–409 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Shaner, N.C. et al. Improved monomeric red, orange and yellow fluorescent proteins derived from Discosoma sp. red fluorescent protein. Nat. Biotechnol. 22, 1567–1572 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Bindels, D.S. et al. mScarlet: a bright monomeric red fluorescent protein for cellular imaging. Nat. Methods 14, 53–56 (2016).

    Article  PubMed  CAS  Google Scholar 

  23. Ori, A. et al. Cell type-specific nuclear pores: a case in point for context-dependent stoichiometry of molecular machines. Mol. Syst. Biol. 9, 648 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Suzuki, A., Badger, B.L. & Salmon, E.D. A quantitative description of Ndc80 complex linkage to human kinetochores. Nat. Commun. 6, 1–14 (2015).

    Google Scholar 

  25. Weir, J.R. et al. Insights from biochemical reconstitution into the architecture of human kinetochores. Nature 537, 249–253 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Bauer, M., Cubizolles, F., Schmidt, A. & Nigg, E.A. Quantitative analysis of human centrosome architecture by targeted proteomics and fluorescence imaging. EMBO J. 35, 1–15 (2016).

    Article  CAS  Google Scholar 

  27. Beck, M. et al. The quantitative proteome of a human cell line. Mol. Syst. Biol. 7, 549 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Zacharias, D.A., Violin, J.D., Newton, A.C. & Tsien, R.Y. Partitioning of lipid-modified monomeric GFPs into membrane microdomains of live cells. Science 296, 913–916 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Cranfill, P.J. et al. Quantitative assessment of fluorescent proteins. Nat. Methods 13, 557–562 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Waters, J.C. Accuracy and precision in quantitative fluorescence microscopy. J. Cell Biol. 185, 1135–1148 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Bancaud, A., Huet, S., Rabut, G. & Ellenberg, J. Fluorescence perturbation techniques to study mobility and molecular dynamics of proteins in live cells: FRAP, photoactivation, photoconversion, and FLIP. Cold Spring Harb. Protoc. 2010, pdb.top90 (2010).

  32. Merzlyak, E.M. et al. Bright monomeric red fluorescent protein with an extended fluorescence lifetime. Nat. Methods 4, 555–557 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Lam, A.J. et al. Improving FRET dynamic range with bright green and red fluorescent proteins. Nat. Methods 9, 1005–1012 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Khmelinskii, A. et al. Tandem fluorescent protein timers for in vivo analysis of protein dynamics. Nat. Biotechnol. 30, 708–714 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Schwanhüusser, B. et al. Global quantification of mammalian gene expression control. Nature 473, 337–342 (2011).

    Article  CAS  Google Scholar 

  36. Wu, J.-Q., McCormick, C.D. & Pollard, T.D. in Methods in Cell Biology 89, 253–273 (Elsevier, 2008).

    Article  CAS  PubMed  Google Scholar 

  37. Verdaasdonk, J.S., Lawrimore, J. & Bloom, K. in Methods in Cell Biology 123, 347–365 (Elsevier, 2014).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Wu, J.-Q. & Pollard, T.D. Counting cytokinesis proteins globally and locally in fission yeast. Science 310, 310–314 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Joglekar, A.P., Bouck, D.C., Molk, J.N., Bloom, K.S. & Salmon, E.D. Molecular architecture of a kinetochore-microtubule attachment site. Nat. Cell Biol. 8, 581–585 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Weidemann, T. et al. Counting nucleosomes in living cells with a combination of fluorescence correlation spectroscopy and confocal imaging. J. Mol. Biol. 334, 229–240 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Maeder, C.I. et al. Spatial regulation of Fus3 MAP kinase activity through a reaction-diffusion mechanism in yeast pheromone signalling. Nat. Cell Biol. 9, 1319–1326 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Shivaraju, M. et al. Cell-cycle-coupled structural oscillation of centromeric nucleosomes in yeast. Cell 150, 304–316 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ulbrich, M.H. & Isacoff, E.Y. Subunit counting in membrane-bound proteins. Nat. Methods 4, 319 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ulbrich, M.H. in Springer Series on Fluorescence 13, 263–291 (2011).

    Article  CAS  Google Scholar 

  45. Ta, H., Wolfrum, J. & Herten, D.-P. An extended scheme for counting fluorescent molecules by photon-antibunching. Laser Phys. 20, 119–124 (2010).

