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:

Visualization and targeting of LGR5+ human colon cancer stem cells

Abstract

The cancer stem cell (CSC) theory highlights a self-renewing subpopulation of cancer cells that fuels tumour growth. The existence of human CSCs is mainly supported by xenotransplantation of prospectively isolated cells, but their clonal dynamics and plasticity remain unclear. Here, we show that human LGR5+ colorectal cancer cells serve as CSCs in growing cancer tissues. Lineage-tracing experiments with a tamoxifen-inducible Cre knock-in allele of LGR5 reveal the self-renewal and differentiation capacity of LGR5+ tumour cells. Selective ablation of LGR5+ CSCs in LGR5-iCaspase9 knock-in organoids leads to tumour regression, followed by tumour regrowth driven by re-emerging LGR5+ CSCs. KRT20 knock-in reporter marks differentiated cancer cells that constantly diminish in tumour tissues, while reverting to LGR5+ CSCs and contributing to tumour regrowth after LGR5+ CSC ablation. We also show that combined chemotherapy potentiates targeting of LGR5+ CSCs. These data provide insights into the plasticity of CSCs and their potential as a therapeutic target in human colorectal cancer.

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: Recapitulation of stem cell hierarchy using colon cancer organoids.
Figure 2: Lineage tracing of human LGR5+ CSCs.
Figure 3: Ablation of LGR5+ CSCs.
Figure 4: Lineage tracing of human KRT20+ CRC cells.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Kreso, A. & Dick, J. E. Evolution of the cancer stem cell model. Cell Stem Cell 14, 275–291 (2014)

    Article  CAS  Google Scholar 

  2. Nassar, D. & Blanpain, C. Cancer stem cells: basic concepts and therapeutic implications. Annu. Rev. Pathol. 11, 47–76 (2016)

    Article  CAS  Google Scholar 

  3. Lapidot, T. et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648 (1994)

    Article  ADS  CAS  Google Scholar 

  4. Nguyen, L. V., Vanner, R., Dirks, P. & Eaves, C. J. Cancer stem cells: an evolving concept. Nat. Rev. Cancer 12, 133–143 (2012)

    Article  CAS  Google Scholar 

  5. Zeuner, A., Todaro, M., Stassi, G. & De Maria, R. Colorectal cancer stem cells: from the crypt to the clinic. Cell Stem Cell 15, 692–705 (2014)

    Article  CAS  Google Scholar 

  6. Plaks, V., Kong, N. & Werb, Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 16, 225–238 (2015)

    Article  CAS  Google Scholar 

  7. Ricci-Vitiani, L. et al. Identification and expansion of human colon-cancer-initiating cells. Nature 445, 111–115 (2007)

    Article  ADS  CAS  Google Scholar 

  8. O’Brien, C. A., Pollett, A., Gallinger, S. & Dick, J. E. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445, 106–110 (2007)

    Article  ADS  Google Scholar 

  9. Kobayashi, S. et al. LGR5-positive colon cancer stem cells interconvert with drug-resistant LGR5-negative cells and are capable of tumor reconstitution. Stem Cells 30, 2631–2644 (2012)

    Article  CAS  Google Scholar 

  10. Kemper, K. et al. Monoclonal antibodies against Lgr5 identify human colorectal cancer stem cells. Stem Cells 30, 2378–2386 (2012)

    Article  CAS  Google Scholar 

  11. Sato, T. et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469, 415–418 (2011)

    Article  ADS  CAS  Google Scholar 

  12. Barker, N. & Clevers, H. Tracking down the stem cells of the intestine: strategies to identify adult stem cells. Gastroenterology 133, 1755–1760 (2007)

    Article  CAS  Google Scholar 

  13. Driessens, G., Beck, B., Caauwe, A., Simons, B. D. & Blanpain, C. Defining the mode of tumour growth by clonal analysis. Nature 488, 527–530 (2012)

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  15. Asfaha, S. et al. Krt19+/Lgr5 cells are radioresistant cancer-initiating stem cells in the colon and intestine. Cell Stem Cell 16, 627–638 (2015)

    Article  CAS  Google Scholar 

  16. Fujii, M. et al. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell 18, 827–838 (2016)

    Article  CAS  Google Scholar 

  17. Merlos-Suárez, A. et al. The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell 8, 511–524 (2011)

    Article  Google Scholar 

  18. Dalerba, P. et al. Single-cell dissection of transcriptional heterogeneity in human colon tumors. Nat. Biotechnol. 29, 1120–1127 (2011)

    Article  CAS  Google Scholar 

  19. Junttila, M. R. et al. Targeting LGR5+ cells with an antibody-drug conjugate for the treatment of colon cancer. Sci. Transl. Med. 7, 314ra186 (2015)

