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:

Lgr5-expressing chief cells drive epithelial regeneration and cancer in the oxyntic stomach

Abstract

The daily renewal of the corpus epithelium is fuelled by adult stem cells residing within tubular glands, but the identity of these stem cells remains controversial. Lgr5 marks homeostatic stem cells and ‘reserve’ stem cells in multiple tissues. Here, we report Lgr5 expression in a subpopulation of chief cells in mouse and human corpus glands. Using a non-variegated Lgr5-2A-CreERT2 mouse model, we show by lineage tracing that Lgr5-expressing chief cells do not behave as corpus stem cells during homeostasis, but are recruited to function as stem cells to effect epithelial renewal following injury by activating Wnt signalling. Ablation of Lgr5+ cells severely impairs epithelial homeostasis in the corpus, indicating an essential role for these Lgr5+ cells in maintaining the homeostatic stem cell pool. We additionally define Lgr5+ chief cells as a major cell-of-origin of gastric cancer. These findings reveal clinically relevant insights into homeostasis, repair and cancer in the corpus.

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: Lgr5 is selectively expressed in a subpopulation of chief cells at the base of mouse and human corpus glands.
Figure 2: Lgr5-expressing chief cells express gastric stem cell markers.
Figure 3: Lgr5-cell ablation impairs epithelial homeostasis in the corpus.
Figure 4: Lgr5-expressing chief cells do not function as stem cells during homeostasis of the adult corpus epithelium.
Figure 5: Lgr5-expressing chief cells drive epithelial regeneration in response to tissue damage.
Figure 6: Damage-inducible modulation of WNT pathway genes during Lgr5+ chief cell activation.
Figure 7: Gastric cancer originates from Lgr5-expressing chief cells.
Figure 8: LGR5 is co-expressed with stem/Wnt markers in human gastric cancer.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Barker, N., Bartfeld, S. & Clevers, H. Tissue-resident adult stem cell populations of rapidly self-renewing organs. Cell Stem Cell 7, 656–670 (2010).

    Article  CAS  Google Scholar 

  2. Arnold, K. et al. Sox2+ adult stem and progenitor cells are important for tissue regeneration and survival of mice. Cell Stem Cell 9, 317–329 (2011).

    Article  CAS  Google Scholar 

  3. Stange, D. E. et al. Differentiated Troy+ chief cells act as reserve stem cells to generate all lineages of the stomach epithelium. Cell 155, 357–368 (2013).

    Article  CAS  Google Scholar 

  4. Hayakawa, Y. et al. Mist1 expressing gastric stem cells maintain the normal and neoplastic gastric epithelium and are supported by a perivascular stem cell niche. Cancer Cell 28, 800–814 (2015).

    Article  CAS  Google Scholar 

  5. Choi, E., Hendley, A. M., Bailey, J. M., Leach, S. D. & Goldenring, J. R. Expression of activated Ras in gastric chief cells of mice leads to the full spectrum of metaplastic lineage transitions. Gastroenterology 150, 918–930.e13 (2016).

    Article  Google Scholar 

  6. Nam, K. T. et al. Mature chief cells are cryptic progenitors for metaplasia in the stomach. Gastroenterology 139, 2028–2037.e9 (2010).

    PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Barker, N. et al. Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6, 25–36 (2010).

    Article  CAS  Google Scholar 

  10. Huch, M. et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 494, 247–250 (2013).

    Article  CAS  Google Scholar 

  11. Leushacke, M., Ng, A., Galle, J., Loeffler, M. & Barker, N. Lgr5+ gastric stem cells divide symmetrically to effect epithelial homeostasis in the pylorus. Cell Rep. 5, 349–356 (2013).

    Article  CAS  Google Scholar 

  12. Tian, H. et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature 478, 255–259 (2011).

    Article  CAS  Google Scholar 

  13. van der Flier, L. G. et al. Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell 136, 903–912 (2009).

