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:

Use of the iNo score to discriminate normal from altered nucleolar morphology, with applications in basic cell biology and potential in human disease diagnostics

Abstract

Ribosome biogenesis is initiated in the nucleolus, a cell condensate essential to gene expression, whose morphology informs cancer pathologists on the health status of a cell. Here, we describe a protocol for assessing, both qualitatively and quantitatively, the involvement of trans-acting factors in the nucleolar structure. The protocol involves use of siRNAs to deplete cells of factors of interest, fluorescence imaging of nucleoli in an automated high-throughput platform, and use of dedicated software to determine an index of nucleolar disruption, the iNo score. This scoring system is unique in that it integrates the five most discriminant shape and textural features of the nucleolus into a parametric equation. Determining the iNo score enables both qualitative and quantitative factor classification with prediction of function (functional clustering), which to our knowledge is not achieved by competing approaches, as well as stratification of their effect (severity of defects) on nucleolar structure. The iNo score has the potential to be useful in basic cell biology (nucleolar structure–function relationships, mitosis, and senescence), developmental and/or organismal biology (aging), and clinical practice (cancer, viral infection, and reproduction). The entire protocol can be completed within 1 week.

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: High-throughput screening pipeline for identifying factors involved in nucleolar structure maintenance.
Fig. 2: Testing the involvement of 86 abundant nucleolar proteins in nucleolar structure maintenance.
Fig. 3: Examples of validation of nucleolar morphological alterations by high-resolution imaging.

Similar content being viewed by others

References

  1. Boulon, S., Westman, B. J., Hutten, S., Boisvert, F. M. & Lamond, A. I. The nucleolus under stress. Mol. Cell 40, 216–227 (2010).

    Article  CAS  Google Scholar 

  2. Boisvert, F. M., van Koningsbruggen, S., Navascues, J. & Lamond, A. I. The multifunctional nucleolus. Nat. Rev. Mol. Cell Biol. 8, 574–585 (2007).

    Article  CAS  Google Scholar 

  3. Nicolas, E. et al. Involvement of human ribosomal proteins in nucleolar structure and p53-dependent nucleolar stress. Nat. Commun. 7, 11390 (2016).

    Article  Google Scholar 

  4. Derenzini, M., Montanaro, L. & Trere, D. What the nucleolus says to a tumour pathologist. Histopathology 54, 753–762 (2009).

    Article  Google Scholar 

  5. Salvetti, A. & Greco, A. Viruses and the nucleolus: the fatal attraction. Biochim. Biophys. Acta 1842, 840–847 (2014).

    Article  CAS  Google Scholar 

  6. Buchwalter, A. & Hetzer, M. W. Nucleolar expansion and elevated protein translation in premature aging. Nat. Commun. 8, 328 (2017).

    Article  Google Scholar 

  7. Fulka, H., Kyogoku, H., Zatsepina, O., Langerova, A. & Fulka, J. Jr. Can nucleoli be markers of developmental potential in human zygotes? Trends Mol. Med. 21, 663–672 (2015).

    Article  CAS  Google Scholar 

  8. Hernandez-Verdun, D., Roussel, P., Thiry, M., Sirri, V. & Lafontaine, D. L. J. The nucleolus: structure/function relationship in RNA metabolism. Wiley Interdiscip. Rev. RNA 1, 415–431 (2010).

    Article  CAS  Google Scholar 

  9. Lafontaine, D., De Vleeschouwer, C., Nicolas, E. & Parisot, P. Nucleolar structure evaluation and manipulation. European patent WO2017191187A2 (2016) (16168087.1-1408).

  10. Feric, M. et al. Coexisting liquid phases underlie nucleolar subcompartments. Cell 165, 1686–1697 (2016).

    Article  CAS  Google Scholar 

  11. Mitrea, D. M. et al. Self-interaction of NPM1 modulates multiple mechanisms of liquid-liquid phase separation. Nat. Commun. 9, 842 (2018).

    Article  Google Scholar 

  12. Thiry, M. & Lafontaine, D. L. J. Birth of a nucleolus: the evolution of nucleolar compartments. Trends Cell Biol. 15, 194–199 (2005).

    Article  CAS  Google Scholar 

  13. Sobecki, M. et al. The cell proliferation antigen Ki-67 organises heterochromatin. eLife 5, e13722 (2016).

    Article  Google Scholar 

  14. Zhu, L. & Brangwynne, C. P. Nuclear bodies: the emerging biophysics of nucleoplasmic phases. Curr. Opin. Cell Biol. 34, 23–30 (2015).

    Article  CAS  Google Scholar 

  15. Courchaine, E. M., Lu, A. & Neugebauer, K. M. Droplet organelles? EMBO J. 35, 1603–1612 (2016).

    Article  CAS  Google Scholar 

  16. Shin, Y. & Brangwynne, C. P. Liquid phase condensation in cell physiology and disease. Science 357, eaaf4382 (2017).

    Article  Google Scholar 

  17. Weber, S. C. & Brangwynne, C. P. Inverse size scaling of the nucleolus by a concentration-dependent phase transition. Curr. Biol. 25, 641–646 (2015).