    Article  CAS  Google Scholar 

  46. Ta, H. et al. Mapping molecules in scanning far-field fluorescence nanoscopy. Nat. Commun. 6, 7977 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Lawrimore, J., Bloom, K.S. & Salmon, E.D. Point centromeres contain more than a single centromere-specific Cse4 (CENP-A) nucleosome. J. Cell Biol. 195, 573–582 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Charpilienne, A. et al. Individual rotavirus-like particles containing 120 molecules of fluorescent protein are visible in living cells. J. Biol. Chem. 276, 29361–29367 (2001).

    Article  CAS  PubMed  Google Scholar 

  49. Picco, A., Mund, M., Ries, J., Nédélec, F. & Kaksonen, M. Visualizing the functional architecture of the endocytic machinery. Elife 4, e04535 (2015).

  50. Keppler, A. et al. A general method for the covalent labeling of fusion proteins with small molecules in vivo. Nat. Biotechnol. 21, 86–89 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Gautier, A. et al. An engineered protein tag for multiprotein labeling in living cells. Chem. Biol. 15, 128–136 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. Los, G.V. et al. HaloTag: a novel protein labeling technology for cell imaging and protein analysis. ACS Chem. Biol. 3, 373–382 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Mütze, J., Ohrt, T. & Schwille, P. Fluorescence correlation spectroscopy in vivo. Laser Photonics Rev. 5, 52–67 (2011).

    Article  CAS  Google Scholar 

  54. Lukinavicius, G. et al. SiR-Hoechst is a far-red DNA stain for live-cell nanoscopy. Nat. Commun. 6, 8497 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Neumann, B. et al. Phenotypic profiling of the human genome by time-lapse microscopy reveals cell division genes. Nature 464, 721–727 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Otsuka, S. et al. Nuclear pore assembly proceeds by an inside-out extrusion of the nuclear envelope. Elife 5, e19071 (2016).

  57. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Schindelin, J., Rueden, C.T., Hiner, M.C. & Eliceiri, K.W. The ImageJ ecosystem: an open platform for biomedical image analysis. Mol. Reprod. Dev. 82, 518–529 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Rüttinger Buschmann, V., Krämer, B., Erdmann, R., MacDonald, R. & Koberling, F.S. et al. Comparison and accuracy of methods to determine the confocal volume for quantitative fluorescence correlation spectroscopy. J. Microsc. 232, 343–352 (2008).

    Article  PubMed  Google Scholar 

  60. Capoulade, J., Wachsmuth, M., Hufnagel, L. & Knop, M. Quantitative fluorescence imaging of protein diffusion and interaction in living cells. Nat. Biotechnol. 29, 835–839 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Kapusta, P. Absolute Diffusion Coefficients: Compilation of Reference Data for FCS Calibration https://www.picoquant.com/images/uploads/page/files/7353/appnote_diffusioncoefficients.pdf (2010).

Download references

Acknowledgements

We thank the mechanical and electronics workshops of EMBL for custom hardware, the Advanced Light Microscopy Facility of EMBL for microscopy support, and the Flow Cytometry Core Facility of EMBL for cell sorting. We gratefully acknowledge B. Nijmeijer, S. Alexander, and A. Rybina for critical reading of the manuscript. We thank T. Ohrt (Zeiss) for support in the hardware control. This work was supported by grants to J.E. from the European Commission EU-FP7-Systems Microscopy NoE (grant agreement 258068), EU-FP7-MitoSys (grant agreement 241548), and iNEXT (grant agreement 653706), as well as by EMBL (A.Z.P., Y.C., N.W., M.J.H., B.K., M.W., and J.E.). Y.C. and N.W. were supported by the EMBL International PhD Programme (EIPP).

Author information

Authors and Affiliations

Authors

Contributions

A.Z.P. performed the experiments and analysis, and designed the software packages. A.Z.P. developed the protocol with the help of Y.C. and M.W. N.W. tested the protocol and helped in writing the software manuals. B.K. created the homozygous cell line and provided the dextran-labeling protocol. M.J.H. provided the code to segment the cells. A.Z.P. wrote the protocol with the help of N.W., B.K., and J.E.

Corresponding author

Correspondence to Jan Ellenberg.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Quality control of FCS traces

(a) Typical trace that passes the quality control (QC) according to the parameters shown in f. The QC is based on thresholds applied to fitting parameters such as the sum of squared residuals χ2, the coefficient of variation R2, and properties of the photon counts traces (e.g. Bleach coefficient, as calculated by FA). A visual inspection of the photon counts and autocorrelation traces can also be used for quality control. Scale bar 10 μm. (b) FCS measurement point is at the boundary of the cytoplasmic compartment causing a decreasing drift in photon counts. (c) FCS measurement point is at the boundary of the cytoplasmic compartment causing an increasing drift in photon counts. (d) FCS measurement point is at the boundary between a dim and a brighter cell causing large fluctuations in the photon counts. (e) FCS measurement point is in a cell that does not express a fluorescent protein. (f) Table of parameters used for the QC. The measurements in b and c do not pass the QC according to the bleach parameter. The measurement in d does not pass the QC according to the χ2 value. The measurement in e does not pass the QC according to the R2 value. None of the traces in b-e pass the visual inspection. The thresholds used were χ2 < 1.2, R2 > 0.9, 0.8 < Bleach < 1.2. The thresholds can be interactively set in the FCSCalibration software (Supplementary Software 4).

Supplementary Figure 2 CPM and effective volume dependency on pinhole diameter and laser power

(a) Counts per molecule (CPM) as function of the pinhole diameter for Alexa488 in water. (b) Effective confocal focal volume estimated as described in the supplement (Supplementary Notes 1–2 and Box 2) as function of the pinhole diameter for Alexa488. (c) Effective confocal focal volume as function of the excitation laser (Argon 488 nm). The pinhole was 34 μm.

Supplementary Figure 3 Simulated FCS-calibrated imaging

(a) Illustration of a homogeneous fluorophore distribution. (b) The slope of the fluorophore concentration as function of pixel fluorescence intensity gives the calibration coefficient. Simulated PSF is a 3D Gaussian (Eq. S1). The size of the PSF is characterized by its e2 decay w0 and z0 in XY and Z direction, respectively. Here and in all subsequent panels background and detector noise are not simulated. (c) Simulated Z-stack of a point source (50 fluorophores). (d) Using the calibration coefficient and Eqs. S15, S16, and S19 the total number of proteins in each plane is calculated. Several planes along the Z-direction are summed (circles). The expected protein number is obtained when a large region along Z is considered (>2*z0). Simulated images have a pixel-size of 0.4, 0.6, 0.8, 1 times w0 in all directions (different colors). Triangles give the result when all pixels are considered (Eq. S19). Squares give the approximation using the integral of the PSF along Z (Eq. S21). Diamonds give the approximation when the integral of the whole PSF and the pixel with the highest intensity is used (Eq. S23). (e) Simulated fluorophores distributed in a XZ plane (density of 100 fluorophores/μm2). Scale bar 10 μm. (f) Protein density is computed using Eqs. S15 and S24. The expected density is obtained for a region width along the X direction of > 2*w0. Triangles give the result when all pixels are considered. Squares give the approximation when the integral of the PSF along X is used (Eq. S26). (g) Simulated fluorophores distributed in a XY plane (density of 100 fluorophores/μm2). (h) The average protein density is summed for planes along Z (Eqs. S15 and S17). The expected density is obtained for a region width > 2*z0. Triangles give the result when all pixels are considered. Squares give the approximation when the integral of the PSF along Z is used (Eq. S29). Each fluorophore has a simulated intensity of 1000 (a.u.), the simulated PSF is characterized by w0 = 250 nm, z0 = 1500 nm. The black lines give the theoretically expected result after integration of the Gaussian function where N e is the expected protein number/density (50 in d, and 100 in f and h) and Rs is the region size in unit of the PSF characteristic size.

Supplementary information

Supplementary Text and Figures

Supplementary figures 1–3, Supplementary Tables 1–2 and Supplementary Notes 1–6 (PDF 1377 kb)

Supplementary Software 1

The FCSRunner software to manually acquire FCS and imaging data. (ZIP 2057 kb)

Supplementary Software 2

The MyPiC software to automatically acquire FCS and imaging data using adaptive-feedback microscopy. (ZIP 7646 kb)

Supplementary Software 3

A FiJi software package to automatically acquire FCS and imaging data using adaptive-feedback microscopy; to be used in combination with Supplementary Software 2. (ZIP 7971 kb)

Supplementary Software 4

FiJi and R tools to analyze the FCS and imaging data and compute image calibration coefficients. (ZIP 114307 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Politi, A., Cai, Y., Walther, N. et al. Quantitative mapping of fluorescently tagged cellular proteins using FCS-calibrated four-dimensional imaging. Nat Protoc 13, 1445–1464 (2018). https://doi.org/10.1038/nprot.2018.040

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2018.040

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