    Article  CAS  Google Scholar 

  14. van der Flier, L. G. et al. The intestinal Wnt/TCF signature. Gastroenterology 132, 628–632 (2007).

    Article  CAS  Google Scholar 

  15. Ahn, Y., Sims, C., Logue, J. M., Weatherbee, S. D. & Krumlauf, R. Lrp4 and Wise interplay controls the formation and patterning of mammary and other skin appendage placodes by modulating Wnt signaling. Development 140, 583–593 (2013).

    Article  CAS  Google Scholar 

  16. Ahn, Y., Sanderson, B. W., Klein, O. D. & Krumlauf, R. Inhibition of Wnt signaling by Wise (Sostdc1) and negative feedback from Shh controls tooth number and patterning. Development 137, 3221–3231 (2010).

    Article  CAS  Google Scholar 

  17. Karam, S. M. & Leblond, C. P. Dynamics of epithelial cells in the corpus of the mouse stomach. I. Identification of proliferative cell types and pinpointing of the stem cell. Anat. Rec. 236, 259–279 (1993).

    Article  CAS  Google Scholar 

  18. Huh, W. J. et al. Tamoxifen induces rapid, reversible atrophy, and metaplasia in mouse stomach. Gastroenterology 142, 21–24.e7 (2012).

    Article  Google Scholar 

  19. Nozaki, K. et al. A molecular signature of gastric metaplasia arising in response to acute parietal cell loss. Gastroenterology 134, 511–522 (2008).

    Article  CAS  Google Scholar 

  20. Engevik, A. C. et al. The development of spasmolytic polypeptide/TFF2-expressing metaplasia (SPEM) during gastric repair is absent in the aged stomach. Cell. Mol. Gastroenterol. Hepatol. 2, 605–624 (2016).

    Article  Google Scholar 

  21. Aihara, E. et al. Epithelial regeneration after gastric ulceration causes prolonged cell-type alterations. Cell. Mol. Gastroenterol. Hepatol. 2, 625–647 (2016).

    Article  Google Scholar 

  22. Brabletz, T., Jung, A., Dag, S., Hlubek, F. & Kirchner, T. β-catenin regulates the expression of the matrix metalloproteinase-7 in human colorectal cancer. Am. J. Pathol. 155, 1033–1038 (1999).

    Article  CAS  Google Scholar 

  23. Goldenring, J. R., Nam, K. T. & Mills, J. C. The origin of pre-neoplastic metaplasia in the stomach: chief cells emerge from the Mist. Exp. Cell Res. 317, 2759–2764 (2011).

    Article  CAS  Google Scholar 

  24. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513, 202–209 (2014).

  25. Deng, N. et al. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut 61, 673–684 (2012).

    Article  CAS  Google Scholar 

  26. Nomura, S. et al. Alterations in gastric mucosal lineages induced by acute oxyntic atrophy in wild-type and gastrin-deficient mice. Am. J. Physiol. Gastrointest. Liver Physiol. 288, G362–G75 (2005).

    Article  CAS  Google Scholar 

  27. Petersen, C. P. et al. Macrophages promote progression of spasmolytic polypeptide-expressing metaplasia after acute loss of parietal cells. Gastroenterology 146, 1727–1738.e8 (2014).

    Article  Google Scholar 

  28. Tan, P. & Yeoh, K. G. Genetics and molecular pathogenesis of gastric adenocarcinoma. Gastroenterology 149, 1153–1162.e3 (2015).

    Article  Google Scholar 

  29. McCracken, K. W. et al. Wnt/β-catenin promotes gastric fundus specification in mice and humans. Nature 541, 182–187 (2017).

    Article  CAS  Google Scholar 

  30. Schepers, A. G. et al. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science 337, 730–735 (2012).

    Article  CAS  Google Scholar 

  31. McDonnell, S., Navre, M., Coffey, R. J. & Matrisian, L. M. Expression and localization of the matrix metalloproteinase pump-1 (MMP-7) in human gastric and colon carcinomas. Mol. Carcinog. 4, 527–533 (1991).

