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.

Stem cell function and stress response are controlled by protein synthesis

Abstract

Whether protein synthesis and cellular stress response pathways interact to control stem cell function is currently unknown. Here we show that mouse skin stem cells synthesize less protein than their immediate progenitors in vivo, even when forced to proliferate. Our analyses reveal that activation of stress response pathways drives both a global reduction of protein synthesis and altered translational programmes that together promote stem cell functions and tumorigenesis. Mechanistically, we show that inhibition of post-transcriptional cytosine-5 methylation locks tumour-initiating cells in this distinct translational inhibition programme. Paradoxically, this inhibition renders stem cells hypersensitive to cytotoxic stress, as tumour regeneration after treatment with 5-fluorouracil is blocked. Thus, stem cells must revoke translation inhibition pathways to regenerate a tissue or tumour.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: HFSCs synthesize less protein than their progeny.
Figure 2: Protein synthesis correlates with differentiation.
Figure 3: Tumour-initiating cells synthesize less protein than their progeny.
Figure 4: Nsun2 deletion promotes stem cell identity and tumorigenesis.
Figure 5: Nsun2 deletion imposes distinct translational programmes.
Figure 6: Nsun2 deletion sensitizes tumour-initiating cells to cytotoxic stress.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Mouse next-generation sequencing data have been deposited in the Gene Expression Omnibus under accession number GSE72067. Human data have been deposited in dbGAP under accession number phs000645.v2.p1.

References

  1. Blanco, S. et al. Aberrant methylation of tRNAs links cellular stress to neuro-developmental disorders. EMBO J. 33, 2020–2039 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tuorto, F. et al. RNA cytosine methylation by Dnmt2 and NSun2 promotes tRNA stability and protein synthesis. Nature Struct. Mol. Biol . 19, 900–905 (2012)

    Article  CAS  Google Scholar 

  3. Schaefer, M. et al. RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes Dev. 24, 1590–1595 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ivanov, P., Emara, M. M., Villen, J., Gygi, S. P. & Anderson, P. Angiogenin-induced tRNA fragments inhibit translation initiation. Mol. Cell 43, 613–623 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Spriggs, K. A., Bushell, M. & Willis, A. E. Translational regulation of gene expression during conditions of cell stress. Mol. Cell 40, 228–237 (2010)

    Article  CAS  PubMed  Google Scholar 

  6. Sobala, A. & Hutvagner, G. Small RNAs derived from the 5′ end of tRNA can inhibit protein translation in human cells. RNA Biol. 10, 553–563 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gebetsberger, J., Zywicki, M., Künzi, A. & Polacek, N. tRNA-derived fragments target the ribosome and function as regulatory non-coding RNA in Haloferax volcanii. Archaea 2012, 260909 (2012)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Khan, M. A. et al. Mutation in NSUN2, which encodes an RNA methyltransferase, causes autosomal-recessive intellectual disability. Am. J. Hum. Genet. 90, 856–863 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Martinez, F. J. et al. Whole exome sequencing identifies a splicing mutation in NSUN2 as a cause of a Dubowitz-like syndrome. J. Med. Genet. 49, 380–385 (2012)

    Article  CAS  PubMed  Google Scholar 

  10. Abbasi-Moheb, L. et al. Mutations in NSUN2 cause autosomal-recessive intellectual disability. Am. J. Hum. Genet. 90, 847–855 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Blanco, S. et al. The RNA-methyltransferase Misu (NSun2) poises epidermal stem cells to differentiate. PLoS Genet. 7, e1002403 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hussain, S. et al. The mouse cytosine-5 RNA methyltransferase NSun2 is a component of the chromatoid body and required for testis differentiation. Mol. Cell. Biol. 33, 1561–1570 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fuchs, E. Cell biology: more than skin deep. J. Cell Biol. 209, 629–632 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Plikus, M. V. & Chuong, C. M. Macroenvironmental regulation of hair cycling and collective regenerative behavior. Cold Spring Harb. Perspect. Med. 4, a015198 (2014)