    Article  CAS  Google Scholar 

  18. Drygin, D., Rice, W. G. & Grummt, I. The RNA polymerase I transcription machinery: an emerging target for the treatment of cancer. Annu. Rev. Pharmacol. Toxicol. 50, 131–156 (2010).

    Article  CAS  Google Scholar 

  19. Bywater, M. J. et al. Inhibition of RNA polymerase I as a therapeutic strategy to promote cancer-specific activation of p53. Cancer Cell 22, 51–65 (2012).

    Article  CAS  Google Scholar 

  20. Peltonen, K. et al. A targeting modality for destruction of RNA polymerase I that possesses anticancer activity. Cancer Cell 25, 77–90 (2014).

    Article  CAS  Google Scholar 

  21. McCann, K. L. & Baserga, S. J. Genetics. Mysterious ribosomopathies. Science 341, 849–850 (2013).

    Article  CAS  Google Scholar 

  22. Narla, A. & Ebert, B. L. Translational medicine: ribosomopathies. Blood 118, 4300–4301 (2011).

    Article  CAS  Google Scholar 

  23. De Keersmaecker, K., Sulima, S. O. & Dinman, J. D. Ribosomopathies and the paradox of cellular hypo- to hyperproliferation. Blood 125, 1377–1382 (2015).

    Article  Google Scholar 

  24. Yelick, P. C. & Trainor, P. A. Ribosomopathies: global process, tissue specific defects. Rare Dis. 3, e1025185 (2015).

    Article  Google Scholar 

  25. Ploton, D. et al. Improvement in the staining and in the visualization of the argyrophilic proteins of the nucleolar organizer region at the optical level. Histochem. J. 18, 5–14 (1986).

    Article  CAS  Google Scholar 

  26. Trere, D. AgNOR staining and quantification. Micron 31, 127–131 (2000).

    Article  CAS  Google Scholar 

  27. Mais, C., Wright, J. E., Prieto, J. L., Raggett, S. L. & McStay, B. UBF-binding site arrays form pseudo-NORs and sequester the RNA polymerase I transcription machinery. Genes Dev. 19, 50–64 (2005).

    Article  CAS  Google Scholar 

  28. Derenzini, M. & Ploton, D. Interphase nucleolar organizer regions in cancer cells. Int. Rev. Exp. Pathol. 32, 149–192 (1991).

    Article  CAS  Google Scholar 

  29. Aubele, M. et al. Guidelines of AgNOR quantitation. Committee on AgNOR quantitation within the European Society of Pathology. Zentralbl. Pathol. 140, 107–108 (1994).

    CAS  PubMed  Google Scholar 

  30. Belin, S. et al. Dysregulation of ribosome biogenesis and translational capacity is associated with tumor progression of human breast cancer cells. PLoS ONE 4, e7147 (2009).

    Article  Google Scholar 

  31. Kodiha, M., Banski, P. & Stochaj, U. Computer-based fluorescence quantification: a novel approach to study nucleolar biology. BMC Cell Biol. 12, 25 (2011).

    Article  CAS  Google Scholar 

  32. Yap, C. K. et al. Automated image based prominent nucleoli detection. J. Pathol. Inform. 6, 39 (2015).

    Article  Google Scholar 

  33. Green, M. R. & Sambrook, J. Molecular Cloning: A Laboratory Manual 4th edn (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2012).

  34. Tafforeau, L. et al. The complexity of human ribosome biogenesis revealed by systematic nucleolar screening of pre-rRNA processing factors. Mol. Cell 51, 539–551 (2013).

    Article  CAS  Google Scholar 

  35. Ideue, T., Hino, K., Kitao, S., Yokoi, T. & Hirose, T. Efficient oligonucleotide-mediated degradation of nuclear noncoding RNAs in mammalian cultured cells. RNA 15, 1578–1587 (2009).

    Article  CAS  Google Scholar 

  36. Mitrea, D. M. et al. Nucleophosmin integrates within the nucleolus via multi-modal interactions with proteins displaying R-rich linear motifs and rRNA. eLife 5, e13571 (2016).

    Article  Google Scholar 

  37. Sloan, K. E., Bohnsack, M. T. & Watkins, N. J. The 5S RNP couples p53 homeostasis to ribosome biogenesis and nucleolar stress. Cell Rep. 5, 237–247 (2013).

    Article  CAS  Google Scholar 

  38. Zhang, J. et al. Assembly factors Rpf2 and Rrs1 recruit 5S rRNA and ribosomal proteins rpL5 and rpL11 into nascent ribosomes. Genes Dev. 21, 2580–2592 (2007).

    Article  CAS  Google Scholar 

  39. Greber, B. J. et al. Insertion of the biogenesis factor Rei1 probes the ribosomal tunnel during 60S maturation. Cell 164, 91–102 (2016).