    Article  CAS  Google Scholar 

  32. Wroblewski, L. E. et al. Stimulation of MMP-7 (matrilysin) by Helicobacter pylori in human gastric epithelial cells: role in epithelial cell migration. J. Cell. Sci. 116, 3017–3026 (2003).

    Article  CAS  Google Scholar 

  33. Shimizu, T. et al. Characterization of progressive metaplasia in the gastric corpus mucosa of Mongolian gerbils infected with Helicobacter pylori. J. Pathol. 239, 399–410 (2016).

    Article  CAS  Google Scholar 

  34. Crawford, H. C. et al. Helicobacter pylori strain-selective induction of matrix metalloproteinase-7 in vitro and within gastric mucosa. Gastroenterology 125, 1125–1136 (2003).

    Article  CAS  Google Scholar 

  35. Weis, V. G. et al. Maturity and age influence chief cell ability to transdifferentiate into metaplasia. Am. J. Physiol. Gastrointest. Liver Physiol. 312, G67–G76 (2017).

    Article  Google Scholar 

  36. Nam, K. T. et al. Spasmolytic polypeptide-expressing metaplasia (SPEM) in the gastric oxyntic mucosa does not arise from Lgr5-expressing cells. Gut 61, 1678–1685 (2012).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  38. Johnson, L. et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature 410, 1111–1116 (2001).

    Article  CAS  Google Scholar 

  39. Shibata, H. et al. Rapid colorectal adenoma formation initiated by conditional targeting of the Apc gene. Science 278, 120–123 (1997).

    Article  CAS  Google Scholar 

  40. Schwank, G. et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 13, 653–658 (2013).

    Article  CAS  Google Scholar 

  41. Snippert, H. J., Schepers, A. G., Delconte, G., Siersema, P. D. & Clevers, H. Slide preparation for single-cell-resolution imaging of fluorescent proteins in their three-dimensional near-native environment. Nat. Protoc. 6, 1221–1228 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Mustafah and the SIgN Flow Facility for FACS assistance, G. Wright, J. Lim and S. Zulkifli for imaging assistance, and A. Fatehullah, C. Leung and R. Seishima for their technical expertise and manuscript proofreading. We also thank Y. Khay Guan and J. So for providing the human material, and D. H. Alpers, Washington University School of Medicine for providing the Gastric Intrinsic Factor antibody. We thank S. Srivastava for analysing and confirming a SPEM phenotype of Lgr5-cell-derived corpus metaplasia. In addition, we thank F. Sauvage, Department of Molecular Biology, Genentech, South San Francisco, California 94080, USA for providing the Lgr5-DTR-EGFP mice. M.L. is supported by the National Medical Research Council (NMRC) Singapore. N.B. is supported by the Agency for Science, Technology and Research (ASTAR), the Singapore Gastric Cancer Consortium (SGCC) and the National Research Foundation (NRF) NRFI2017-03.

Author information

Authors and Affiliations

Authors

Contributions

M.L. designed, performed experiments, analysed data and wrote the manuscript. S.H.T. performed FACS and microarray experiments, bioinformatics analyses and wrote the manuscript. A.W., L.T.T. and J.G. performed histology, FACS and organoid experiments. K.M. generated inducible expression constructs and performed organoid experiments. Y.S. performed microarray experiments and generated the Lgr5-2A-CreERT2 mouse line. A.H. performed histology experiments. S.L.I.J.D. performed bioinformatics analyses. E.W. generated the Lgr5-2A-CreERT2 mouse line. N.B. supervised the project, generated the Lgr5-2A-CreERT2 mouse line, analysed the data and wrote the manuscript.

Corresponding author

Correspondence to Nick Barker.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Histological analysis of Lgr5+ corpus cells.

(a) ISH for DapB (negative control) GIF, Muc6 and Muc5ac on mouse corpus tissue (image representative of 5 repeats). (b) Co-ISH for DapB (negative control) (image representative of 5 repeats). (c) Co-ISH for Lgr5 together with GIF and Mist1 (image representative of 5 repeats). (d) IHC for Lgr5-driven EGFP on gastric corpus and antral tissue using Lgr5-EGFP-ires-CreERT2(upper panel) or Lgr5-DTR-EGFP (lower panel) (image representative of 5 repeats). (e) Co-IF for EGFP and MUC5AC on Lgr5-DTR-EGFP gastric corpus tissue (image representative of 5 repeats). Scale bars represent 50 μm.

Supplementary Figure 2 Characterization of Lgr5-expressing corpus cells.

(a) EGFP FACS profile of single corpus cells from Lgr5-DTR-EGFP mice and wildtype animals (n = 3). (b) Cytospin of sorted Lgr5-GFPhigh cells from Lgr5-DTR-EGFP mice stained for GFP and GIF (Image representative of 3 repeats). Scale bar represents 5 μm. (c) Hierarchical clustering of 1832 genes that were differentially expressed by more than 2-fold in the Lgr5-GFPhigh population when compared to their Lgr5-GFPneg counterpart (n = 3). (d) ISH for Sostdc1 on mouse antral and small intestinal tissue (image representative of 3 repeats). Scale bars represent 50 μm. (e) Sostdc1 qPCR analysis on mouse corpus, antral, small intestinal and colon tissue (n = 3). Error bars represented as mean ± s.e.m.

Supplementary Figure 3 Lgr5+ cell-ablation in the stomach.

(a) Experimental strategy to ablate Lgr5-expressing cells using the Lgr5-DTR-EGFP mouse model. (b) Histological analysis of the corpus tissue from Lgr5-DTR-EGFP mice and control treated animals at 24 h post DT administration (image representative of 3 repeats). (c) Experimental strategy to ablate Lgr5-expressing cells over a 3 week period using Lgr5-DTR-EGFP mice and control animals. (d) IF for KI67 and CDH1 on Lgr5-cell ablated corpus tissue from Lgr5-DTR-EGFP mice and wildtype-control animals described in c (image representative of 3 repeats). (e) Experimental strategy to ablate Lgr5-expressing cells over a 3 week period followed by a 3 week recovery phase using Lgr5-DTR-EGFP mice and control animals. (f) Histological analysis of the corpus epithelium from Lgr5-DTR-EGFP mice and wildtype control animals described in e (image representative of 3 repeats). (g) Experimental strategy to ablate Lgr5-expressing cells over a 4-day period (n = 3). (h) qPCR analysis for Lgr5, Troy, Mist1 and Sox2 in corpus tissue from Lgr5-DTR-EGFP mice and wildtype control animals described in g (n = 3). (i) The aberrant morphology of the corpus epithelium in Lgr5+ cell-depleted tissue mirrors altered characteristics induced by acute reduction of the peptide hormone gastrin that typically stimulates parietal cells to secrete gastric acid42,43. Gastrin producing G-cells reside along the Lgr5-cell stem cell zone in the lower compartment of antral glands. Given the spatial overlap of antral Lgr5+ and gastrin-producing G cells, we sought to determine if Lgr5-cell ablation would affect antral G cells, which could in turn influence the corpus epithelium. IF for gastrin in antral tissue from Lgr5-DTR-EGFP mice and wildtype control animals described in g (image representative of 3 repeats). (j) Quantification of Gastrin+ cells described in i (n = 3). Data were tested for significance by an unpaired two-tailed t-test. P-values of statistical significance are represented as P < 0.001,P < 0.01, P < 0.05. Scale bars represent 50 μm; error bars represented as mean ± s.e.m.