    Google Scholar 

  15. Müller-Röver, S. et al. A comprehensive guide for the accurate classification of murine hair follicles in distinct hair cycle stages. J. Invest. Dermatol. 117, 3–15 (2001)

    Article  PubMed  Google Scholar 

  16. Jaks, V. et al. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nature Genet. 40, 1291–1299 (2008)

    Article  CAS  PubMed  Google Scholar 

  17. Trempus, C. S. et al. Enrichment for living murine keratinocytes from the hair follicle bulge with the cell surface marker CD34. J. Invest. Dermatol. 120, 501–511 (2003)

    CAS  PubMed  Google Scholar 

  18. Youssef, K. K. et al. Identification of the cell lineage at the origin of basal cell carcinoma. Nature Cell Biol. 12, 299–305 (2010)

    Article  CAS  PubMed  Google Scholar 

  19. Signer, R. A., Magee, J. A., Salic, A. & Morrison, S. J. Haematopoietic stem cells require a highly regulated protein synthesis rate. Nature 509, 49–54 (2014)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  20. Fuchs, E. Scratching the surface of skin development. Nature 445, 834–842 (2007)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  21. Hirai, Y., Nose, A., Kobayashi, S. & Takeichi, M. Expression and role of E- and P-cadherin adhesion molecules in embryonic histogenesis. II. Skin morphogenesis. Development 105, 271–277 (1989)

    CAS  PubMed  Google Scholar 

  22. Joshi, R. S. The inner root sheath and the men associated with it eponymically. Int. J. Trichology 3, 57–62 (2011)

    Article  PubMed  PubMed Central  Google Scholar 

  23. Sibilia, M. et al. The EGF receptor provides an essential survival signal for SOS-dependent skin tumor development. Cell 102, 211–220 (2000)

    Article  CAS  PubMed  Google Scholar 

  24. Egeblad, M., Nakasone, E. S. & Werb, Z. Tumors as organs: complex tissues that interface with the entire organism. Dev. Cell 18, 884–901 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Malanchi, I. et al. Cutaneous cancer stem cell maintenance is dependent on β-catenin signalling. Nature 452, 650–653 (2008)

    Article  CAS  PubMed  ADS  Google Scholar 

  26. Atsumi, N. et al. Podoplanin, a novel marker of tumor-initiating cells in human squamous cell carcinoma A431. Biochem. Biophys. Res. Commun. 373, 36–41 (2008)

    Article  CAS  PubMed  Google Scholar 

  27. Schober, M. & Fuchs, E. Tumor-initiating stem cells of squamous cell carcinomas and their control by TGF-β and integrin/focal adhesion kinase (FAK) signaling. Proc. Natl Acad. Sci. USA 108, 10544–10549 (2011)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  28. Owens, D. M., Romero, M. R., Gardner, C. & Watt, F. M. Suprabasal α6β4 integrin expression in epidermis results in enhanced tumourigenesis and disruption of TGFβ signalling. J. Cell Sci. 116, 3783–3791 (2003)

    Article  CAS  PubMed  Google Scholar 

  29. Lin, H. C. et al. High-level β1-integrin expression in a subpopulation of highly tumorigenic oral cancer cells. Clin. Oral Investig. 18, 1277–1284 (2014)

    Article  PubMed  Google Scholar 

  30. Ruggero, D. Translational control in cancer etiology. Cold Spring Harb. Perspect. Biol . 5, a012336 (2013)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Frye, M. & Watt, F. M. The RNA methyltransferase Misu (NSun2) mediates Myc-induced proliferation and is upregulated in tumors. Curr. Biol. 16, 971–981 (2006)

    Article  CAS  PubMed  Google Scholar 

  32. Frye, M. et al. Genomic gain of 5p15 leads to over-expression of Misu (NSUN2) in breast cancer. Cancer Lett. 289, 71–80 (2010)

    Article  CAS  PubMed  Google Scholar 

  33. Schaefer, M., Pollex, T., Hanna, K. & Lyko, F. RNA cytosine methylation analysis by bisulfite sequencing. Nucleic Acids Res. 37, e12 (2009)