    Article  Google Scholar 

  20. Vermeulen, L. et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol. 12, 468–476 (2010)

    Article  CAS  Google Scholar 

  21. Cernat, L. et al. Colorectal cancers mimic structural organization of normal colonic crypts. PLoS One 9, e104284 (2014)

    Article  ADS  Google Scholar 

  22. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010)

    Article  CAS  Google Scholar 

  23. Kemper, K., Rodermond, H., Colak, S., Grandela, C. & Medema, J. P. Targeting colorectal cancer stem cells with inducible caspase-9. Apoptosis 17, 528–537 (2012)

    Article  CAS  Google Scholar 

  24. van Es, J. H. et al. Dll1+ secretory progenitor cells revert to stem cells upon crypt damage. Nat. Cell Biol. 14, 1099–1104 (2012)

    Article  CAS  Google Scholar 

  25. Metcalfe, C., Kljavin, N. M., Ybarra, R. & de Sauvage, F. J. Lgr5+ stem cells are indispensable for radiation-induced intestinal regeneration. Cell Stem Cell 14, 149–159 (2014)

    Article  CAS  Google Scholar 

  26. Tetteh, P. W. et al. Replacement of lost Lgr5-positive stem cells through plasticity of their enterocyte-lineage daughters. Cell Stem Cell 18, 203–213 (2016)

    Article  CAS  Google Scholar 

  27. Buczacki, S. J. et al. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 495, 65–69 (2013)

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  29. Basak, O. et al. Induced quiescence of Lgr5+ stem cells in intestinal organoids enables differentiation of hormone-producing enteroendocrine cells. Cell Stem Cell 20, 177–190.e4 (2017)

    Article  CAS  Google Scholar 

  30. Chaffer, C. L. et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell 154, 61–74 (2013)

    Article  CAS  Google Scholar 

  31. Brooks, M. D., Burness, M. L. & Wicha, M. S. Therapeutic Implications of cellular heterogeneity and plasticity in breast cancer. Cell Stem Cell 17, 260–271 (2015)

    Article  CAS  Google Scholar 

  32. Gupta, P. B. et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 146, 633–644 (2011)

    Article  CAS  Google Scholar 

  33. Todaro, M., Perez Alea, M., Scopelliti, A., Medema, J. P. & Stassi, G. IL-4-mediated drug resistance in colon cancer stem cells. Cell Cycle 7, 309–313 (2008)

    Article  CAS  Google Scholar 

  34. Kaiser, J. The cancer stem cell gamble. Science 347, 226–229 (2015)

    Article  ADS  CAS  Google Scholar 

  35. Takebe, N. et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat. Rev. Clin. Oncol. 12, 445–464 (2015)

    Article  CAS  Google Scholar 

  36. Ran, F. A. et al. Genome engineering using the CRISPR–Cas9 system. Nat. Protocols 8, 2281–2308 (2013)

    CAS  Google Scholar 

  37. Fujii, M., Matano, M., Nanki, K. & Sato, T. Efficient genetic engineering of human intestinal organoids using electroporation. Nat. Protocols 10, 1474–1485 (2015)

    Article  CAS  Google Scholar 

  38. Muñoz, J. et al. The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent ‘+4’ cell markers. EMBO J. 31, 3079–3091 (2012)

    Article  Google Scholar 

  39. Ito, M. et al. NOD/SCID/γcnull mouse: an excellent recipient mouse model for engraftment of human cells. Blood 100, 3175–3182 (2002)

    Article  CAS  Google Scholar 

  40. Matano, M. et al. Modeling colorectal cancer using CRISPR–Cas9-mediated engineering of human intestinal organoids. Nat. Med. 21, 256–262 (2015)

    Article  CAS  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  42. Bolte, S. & Cordelières, F. P. A guided tour into subcellular colocalization analysis in light microscopy. J. Microsc. 224, 213–232 (2006)

    Article  MathSciNet  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Project for Cancer Research And Therapeutic Evolution (P-CREATE) from the Japan Agency for Medical Research and Development (AMED), by a Grant-in-Aid for Scientific Research on Innovative Areas ‘Stem Cell Ageing and Disease’, and by Grants-in-Aid for Scientific Research funded by the Ministry of Education, Culture, Sports, Science and Technology of Japan. Y.O., M.F. and S.S. were supported by the Japan Society for the Promotion of Science Research Fellowships for Young Scientists. We also thank the Collaborative Research Resources, School of Medicine, Keio University for the technical assistance provided.

Author information

Authors and Affiliations

Authors

Contributions

M.S. performed genetic construction and genetic data analyses. Y.O. performed imaging analyses. S.N., A.T., S.D. and S.S. performed and analysed animal experiments. M.M. engineered organoids. M.F. established CRC organoids. T.K. provided clinical materials. T.S. conceived the project and designed experiments. M.S., Y.O., M.F. and T.S. wrote the manuscript.

Corresponding author

Correspondence to Toshiro Sato.

Ethics declarations

Competing interests

S.N. and S.D. are employees of Otsuka Pharmaceutical Co., Ltd.

Additional information

Reviewer Information Nature thanks C. Blanpain, A. Ventura and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Figure 1 Generation of LGR5–GFP organoids.

a, Targeting strategy for the generation of LGR5–GFP organoids. The locations of PCR primers, restriction sites and loxP sites in the targeted and wild-type alleles are indicated. b, Gel electrophoresis of the PCR products from knocked-in organoids using the primers shown in a. Primer pairs a/b and c/e detected knock-in of the 5′ and 3′ arms, respectively. Primer pair d/e detected Cre-recombined alleles in the organoids treated with adenoviral-Cre. M, size marker; C, control organoids; KI, knock-in organoids. c, Left, Southern blot analyses with a 5′ external probe of NdeI/SacI-digested DNA with 4.6 kb size in control CCO. In the LGR5–GFP organoids, 2.0-kb bands appeared. Right, Southern blot with an internal probe against SacI-digested DNA from LGR5–GFP organoids authenticated locus-specific knock-in. d, Confocal images of LGR5–GFP organoids. Each organoid clone was derived from a different patient-derived clone as summarized in Supplementary Table 2. Scale bars, 25 μm.

Extended Data Figure 2 Generation of KRT20–GFP organoids.

a, Targeting strategy for the generation of KRT20–GFP organoids. The locations of PCR primers and loxP sites in the targeted and wild-type alleles are indicated. b, Gel electrophoresis of the PCR products from knocked-in organoids using the primers shown in a. Primer pairs a/b and c/e detected knock-in of the 5′ and 3′ arms, respectively. Primer pair d/e detected Cre-recombined alleles in the organoids treated with adenoviral-Cre. M, size marker; C, control organoids; KI, knock-in organoids. c, Confocal images of KRT20–GFP CCOs. Each organoid clone was derived from different patient-derived clones as summarized in Supplementary Table 2. Confocal image of LGR5–GFP/KRT20–tdTomato CCO7 (c, bottom right) GFP (green) and tdTomato (red) are visualized. Scale bars, 25 μm.

Extended Data Figure 3 Gene expression and histological expression pattern of LGR5+ CRCs.

a, b, Gene-set enrichment analysis (GSEA). Genes are ranked according to their differential expression between sorted LGR5–GFP+ and LGR5–GFP CCO cells. GSEA confirmed significant enrichment of two independent intestinal stem cell signature gene sets17,38 in LGR5–GFP+ CCO cells (top). GSEA for KRT20–GFP+ and KRT20–GFP cells show inverse correlation with the intestinal stem cell signature genes (bottom). FDR, false discovery rate; NES, normalized enrichment score. c, Immunostaining for the LGR5 reporter (green) and α-smooth muscle actin (αSMA, red) in xenografted (xeno) LGR5-reporter CCOs (top). LGR5 ISH (green) and immunostaining of αSMA (red) in the corresponding parental tissue samples (bottom). αSMA+ cells (red) are found adjacent to LGR5-reporter+ cells (top) or LGR5 mRNA (bottom) in the xenograft and the original patient samples, respectively. Nuclear counterstaining (Hoechst33342) is shown in white. Scale bars, 100 μm.

Extended Data Figure 4 Generation of LGR5-CreER organoids.

a, Targeting strategy for the generation of LGR5-CreER organoids. The locations of PCR primers and loxP sites in the targeted and wild-type alleles are indicated. b, Gel electrophoresis of PCR products from knocked-in organoids using the primers shown in a. Primer pair a/b detected knock-in of the 5′ arm. Primer pair c/e detected the Cre-recombined 3′ arm in the organoids treated with adenoviral-Cre. c, d, In vitro tracing of LGR5+ cells using LGR5-CreER/rainbow CCO12 (c) and CCO7 (d). Time (hours) after the tamoxifen treatment is indicated. Recombined clones are detected by dual (c) and quad (d) colour fluorescence reporters: nGFP (green), YFP (yellow), RFP (red) and mCFP (cyan). Scale bars, 20 μm.

Extended Data Figure 5 Lineage tracing of LGR5-CreER organoids.

ac, Lineage tracing of LGR5-CreER/rainbow CCO7 (a), CCO20 (b) and CCO12 (c). Number of days after tamoxifen treatment is indicated. Descendants of LGR5-CreER+ cells are traced by the RFP reporter (LGR5-tr, red) Non-traced cells are indicated by nuclear GFP (nGFP, green) and no reporter leakage by unintended recombination was observed in the absence of tamoxifen (ac, far left). For some images, differentiated cells are visualized with KRT20 staining (green) as indicated. Persistent expansion of the RFP reporter in a secondary xenografted tumour (a, far right). Nuclear counterstaining (Hoechst33342, white). Scale bars, 100 μm.