Supplementary Figure 4 Strategy to knock-in the 2A-CreERT2 cassette into the Lgr5 locus.

(a) Schematic structure of the 2A-CreERT2 cassette knocked into the mouse Lgr5 gene locus. (b) Lgr5-derived lineage tracing in the antrum, small intestine and colon using Lgr5-2A-CreER-T2tg/+/R26R-tdTomatotg/+ mice at 12 h and 14 days post tamoxifen induction (image representative of 3 repeats). (c) Lgr5-derived lineage tracing at 6 months as described in (b) in the antrum, small intestine and colon (image representative of 3 repeats). Scale bars represent 50 μm.

Supplementary Figure 5 Lgr5+ cell-derived RFP+ cell clusters do not expand under homeostatic conditions.

(a) RFP+ cluster analysis in corpus glands from Lgr5-2A-CreER-T2tg/+/R26R-tdTomatotg/+ mice at 2 days, 14 days, 3 months and 6 months post tamoxifen administration (0.1 mg g−1 body weight) (n = 3 for each time point). (b) IF for H-K-ATPase on corpus sections from wildtype animals treated with low dose (0.1 mg g−1 body weight) or high-dose (0.27 mg g−1 body weight) tamoxifen at 2 days post induction (image representative of 3 repeats). (c) IHC for EGFP on corpus sections from Lgr5-DTR-EGFP animals treated with high-dose (0.27 mg g−1 body weight) tamoxifen at 2 days post induction (image representative of 3 repeats). (d) IF for H-K-ATPase and TFF2 on corpus sections from Lgr5-2A-CreER-T2tg/+/R26R-tdTomatotg/+ mice treated with high-dose (0.27 mg g−1 body weight) tamoxifen at 14 days, 3 months and 6 months post administration (image representative of 3 repeats). Scale bar represents 50 μm; error bars represented as mean ± s.e.m.

Supplementary Figure 6 Lgr5+ cell-derived RFP+ cell clusters expand post injury.

(a) RFP+ cluster analysis in corpus glands from Lgr5-2A-CreER-T2tg/+/R26R-tdTomatotg/+ mice at 2 days, 14 days, 3 months and 6 months post tamoxifen administration (0.27 mg g−1 body weight) (n = 3 for each time point). b, Lgr5-EGFP FACS profile of single corpus cells from Lgr5-DTR-EGFP mice (n = 5). (c) Corpus organoid formation efficiency of sorted Lgr5-GFPpos. and GFPneg. cells (n = 10). (d) Representative example of a developing corpus organoid derived from a single Lgr5-GFPhigh corpus cell (n = 10). (e) Experimental strategy to analyze the differentiation capacity of Lgr5-cell derived organoids in response to growth factor withdrawal (n = 3 for each time point). (f) qPCR analysis for Lgr5, Axin2 and Muc5ac in Lgr5-derived organoids cultured in normal (ENRGFW) medium and medium containing only EGF, Rspondin and Gastrin (ERG) (n = 3). Data were tested for significance by an unpaired two-tailed t-test. P-values of statistical significance are represented as P < 0.001, P < 0.01, P < 0.05. Scale bars represent 50 μm; error bars represented as mean ± s.e.m.

Supplementary Figure 7 Damaged-induced differential gene expression during Lgr5+ chief cell activation.

(a) Venn diagram illustrating overlapping genes differentially expressed in tamoxifen-treated Lgr5-GFPhigh cells compared to non-treated Lgr5-GFPhigh control cells and genes that were differentially expressed between tamoxifen-treated Lgr5-GFPhigh/low and tamoxifen-treated Lgr5-GFPneg cells (n = 3). (b) qPCR for Mmp7 and Sostdc1 on corpus tissue samples from wildtype animals treated with high-dose (0.27 mg g−1 body weight) tamoxifen at 24 h post induction (n = 3). (c) ISH for Lgr5, Mmp7 and Sostdc1on corpus tissue sections from wildtype animals treated with high-dose (0.27 mg g−1 body weight) tamoxifen at 48 h post induction (image representative of 3 repeats). d , ISH for DapB (negative control) on corpus tissue sections from wildtype animals treated with high-dose (0.27 mg g−1 body weight) tamoxifen at 24 and 48 h post induction (image representative of 3 repeats). (e) Doxycycline-inducible Synthesized Mmp7 plasmid map and qPCR analysis for Mmp7 (synthesized) in Mmp7 transfected Lgr5+ cell-derived corpus organoids at 3 days post doxycycline administration (n = 3). (f) Doxycycline-inducible Synthesized Sostdc1 plasmid map and qPCR analysis for Sostdc1 (synthesized) in Sostdc1 transfected Lgr5-cell derived corpus organoids at 3 days post doxycycline administration (n = 3). Data were tested for significance by an unpaired two-tailed t-test. P-values of statistical significance are represented as P < 0.001, P < 0.01, P < 0.05. Scale bars represent 50 μm; error bars represented as mean ± s.e.m.

Supplementary Figure 8 Histology on cancer and control corpus tissue.

(a) Experimental strategy to induce control animals (LSL-Kras(G12D)tg/+). (b) IHC staining for Phospho-MAPK, KI67, MUC5AC, TFF2 and H-K-ATPase on control (LSL-Kras(G12D)tg/+) corpus sections at 4 months post-tamoxifen induction (image representative of 3 repeats). (c) Experimental strategy to induce Lgr5-driven active Kras(G12D) expression. (d) IF staining for CDH1, VIMENTIN and GIF on (Lgr5-2A-CreER-T2tg/+/LSL-Kras(G12D)tg/+) corpus sections at 4 months post-tamoxifen induction (image representative of 3 repeats). (e) IHC analysis on ‘normal’ human corpus tissue (image representative of 3 repeats). (f) ISH for DapB (negative control) on human corpus tumor tissue (image representative of 3 repeats). g, Co-ISH for DapB (negative control) on human corpus tumor tissue (image representative of 3 repeats). Scale bars represent 50 μm.

Supplementary information

Supplementary Information

Supplementary Information (PDF 35481 kb)

Supplementary Table 1

Supplementary Information (XLSX 65 kb)

Supplementary Table 2

Supplementary Information (XLSX 32 kb)

Supplementary Table 3

Supplementary Information (XLSX 93 kb)

Long-term ablation of Lgr5-expressing chief cells (xy-plane).

xy-plane imaging of wholemount corpus tissue from Lgr5-DTR-EGFP mice after 3 weeks of sustained DT administration (β-catenin staining to demarcate cell boundaries in white, nuclear staining using DAPI in blue). (AVI 5430 kb)

Control-treated corpus tissue (xy-plane).

xy-plane imaging of wholemount corpus tissue from wildtype mice after 3 weeks of sustained DT administration (β-catenin staining to demarcate cell boundaries in white, nuclear staining using DAPI in blue). (AVI 8159 kb)

Long-term ablation of Lgr5-expressing Chief Cells (xz-plane).

xz-plane imaging of wholemount corpus tissue from Lgr5-DTR-EGFP mice after 3 weeks of sustained DT administration (β-catenin staining to demarcate cell boundaries in white, nuclear staining using DAPI in blue). (AVI 14641 kb)

Control-treated Corpus Tissue (xz-plane).

xz-plane imaging of wholemount corpus tissue from wildtype mice after 3 weeks of sustained DT administration (β-catenin staining to demarcate cell boundaries in white, nuclear staining using DAPI in blue). (AVI 13821 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leushacke, M., Tan, S., Wong, A. et al. Lgr5-expressing chief cells drive epithelial regeneration and cancer in the oxyntic stomach. Nat Cell Biol 19, 774–786 (2017). https://doi.org/10.1038/ncb3541

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3541

This article is cited by

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