    Article  PubMed  CAS  Google Scholar 

  34. Hussain, S. et al. NSun2-mediated cytosine-5 methylation of vault noncoding RNA determines its processing into regulatory small RNAs. Cell Reports 4, 255–261 (2013)

    Article  CAS  PubMed  Google Scholar 

  35. Hussain, S., Aleksic, J., Blanco, S., Dietmann, S. & Frye, M. Characterizing 5-methylcytosine in the mammalian epitranscriptome. Genome Biol. 14, 215 (2013)

    Article  PubMed  PubMed Central  Google Scholar 

  36. Khoddami, V. & Cairns, B. R. Identification of direct targets and modified bases of RNA cytosine methyltransferases. Nature Biotechnol. 31, 458–464 (2013)

    Article  CAS  Google Scholar 

  37. Zhang, X. et al. The tRNA methyltransferase NSun2 stabilizes p16INK4 mRNA by methylating the 3′-untranslated region of p16. Nature Commun . 3, 712 (2012)

    Article  ADS  CAS  Google Scholar 

  38. Tang, H. et al. NSun2 delays replicative senescence by repressing p27 (KIP1) translation and elevating CDK1 translation. Aging 7, 1143–1158 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Xing, J. et al. NSun2 promotes cell growth via elevating cyclin-dependent kinase 1 translation. Mol. Cell. Biol. 35, 4043–4052 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Baranov, P. V. & Michel, A. M. Illuminating translation with ribosome profiling spectra. Nature Methods 13, 123–124 (2016)

    Article  CAS  PubMed  Google Scholar 

  41. Ingolia, N. T., Lareau, L. F. & Weissman, J. S. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell 147, 789–802 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  43. Gerashchenko, M. V., Lobanov, A. V. & Gladyshev, V. N. Genome-wide ribosome profiling reveals complex translational regulation in response to oxidative stress. Proc. Natl Acad. Sci. USA 109, 17394–17399 (2012)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  44. Ingolia, N. T., Ghaemmaghami, S., Newman, J. R. & Weissman, J. S. Genome-wide analysis in vivo of translation with nucleotide resolution using ribosome profiling. Science 324, 218–223 (2009)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  45. Calvo, S. E., Pagliarini, D. J. & Mootha, V. K. Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans. Proc. Natl Acad. Sci. USA 106, 7507–7512 (2009)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  46. Schleich, S. et al. DENR-MCT-1 promotes translation re-initiation downstream of uORFs to control tissue growth. Nature 512, 208–212 (2014)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  47. Lu, P. D., Harding, H. P. & Ron, D. Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response. J. Cell Biol. 167, 27–33 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Morris, D. R. & Geballe, A. P. Upstream open reading frames as regulators of mRNA translation. Mol. Cell. Biol. 20, 8635–8642 (2000)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Longley, D. B., Harkin, D. P. & Johnston, P. G. 5-Fluorouracil: mechanisms of action and clinical strategies. Nature Rev. Cancer 3, 330–338 (2003)

    Article  CAS  Google Scholar 

  50. Kao, R. Y. et al. A small-molecule inhibitor of the ribonucleolytic activity of human angiogenin that possesses antitumor activity. Proc. Natl Acad. Sci. USA 99, 10066–10071 (2002)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  51. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nature Neurosci. 13, 133–140 (2010)

    Article  CAS  PubMed  Google Scholar 

  52. Means, A. L., Xu, Y., Zhao, A., Ray, K. C. & Gu, G. A CK19CreERT knockin mouse line allows for conditional DNA recombination in epithelial cells in multiple endodermal organs. Genesis 46, 318–323 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007)

    Article  CAS  PubMed  ADS  Google Scholar 

  54. Braun, K. M. et al. Manipulation of stem cell proliferation and lineage commitment: visualisation of label-retaining cells in wholemounts of mouse epidermis. Development 130, 5241–5255 (2003)

    Article  CAS  PubMed  Google Scholar 

  55. Hussain, S. et al. The nucleolar RNA methyltransferase Misu (NSun2) is required for mitotic spindle stability. J. Cell Biol. 186, 27–40 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ingolia, N. T., Brar, G. A., Rouskin, S., McGeachy, A. M. & Weissman, J. S. The ribosome profiling strategy for monitoring translation in vivo by deep sequencing of ribosome-protected mRNA fragments. Nature Protocols 7, 1534–1550 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ingolia, N. T. Genome-wide translational profiling by ribosome footprinting. Methods Enzymol. 470, 119–142 (2010)

    Article  CAS  PubMed  Google Scholar 

  58. Schneider-Poetsch, T. et al. Inhibition of eukaryotic translation elongation by cycloheximide and lactimidomycin. Nature Chem. Biol. 6, 209–217 (2010)

    Article  CAS  Google Scholar 

  59. Weinberg, D. E. et al. Improved ribosome-footprint and mRNA measurements provide insights into dynamics and regulation of yeast translation. Cell Reports 14, 1787–1799 (2015)

    Article  CAS  Google Scholar 

  60. Hintze, J. L. & Nelson, R. D. Violin plots: a box plot-density trace synergism. Am. Stat. 52, 4 (1998)

    Google Scholar 

Download references

Acknowledgements

We thank J. Marioni and D. Odom for their advice on analysing the sequencing data. This work was funded by Cancer Research UK, Worldwide Cancer Research, the Medical Research Council (MRC), the European Research Council, and EMBO. Research in M.F.’s laboratory is supported by a core support grant from the Wellcome Trust and MRC to the Wellcome Trust-Medical Research Cambridge Stem Cell Institute.

Author information

Authors and Affiliations

Authors

Contributions

M.F., S.B. and R.B. designed experiments and performed data analysis. S.B., R.B., M.P., S.H., A.S., H.T., R.C.-G. and N.G. performed experiments. P.L., J.A. and S.D. performed bioinformatics analysis. M.F., S.B. and R.B. wrote the manuscript.

Corresponding author

Correspondence to Michaela Frye.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Protein synthesis in epidermal populations.

a, Hair cycle stages and genetic lineage marking using K19- and LGR5 tdTom mice. Cell surface markers to isolated bulge stem cells are CD34 and ITGA6. Telogen: stem cells (CD34+/ITGA6+) are quiescent and resting in the bulge (BG). Early anagen: stem cells divide and give rise to committed progenitors in the hair germ (HG), which then grow downwards into the bulb (BU) surrounding the dermal papilla (DP). Late anagen: cells differentiate upwards to form the hair. Catagen: intermediate phase, when the hair bulb degenerates into a new resting bulge. IFE, interfollicular epidermis; SG, sebaceous glands. Mouse transgenes label K19- (red) and LGR5- (orange) positive stem cells and their progeny. b, OP-puro detection in mouse epidermis at all hair cycle stages. Dotted lines indicate hair follicle and epidermal basal layer. Arrows indicate OP-purohigh cells in the hair follicle. Arrowheads indicate OP-purolow cells in the interfollicular epidermis. Nuclei are stained with DAPI. c, d, tdTom and OP-puro detection in back skin of K19tdTom and Lgr5tdTom mice in telogen and late anagen. Arrows indicate tdTom+ cells. Arrowheads indicate Tomato+/OP-purohigh cells. Dotted line indicates lower bulge. Merged panels from c, d are shown in Fig. 1c–f. e, Hair follicle lineages and differentiation markers used in Fig. 1g–j. Ci, cuticle of inner root sheet; Ch, cuticle; Co, cortex; Cp, companion layer; He, Henle’s layer; Hu, Huxley layer; IRS, inner root sheet; Me, medulla; ORS, outer root sheet. f, P-cadherin and OP-puro detection in a late anagen. Scale bars, 50 μm.

Extended Data Figure 2 Quantification of protein synthesis in epidermal populations.

a, b, Top 2.5%, 10%, 25% and 50% translating epidermal cells (OP-purohigh) (a) were sorted for CD34 and ITGA6 (b). c, Protein synthesis in CD34+/ITGA6+, CD34/ITGA6+ and CD34/ITGA6 epidermal populations in the top 2.5%, 10%, 25%, 50% or 100% (all) translating cells at indicated hair follicle stages. d, Percentage of CD34+/ITGA6+, CD34/ITGA6+ and CD34/ITGA6 cells in the top 2.5%, 10%, 25%, 50% or 100% (all) of translating epidermal cells at indicated stages of the hair cycle. Error bars show mean ± s.d. e, f, Violin plots of protein synthesis in top 2.5% OP-purohigh cells in tdTom epidermal cells sorted for CD34 and ITGA6 from K19-tdTom (e) or Lgr5-tdTom mice (f) at all stages of the hair cycle. (n = mice). Source Data for this figure is available in the online version of the paper.

Source data

Extended Data Figure 3 Protein synthesis and cell cycle analyses in epidermal cells.

ac, Violin plots of protein synthesis in indicated epidermal populations sorted for K19-tdTom-positive (a) and LGR5-tdTom-positive (b) and -negative (c) populations. Protein synthesis is shown for top 10%, 25% or 50% OP-purohigh cells. d, e, Cell cycle analysis (d) and percentage of cells in G1/G0 or S/G2/M in the top 2.5%, 10%, 25% or 50% OP-purohigh cells in late anagen (e). Data represent mean ± s.d. f, Scatter plots correlating protein synthesis in the 2.5% OP-purohigh population with percentage of cells in S/G2/M (top) and G1/G0 (bottom) using all samples independent of hair cycle stage. Linear regression, correlation coefficient (r2) and P value are shown. g, Box plots of protein synthesis (top) and number of cycling cells (bottom) in the top 2.5% translating cell populations (OP-purohigh). h, Box plots of protein synthesis in cycling (S/G2/M) and non-dividing (G1/G0) cells in the 2.5% OP-purohigh population isolated from Lgr5-tdTom mice. Shown are all cells (top), tdTom (Tom) (middle) and tdTom+ (Tom+) (bottom) cells at the indicated hair cycle stages. **P < 0.01, ***P < 0.001, ****P < 0.0001, two-tailed Student’s t-test. n = mice. Source Data for this figure is available in the online version of the paper.

Source data

Extended Data Figure 4 Protein synthesis in squamous tumours.

ac, Co-labelling of OP-puro with markers for undifferentiated basal cells: ITGA6 (a), CD44 (b) and PDPN (c) in mouse tumours. Nuclei are stained with DAPI. Arrows indicate low translating and marker-positive cells. Dotted line indicates invasive front of the tumour. Boxed areas are magnified on the right. d, Gating of low, medium and high OP-puro cells in Nsun2+/+ (wild type) and Nsun2−/−; K5-Sos skin tumours analysed in eg. e, Percentage of OP-purolow cells in tumours from Nsun2+/+ (wild type) and Nsun2−/−; K5-Sos mice. f, g, Flow cytometry for ITGA6 and CD34 in unfractionated epithelial cells from mouse tumours (all cells) or epithelial cells with high, medium and low OP-puro incorporation (f) and quantification (g) (mean ± s.d.; n = 3 mice). h, Flow cytometry for ITGA6 and CD44 in unfractionated epithelial cells from mouse tumours. i, j, Histogram (i) and quantification (j) showing OP-puro incorporation of cells as gated and quantified in h (mean ± s.d.; n = 4 mice). k, l, Detection of endogenous expression of NSUN2 (LacZ) in early (P23) (k) and late (P30) anagen (l) hair follicles. Sections were co-stained with eosin or markers for bulge stem cells K15 and the hair lineages Huxley’s (Hu), cuticle (Ci) (GATA3), and cortex (Co) (LEF1). mo, Haematoxylin and eosin staining (m) and immunostaining for LEF1 (n), K72 and DLX3 (o) in wild-type (WT) and Nsun2−/− skin at P1. Nuclei are stained with DAPI. Insets: magnified boxed area (1, 2). Scale bars, 50 μm. p, Correlation between proliferation and protein synthesis with differentiation of quiescent (QSC) or committed stem cells (CSC), committed progenitors (CP), differentiating progenitors (DP), and terminally differentiated (TD) cells. Source Data for this figure is available in the online version of the paper.

Source data

Extended Data Figure 5 NSUN2 in mouse skin squamous cell carcinomas.

a, Immunostaining for NSUN2, ITGA6, K10 (differentiation marker), laminin 5α and K8 (tumour progression markers), and Slug (epithelial to mesenchymal transition-related gene) at different stages of DMBA-TPA-induced malignant progression to squamous cell carcinoma (SCC). bd, Quantification of tumour diameter normalized to body weight (BW) (b), tumours per mouse (c), and mouse life span (d) in K5-Sos/Nsun2+/+ (K5-Sos), K5-Sos/Nsun2+/− and K5-Sos/Nsun2−/− littermates. Measurements start at P16. Data collection discontinued when mice died (indicated by a dagger). Data represent mean, n ≥ 5 mice of each genotype. e, f, Haematoxylin and eosin staining (e) and immunostaining for ITGB1 (f) in sections from K5-Sos (K5-Sos/Nsun2+/+) and K5-Sos/Nsun2−/− skin tumours. b, basal undifferentiated cells; sb, suprabasal layers. Arrows indicate ITGB1+ cells. g, Relative mRNA expression levels of the indicated transcripts in skin tumours (mean ± s.d.; n = mice). h, Flow cytometry using ITGA6 and CD44 in K5-Sos/Nsun2−/− and control K5-Sos (K5-Sos/Nsun2+/+) tumours. i, Percentage of cells in cell populations as gated in h (mean ± s.d.; n = mice). *P < 0.05; ***P < 0.001 (two-tailed Student’s t-test) (i). j, TdT-mediated dUTP nick end labelling (assay) (TUNEL) assay on sections of K5-Sos tumours expressing (K5-Sos/Nsun2+/+) or lacking Nsun2 (K5-Sos/Nsun2−/−). Arrows indicate TUNEL+ (apoptotic) cells. Nuclei are stained with DAPI. Dotted line indciates boundary of epithelia and stroma (f, j). Scale bars: 25 μm (a), 100 μm (e, f, j). Source Data for this figure is available in the online version of the paper.

Source data

Extended Data Figure 6 Deletion of Nsun2 enhances self-renewal of tumour-initiating cells in a cell-autonomous manner and NSUN2 expression in human skin tumours.

a, Tumour size after grafting of K5-Sos/Nsun2+/+ (K5-Sos) and K5-Sos/Nsun2−/− tumour cells subcutaneously into nude mice (mean ± s.d.; n = 3 mice). bf, Histology (haematoxylin and eosin staining) (b), staining for GFP (c), Ki67 (d), ITGB1 (e) and PDPN (f) in grafted tumour sections. Dotted line indicates boundary between epithelia and stroma. Arrows indicate basal and suprabasal expression. Nuclei are stained with DAPI. gl, Immunohistochemistry for NSUN2 in human normal skin, benign tumours, malignant basal cell carcinomas (BCC) and squamous cell carcinomas (SCC) with increased malignancy (stages classified using the TNM system). Arrows indicate NSUN2high cells. Arrowheads indicate NSUN2low cells. m, Distribution of cells shown in gl according to NSUN2 protein levels. (n ≥ 3 samples). Scale bars, 100 μm. Source Data for this figure is available in the online version of the paper.

Source data

Extended Data Figure 7 NSUN2-dependent RNA methylation of coding and non-coding RNA in mouse tumours.

a, Percentage of NSUN2-methylated sites (>0.15 m5C in Nsun2+/+; <0.05 m5C in Nsun2−/−) out of all covered sites (left) and in non-coding RNA (ncRNA) or introns and exons (right). b, Methylation level in coding and non-coding RNAs (>0.15 m5C in Nsun2+/+; <0.05 m5C in Nsun2−/−; coverage >10 reads). ce, Examples of NSUN2-targeted non-coding RNA (Rpph1) and mRNA (Elf1 and Dscaml1) in Nsun2+/+ (top) and Nsun2−/− (bottom) tumours. f, Number of NSun2-methylated sites in exons 1 to 60 (top) or distance from the transcriptional start site (TSS) in introns (bottom). Plotted sites: >0.1 m5C in Nsun2+/+; <0.05 m5C in Nsun2−/−; coverage >10 reads. g, No correlation between NSUN2 methylation shown in b and RNA abundance in normal skin or K5-Sos skin tumours. NSun2 is highlighted as a control. h, Venn diagram with no significant overlap between NSUN2-methylation targets shown in b and differentially translated mRNAs (P < 0.05; measured as ribosome density of Nsun2+/+ versus Nsun2−/− tumours). il, NSUN2 methylation in tRNAs (>0.15 m5C in Nsun2+/+; <0.05 m5C in Nsun2−/−; coverage >10 reads) (i). Number and location of lost (red) or unchanged (grey) m5C sites in K5-Sos/Nsun2−/− tumours. Nucleotide position in tRNA is shown on the x-axis (j). Examples of NSUN2-targeted tRNAs in Nsun2+/+ (top) and Nsun2−/− (bottom) K5-Sos tumours (k, l). Heatmaps show methylated (red) and unmethylated (grey) cytosines. Cytosines are shown on the x-axis, and sequence reads on the y-axis. Numbers indicate the m5C position in the RNA (ce; k, l). Bisulfite-seq and RNA-seq data represent average of 4 replicates per condition.

Extended Data Figure 8 Nsun2 deletion drives translational changes independent of mRNA expression.

a, Ribosome profiling and RNA-sequencing experiments (see Fig. 5) using Nsun2-expressing (Nsun2+/+) and Nsun2-deficient (Nsun2−/−) K5-Sos skin tumours, or cultured human skin fibroblasts (NSUN2−/−line1 and NSUN2−/−line2 and healthy donors: NSUN2+/−, NSUN2+/+). HTS, high-throughput sequencing. b, Correlation between protein synthesis (ribosome footprint density) in Nsun2+/+ and Nsun2−/− tumours. c, Example of triplet periodicity in ribosome footprints (K5-Sos/Nsun2+/+, replicate 1) shown as number of reads against nucleotide position relative to the translational start site for all ORFs. d, Heatmaps showing ribosome footprint reads around the translational start site (0) in Nsun2+/+ and Nsun2−/− tumours (3 replicates per condition; ribosome density >0; colour indicates RPKM values of footprints). e, Log2 fold change (FC) per transcript in normal skin (left) and tumour samples (right) of significant (P < 0.05) expression differences. Nsun2 RNA levels (red). fj, Scatter plots, linear regression lines and coefficient of correlation (r2) of mRNA expression and protein synthesis (density of ribosome footprints per kb) in Nsun2+/+ (grey) and Nsun2−/− (red) mouse tumours (f) and human fibroblasts (gj). k, Venn diagram of transcripts with significant (P < 0.05) different ribosome footprint density in the 5′ UTR in NSUN2+/−, NSUN2−/−line1 and NSUN2−/−line2 human fibroblasts relative to NSUN2+/+ cells. l, Box plots of ribosome footprint read counts in the 5′ UTR (left) and corresponding CDS (right) of the 192 transcripts in k. ****P < 0.0001 (two-tailed Student’s t-test).

Extended Data Figure 9 RNA methylation-dependent changes of protein synthesis.

a, Venn diagram of transcripts with differential protein synthesis in NSUN2+/− and NSUN2−/− human fibroblasts relative to NSUN2+/+ cells. b, GO terms enriched in 424 commonly differentially translated transcripts in NSUN2−/− lines (a). c, Western blot for NSUN2 and tubulin in NSUN2−/− human fibroblasts rescued with viral constructs expressing wild-type NSUN2 (NSUN2-wt), two catalytically dead mutants (C271A and C321A) or the empty vector. d, Venn diagram of differentially translated transcripts in the indicated rescued cells relative to empty vector-infected control cells. Translation of 173 out of 746 of transcripts (23%) depended on the enzymatic activity of NSUN2. eg, Differential translation of transcripts relative to NSUN2−/− cells (infected with empty vector) showing reduced translation in the presence of wild-type NSUN2 but not the enzymatic-dead versions of NSUN2 (C271A, C321A), corresponding GO categories (f) and examples (g). h, Boyden chamber migration assay towards epidermal growth factor (EGF) or control medium (ctr) using primary human keratinocytes transduced with a siRNA for NSUN2 (si_NSUN2) or a scrambled construct (si_ctr). Data represent mean ± s.d. (n = 3 assays). Western blot confirms downregulation of NSUN2 in the presence of the siRNA construct. i, j Reduced motility in keratinocytes expressing the enzymatic-dead NSUN2 construct (K190M) (K190M: n = 13; NSUN2: n = 19 cells) (i). Western blot confirms equal protein expression levels of K190M and NSUN2 (j). k, Reduced differentiation in primary human keratinocytes expressing the enzymatic-dead NSUN2 (K190M). Staining for NSUN2, ITGA6 or involucrin (IVL) and nuclei (DAPI). Control: empty vector (left); NSUN2: wild-type NSUN2 (middle); K190: enzymatic-dead NSUN2 (right). Arrows indicate NSUN2-expressing ITGA6/IVL+ cells. Arrowheads indicate K190M-expressing ITGA6+/IVL cells. l, Flow cytometry for ITGA6 of keratinocytes transduced with NSUN2 (blue line, top panel), K190M (blue line, bottom panel) or the empty vector (eVector) (red line). Negative control (grey line) represents unstained cells. m, Quantification of IVL+ infected keratinocytes grown in suspension for 24 h to stimulate differentiation. *P < 0.05, **P < 0.01 (two-tailed Student’s t-test) (hm). Scale bar, 100 μm. Source Data for this figure is available in the online version of the paper.

Source data

Extended Data Figure 10 Protein expression differences, drug treatment of Nsun2−/− tumours and graphical summary.

a, b, Western blot analysis of translationally repressed (a) or induced (b) mRNAs in Nsun2−/− (−/−) compared to Nsun2+/+ (WT) skin tumours with quantification of band densitometry on the right (mean ± s.d.; n = 3 mice). *P < 0.05, ***P < 0.001 (two-tailed Student’s t-test). c, d, Control and 5FU-treated tumours, before and after treatment. e, f, Immunohistochemistry for p53 in tumours shown in c, d. gi, Immunostaining for cleaved caspase 3 (Cl−CASP3) (g), Ki67 (h), ITGA6 and K10 (i) in K5-Sos tumours expressing (+/+) or lacking (−/−) Nsun2 and treated with CDDP (see Methods). Scale bars, 100 μm. j, Graphical summary: (1) quiescent undifferentiated stem and progenitor cells are characterized by the absence of NSUN2 and low global protein synthesis; (2) upregulation of NSUN2 counteracts angiogenin-mediated cleavage of tRNAs through site-specific methylation of tRNAs, allowing increased translation of lineage-specific transcripts driving terminal differentiation; (3) cytotoxic stress inhibits NSUN2 and global protein synthesis in particular of lineage-specific transcripts and promotes an undifferentiated quiescent cell state. Yet cell survival after the insult requires re-methylation of tRNAs by NSUN2 (see (2)); (4) the inability to upregulate NSUN2 in response to the cytotoxic insult leads to cell death. Source Data for this figure is available in the online version of the paper.

Source data

Supplementary information

Supplementary Information

This file contains a Supplementary Discussion, Supplementary references and Supplementary Figure 1. (PDF 1133 kb)

Supplementary Tables

This zipped file contains Supplementary Tables 1-10. (ZIP 19080 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Blanco, S., Bandiera, R., Popis, M. et al. Stem cell function and stress response are controlled by protein synthesis. Nature 534, 335–340 (2016). https://doi.org/10.1038/nature18282

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature18282

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