    Article  CAS  Google Scholar 

  40. Angers, S. et al. Molecular architecture and assembly of the DDB1-CUL4A ubiquitin ligase machinery. Nature 443, 590–593 (2006).

    CAS  PubMed  Google Scholar 

  41. Thiry, M. et al. A protocol for studying the kinetics of RNA within cultured cells: application to ribosomal RNA. Nat. Protoc. 3, 1997–2004 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

V.S. was the recipient of a fellowship from the Fonds National de la Recherche Scientifique (F.R.S./FNRS). The De Vleeschouwer lab is supported by the Fonds National de la Recherche Scientifique (F.R.S./FNRS) and the Walloon Region (DGO6). The Lafontaine lab is supported by the Université Libre de Bruxelles (ULB), the Fonds National de la Recherche Scientifique (F.R.S./FNRS), the Walloon Region (DGO6), the Fédération Wallonie-Bruxelles, and the European Research Development Fund (ERDF) through its affiliation with the Centre for Microscopy and Molecular Imaging (CMMI). The Lafontaine Lab is also affiliated with the ULB Cancer Research Center (U-CRC).

Author information

Authors and Affiliations

Authors

Contributions

V.S. performed the wet lab experimental work. V.S. and D.L.J.L. interpreted the data. P.P. wrote the computer script and performed the image processing analysis. C.D.V. supervised the code production and statistical analysis. D.L.J.L. designed the project, helped with code production, and integrated the data. All authors contributed to writing the manuscript.

Corresponding author

Correspondence to Denis L. J. Lafontaine.

Ethics declarations

Competing interests

The 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.

Related link

Key reference using this protocol

1. Nicolas, E. et al. Nat. Commun. 7, 11390 (2016): https://www.nature.com/articles/ncomms11390

Integrated supplementary information

Supplementary Figure 1 Analysis of the role of DDB1 and ZNF622 in pre-rRNA synthesis, pre-rRNA processing, and nucleolar stress activation.

a, b. Dynamic analysis of pre-rRNA synthesis and processing by metabolic pulse-chase labeling. Cells treated for 3 days with an siRNA targeting DDB1 a, or ZNF622 b, or with a non-targeting SCR siRNA control were labeled with tritiated methionine for 30 min and the label was then “chased”, by incubating cells with non-radioactive methionine, for the indicated times. At each time point, total RNA was extracted, resolved by denaturing agarose-gel electrophoresis and analyzed by fluorography. In order to compare RNA synthesis under different conditions, the signal was quantitated with a phosphor imager at the 0-min time point (black profile for cells treated with SCR siRNA, red profiles for DDB1-depleted and ZNF622-depleted cells). Pre-rRNA intermediates (labeled in cyan) and mature rRNAs (the 18S and 28S rRNAs, in green) are indicated. Depletion of DDB1 expression strongly influences the accumulation of the high-molecular-weight RNA species (45S, 43S, 41S). This observation confirms the PCA-based prediction that this protein is required for RNA synthesis (see also flat profile in red). By comparison, ZNF622 expression depletion influences RNA synthesis only marginally. Pre-rRNA processing remains active after depletion of either proteins (the mature rRNAs 18S and 28S are produced) but it is severely delayed after depletion of DDB1. c, In situ analysis of RNA synthesis by metabolic labeling. Cells treated for 3 days with an siRNA targeting TIF1A or DDB1 or with a non-targeting SCR siRNA control were incubated for 1 h with 5-ethynyl uridine (EU). EU-labeled RNAs were detected by chemoselective ligation (‘click’ chemistry). In cells treated with SCR, intense EU signals are detected in the DAPI-counterstained nucleoli (here the signal corresponds to robust RNA Pol I activity) and the DAPI-stained nucleoplasm (where the signal corresponds largely to transcription by RNA Pol II). In cells depleted of TIF1A or DDB1, the nucleolar signal is lost and only the nucleoplasm displays an EU signal. In control cells incubated for 2 h with α-amanitin, which inhibits RNA Pol II, the nucleoplasmic signal is lost while the nucleolar staining remains. DNA was labeled with DAPI. Scale bar: 10 µm. d, nucleolar stress activation assessed by western-blot detection of the p53 steady-state level. Three independent siRNAs (nos. 1, 2, and 3) were used to deplete the expression of target proteins DDB1 or ZNF622 for 3 days. DDB1 depletion, which severely inhibits rRNA synthesis (see panels a and c), leads to a marked activation of nucleolar stress (p53 accumulation). By contrast, ZNF622 depletion, which only marginally affects pre-rRNA synthesis and processing (panel b), does not activate nucleolar stress. As loading controls, blots were probed for β-actin.

Supplementary information

Supplementary Text and Figures

Supplementary Figure 1

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Stamatopoulou, V., Parisot, P., De Vleeschouwer, C. et al. Use of the iNo score to discriminate normal from altered nucleolar morphology, with applications in basic cell biology and potential in human disease diagnostics. Nat Protoc 13, 2387–2406 (2018). https://doi.org/10.1038/s41596-018-0044-3

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-018-0044-3

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: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer