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:

Cell cycle staging of individual cells by fluorescence microscopy

Abstract

Progression through the cell cycle is one of the most fundamental features of cells. Studies of the cell cycle have traditionally relied on the analysis of populations, and they often require specific markers or the use of genetically modified systems, making it difficult to determine the cell cycle stage of individual, unperturbed cells. We describe a protocol, suitable for use in high-resolution imaging approaches, for determining cell cycle staging of individual cells by measuring their DNA content by fluorescence microscopy. The approach is based on the accurate quantification by image analysis of the integrated nuclear intensity of cells stained with a DNA dye, and it can be used in combination with several histochemical methods. We describe and provide the algorithms for two automated image analysis pipelines and the derivation of cell cycle profiles with both commercial and open-source software. This 1–2-d protocol is applicable to adherent cells, and it is adaptable for use with several DNA dyes.

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: Overview of the protocol.
Figure 2: Cell cycle profiles derived by using high-throughput confocal fluorescence microscopy.
Figure 3: Cell cycle histograms obtained by using standard wide-field fluorescence microscopy.
Figure 4: Calculation of population of cells within distinct cell cycle phases.

Similar content being viewed by others

References

  1. Malumbres, M. & Barbacid, M. Cell cycle, CDKs and cancer: a changing paradigm. Nat. Rev. Cancer 9, 153–166 (2009).

    Article  CAS  Google Scholar 

  2. Nurse, P. A long twentieth century of the cell cycle and beyond. Cell 100, 71–78 (2000).

    Article  CAS  Google Scholar 

  3. Hanahan, D. & Weinberg, R.A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  Google Scholar 

  4. Darzynkiewicz, Z. & Huang, X. Analysis of cellular DNA content by flow cytometry. Curr. Protoc. Immunol. 60, 5.7.1–5.7.18 (2004).

    Google Scholar 

  5. Jackson, D. & Cook, P.R. Analyzing DNA replication I: labeling animals, tissues, and cells with bromodeoxyuridine (BrdU). CSH Protoc. 2008 doi:10.1101/pdb.prot5031 (2008).

  6. Rodgers, L. Measurement of DNA content using propidium iodide (PI) staining of fixed whole cells. CSH Protoc. 2006 doi:10.1101/pdb.prot4436 (2006).

  7. Salic, A. & Mitchison, T.J. A chemical method for fast and sensitive detection of DNA synthesis in vivo. Proc. Natl. Acad. Sci. USA 105, 2415–2420 (2008).

    Article  CAS  Google Scholar 

  8. Cappella, P., Gasparri, F., Pulici, M. & Moll, J. A novel method based on click chemistry, which overcomes limitations of cell cycle analysis by classical determination of BrdU incorporation, allowing multiplex antibody staining. Cytometry A 73, 626–636 (2008).

    Article  Google Scholar 

  9. Azuara, V. Profiling of DNA replication timing in unsynchronized cell populations. Nat. Protoc. 1, 2171–2177 (2006).

    Article  CAS  Google Scholar 

  10. Sterzel, W., Bedford, P. & Eisenbrand, G. Automated determination of DNA using the fluorochrome Hoechst 33258. Anal. Biochem. 147, 462–467 (1985).

    Article  CAS  Google Scholar 

  11. Dolezel, J., Greilhuber, J. & Suda, J. Estimation of nuclear DNA content in plants using flow cytometry. Nat. Protoc. 2, 2233–2244 (2007).

    Article  CAS  Google Scholar 

  12. Gratzner, H.G. Monoclonal antibody to 5-bromo- and 5-iododeoxyuridine: A new reagent for detection of DNA replication. Science 218, 474–475 (1982).

    Article  CAS  Google Scholar 

  13. Diermeier-Daucher, S. et al. Cell type specific applicability of 5-ethynyl-2′-deoxyuridine (EdU) for dynamic proliferation assessment in flow cytometry. Cytometry A 75, 535–546 (2009).

    Article  Google Scholar 

  14. Nishitani, H. et al. Two E3 ubiquitin ligases, SCF-Skp2 and DDB1-Cul4, target human Cdt1 for proteolysis. EMBO J. 25, 1126–1136 (2006).

    Article  CAS  Google Scholar 

  15. Roukos, V. et al. Dynamic recruitment of licensing factor Cdt1 to sites of DNA damage. J. Cell Sci. 124, 422–434 (2011).

    Article  CAS  Google Scholar 

  16. Leonhardt, H. et al. Dynamics of DNA replication factories in living cells. J. Cell Biol. 149, 271–280 (2000).

    Article  CAS  Google Scholar 

  17. Essers, J. et al. Nuclear dynamics of PCNA in DNA replication and repair. Mol. Cell Biol. 25, 9350–9359 (2005).

    Article  CAS  Google Scholar 

  18. Easwaran, H.P., Leonhardt, H. & Cardoso, M.C. Cell cycle markers for live cell analyses. Cell Cycle 4, 453–455 (2005).

    Article  CAS  Google Scholar 

  19. Sakaue-Sawano, A. et al. Visualizing spatiotemporal dynamics of multicellular cell-cycle progression. Cell 132, 487–498 (2008).

    Article  CAS  Google Scholar 

  20. Hesse, M. et al. Direct visualization of cell division using high-resolution imaging of M-phase of the cell cycle. Nat. Commun. 3, 1076 (2012).

    Article  Google Scholar 

  21. Clute, P. & Pines, J. Temporal and spatial control of cyclin B1 destruction in metaphase. Nat. Cell Biol. 1, 82–87 (1999).

    Article  CAS  Google Scholar 

  22. Campisi, J. & d′Adda di Fagagna, F. Cellular senescence: when bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 8, 729–740 (2007).

    Article  CAS  Google Scholar 

  23. Evan, G.I. & Vousden, K.H. Proliferation, cell cycle and apoptosis in cancer. Nature 411, 342–348 (2001).

    Article  CAS  Google Scholar 

  24. Trotta, E. & Paci, M. Solution structure of DAPI selectively bound in the minor groove of a DNA T.T mismatch-containing site: NMR and molecular dynamics studies. Nucleic Acids Res. 26, 4706–4713 (1998).

    Article  CAS  Google Scholar 

  25. Roukos, V. et al. Spatial dynamics of chromosome translocations in living cells. Science 341, 660–664 (2013).

    Article  CAS  Google Scholar 

  26. Branzei, D. & Foiani, M. Regulation of DNA repair throughout the cell cycle. Nat. Rev. Mol. Cell Biol. 9, 297–308 (2008).

    Article  CAS  Google Scholar 

  27. Darzynkiewicz, Z., Crissman, H. & Jacobberger, J.W. Cytometry of the cell cycle: cycling through history. Cytometry A 58, 21–32 (2004).

    Article  Google Scholar 

  28. Darzynkiewicz, Z. Critical aspects in analysis of cellular DNA content. Curr. Protoc. Cytom. 56, 7.2.1–7.2.8 (2011).

    Article  Google Scholar 

  29. Kamentsky, L.A., Burger, D.E., Gershman, R.J., Kamentsky, L.D. & Luther, E. Slide-based laser scanning cytometry. Acta Cytol. 41, 123–143 (1997).

    Article  CAS  Google Scholar 

  30. Pozarowski, P., Holden, E. & Darzynkiewicz, Z. Laser scanning cytometry: principles and applications-an update. Methods Mol. Biol. 931, 187–212 (2013).

    Article  CAS  Google Scholar 

  31. Darzynkiewicz, Z., Halicka, H.D. & Zhao, H. Analysis of cellular DNA content by flow and laser-scanning cytometry. Adv. Exp. Med. Biol. 676, 137–147 (2010).

    Article  CAS  Google Scholar 

  32. Gorczyca, W., Darzynkiewicz, Z. & Melamed, M.R. Laser scanning cytometry in pathology of solid tumors. A review. Acta Cytol. 41, 98–108 (1997).

    Article  CAS  Google Scholar 

  33. Lyman, S.K. et al. High-content, high-throughput analysis of cell cycle perturbations induced by the HSP90 inhibitor XL888. PLoS ONE 6, e17692 (2011).

    Article  CAS  Google Scholar 

  34. Floyd, S.R. et al. The bromodomain protein Brd4 insulates chromatin from DNA damage signalling. Nature 498, 246–250 (2013).

    Article  CAS  Google Scholar 

  35. Smith, P.J., Wiltshire, M. & Errington, R.J. DRAQ5 labeling of nuclear DNA in live and fixed cells. Curr. Protoc. Cytom. 28, 7.25.1–7.25.11 (2004).

    Google Scholar 

  36. Martin, R.M., Leonhardt, H. & Cardoso, M.C. DNA labeling in living cells. Cytometry A 67, 45–52 (2005).

    Article  Google Scholar 

  37. Mari, P.O. et al. Influence of the live cell DNA marker DRAQ5 on chromatin-associated processes. DNA Repair (Amst) 9, 848–855 (2010).

    Article  CAS  Google Scholar 

  38. Zhao, H., Traganos, F., Dobrucki, J., Wlodkowic, D. & Darzynkiewicz, Z. Induction of DNA damage response by the supravital probes of nucleic acids. Cytometry A 75, 510–519 (2009).

    Article  Google Scholar 

  39. Nandy, K. et al. Automatic segmentation and supervised learning-based selection of nuclei in cancer tissue images. Cytometry A 81, 743–754 (2012).

    Article  Google Scholar 

  40. Perez-Cadahia, B., Drobic, B. & Davie, J.R. H3 phosphorylation: dual role in mitosis and interphase. Biochem. Cell Biol. 87, 695–709 (2009).

    Article  CAS  Google Scholar 

  41. Yokochi, T. & Gilbert, D.M. Replication labeling with halogenated thymidine analogs. Curr. Protoc. Cell Biol. 35, 22.10.1–22.10.14 (2007).

    Article  Google Scholar 

  42. Brott, D.A., Alvey, J.D., Bleavins, M.R., de la Iglesia, F.A. & Lalwani, N.D. Cell cycle dependent distribution of proliferating cell nuclear antigen/cyclin and cdc2-kinase in mouse T-lymphoma cells. J. Cell Biochem. 52, 362–372 (1993).

    Article  CAS  Google Scholar 

  43. Pines, J. & Hunter, T. Human cyclin A is adenovirus E1A-associated protein p60 and behaves differently from cyclin B. Nature 346, 760–763 (1990).

    Article  CAS  Google Scholar 

  44. Stathopoulou, A. et al. Cdt1 is differentially targeted for degradation by anticancer chemotherapeutic drugs. PLoS ONE 7, e34621 (2012).

    Article  CAS  Google Scholar 

  45. McGarry, T.J. & Kirschner, M.W. Geminin, an inhibitor of DNA replication, is degraded during mitosis. Cell 93, 1043–1053 (1998).

    Article  CAS  Google Scholar 

  46. Nishitani, H., Taraviras, S., Lygerou, Z. & Nishimoto, T. The human licensing factor for DNA replication Cdt1 accumulates in G1 and is destabilized after initiation of S-phase. J. Biol. Chem. 276, 44905–44911 (2001).

    Article  CAS  Google Scholar 

  47. Ridenour, D.A., McKinney, M.C., Bailey, C.M. & Kulesa, P.M. CycleTrak: a novel system for the semi-automated analysis of cell cycle dynamics. Dev. Biol. 365, 189–195 (2012).

    Article  CAS  Google Scholar 

  48. Carpenter, A.E. et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 7, R100 (2006).

    Article  Google Scholar 

  49. R Core Team R: A Language and Environment for Statistical Computing. (R Foundation for Statistical Computing, 2014).

  50. Wickham, H. ggplot2: Elegant Graphics for Data Analysis. (Springer, 2009).

  51. Wickham, H. The split-apply-combine strategy for data analysis. J. Stat. Software 40, 1–29 (2011).

    Google Scholar 

Download references

Acknowledgements

Work in the Misteli laboratory and in the High-Throughput Imaging Facility is supported by the Intramural Research Program of the US National Institutes of Health (NIH), National Cancer Institute (NCI), Center for Cancer Research. We thank S. Burke for modeling the cell cycle data, K. MacKinnon for help with flow cytometry and T. Karpova (NCI Fluorescence Imaging Facility) for help with microscopy.

Author information

Authors and Affiliations

Authors

Contributions

V.R., T.C.V. and G.P. developed the protocol; V.R wrote the manuscript; G.P edited the manuscript; and T.M. supervised the study and edited the manuscript.

Corresponding authors

Correspondence to Vassilis Roukos or Tom Misteli.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Optimization of the imaging parameters for Option A.

A. Selection of the optimal middle, upper and lower planes in z of the field shown in Figure 2A. B. Images of sequential 1μm z stacks of nuclei stained with DAPI. The middle plane (marked by a red box) is selected as the image with the higher nuclei average intensity (Av). The upper plane (yellow box) and the lower plane (turquoise box), are selected to include cells in mitosis (white arrows) and to range the same distance from the middle plane (lower plane: z=-7μm, middle plane: z=0, upper plane: z=+7μm). C. DNA content histograms derived by image analysis on three focal plane average projections (left panel) and by analysis of only the middle plane (right panel). The inability to capture mitotic cells when the middle plane is only acquired is reflected by the lower percentage of cells within the G2/M population (right panel).

Supplementary Figure 2 Cell cycle profiles obtained using different numbers of NIH3T3 cells.

Cells were plated in 16 wells of a 384 well imaging plate and processed as described in Option A of the protocol. Histograms were plotted by pooling data from 16, 10, 8, 4, 2 or a single well (9 different fields per well) corresponding to ~7500, 4500, 3500, 1800, 900 and 500 cells, respectively.

Supplementary Figure 3 Examples of text file structures produced in Step 4Av and required for Step 4Avii.

A. The text file structure that is produced by the image analysis in Step 4Av opened in Excel file. Column F, named attribute “Z_IntegratedDAPIInt” (marked by the red arrow) shows the averaged z integrated nuclear intensity of each individual nucleus detected per well. B. An example of a “GlobalLayout.txt” table file that maps the experimental treatments to the appropriate well positions by row and column numbers is shown.

Supplementary Figure 4 Direct comparison of the cell cycle distributions derived by DNA content analysis using flow cytometry (propidium iodide staining, left side) or produced by single-cell fluorescence microscopy (DAPI staining, right side) by following this protocol.

Flow cytometry and image analysis data obtained by high-throughput (A) or wide-field microscopy (B) of control cells or cells treated with etoposide, were imported in FCS Express and modeled via the multicycle plugin. The percentage of cells within the different cell cycle phases is shown.

Supplementary information

Supplementary Figures

Supplementary Figures 1–4 (PDF 645 kb)

Supplementary Data

Scripts and example imaging data for testing purposes. (ZIP 50657 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roukos, V., Pegoraro, G., Voss, T. et al. Cell cycle staging of individual cells by fluorescence microscopy. Nat Protoc 10, 334–348 (2015). https://doi.org/10.1038/nprot.2015.016

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2015.016

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