Extended Data Figure 6 Generation of LGR5-iCT organoids.

a, Targeting strategy for the generation of LGR5-iCT organoids. The locations of PCR primers and loxP sites in the targeted and wild-type alleles are indicated. b, Gel electrophoresis of PCR products from knocked-in organoids using the primers shown in a and Supplementary Table 1. Primer pairs a/b and c/e detected knock-in of 5′ and 3′ arms. c, d, LGR5-iCT CCO7 was cultured with (c) or without (d) dimerizer. LGR5+ cells (red) and dead cells (blue) are depicted by tdTomato reporter and DAPI staining, respectively. eg, After 1 nM dimerizer treatment, residual LGR5 cancer cells regrew at the same time as re-expression of LGR5–tdTomato (LGR5-r, red). Bright field (top) and epifluorescence (bottom) images. Time (days) after dimerizer treatment is indicated. Scale bars, 100 μm.

Extended Data Figure 7 Validation of LGR5+ cell ablation.

a, Ablation of LGR5+ cells in LGR5-iCT CCO7 in vivo. Five days after dimerizer treatment, LGR5 mRNA (red) expression was lost, whereas KRT20+ (green) cells were preserved. b, Immunostaining of cleaved caspase-3. After dimerizer treatment, apoptotic cells appeared along the tumour edge (red arrowheads). cf, Ablation of LGR5+ cells in vivo using a mixture of LGR5-iCT and CMV-GFP-Luc CCO7 cells. cf, Time course (c) of LGR5+ cells ablation experiments (df). d, Relative total flux value of the vehicle-treated (n = 8) and dimerizer-treated (n = 8) tumours to their original value. e, f, LGR5-iCT (red) and CMV-GFP-Luc (green) organoids intermingled in the tumour after vehicle treatment (e), whereas LGR5–tdTomato expression (red) was lost after dimerizer treatment (f) without reduction in the tumour size (d). Number of tumours for vehicle, n = 8; dimerizer, n = 8. gi, Double-labelling nucleotide pulse-chase assay in LGR5-iCT CCO7. g, Schematic of the experiment. h, Percentage of cells positive for BrdU, EdU and BrdU/EdU staining in control (grey) and on day 15 (dark blue) and day 56 (light blue). i, Confocal imaging of LGR5–tdTomato (purple), BrdU (red) and EdU (green) on day 15 after vehicle (left) or dimerizer treatment (middle), and day 56 after dimerizer treatment (right). EdU-labelled short-term cycling cells exist over time. EdU/BrdU-double-labelled long-term cycling cells decreased on day 15 and recovered on day 56 after dimerizer treatment. LGR5-recovering regions on day 15 after dimerizer treatment are indicated (dotted circle). Scale bars, 100 μm. Data are mean ± s.e.m. (d, h); *P = 0.004, one-way ANOVA (h).

Source data

Extended Data Figure 8 Generation of KRT20-CreER organoids.

a, Targeting strategy for the generation of KRT20-CreER organoids. The locations of PCR primers and loxP sites in the targeted and wild-type alleles are indicated. b, Gel electrophoresis of PCR products from knocked-in organoids using the primers shown in a. Primer pair a/b detected knock-in of the 5′ arm. Primer pair c/e detected the Cre-recombined 3′ arm in the organoids treated with adenoviral-Cre. c, d, Lineage tracing of KRT20-CreER/rainbow organoids derived from CCO12 (c) and CCO7 (d). Number of days after tamoxifen treatment is indicated. c, Lineage tracing of KRT20-CreER/rainbow organoids derived from CCO12. No reporter leakage by unintended recombination was observed in the absence of tamoxifen (left). KRT20-CreER RFP reporter+ cells (KRT20-tr, red) were devoid of Ki67 expression (green) at day 6 (middle), whereas rare reporter+ clonal ribbons appeared at day 28 and co-expressed Ki67 (right). d, Diminishment of KRT20-CreER YFP-tracing reporter+ cells (KRT20-tr, red) was also observed in organoids derived from CCO7. e, Rare clonal ribbons at day 28 detected by four-colour fluorescence; nGFP (green), YFP (yellow), RFP (red) and mCFP (cyan). Scale bars, 100 μm.

Extended Data Figure 9 In vitro colony formation from sorted CRC organoid cells.

a, b, In vitro colony-formation assay using LGR5–GFP (a) or KRT20–GFP (b) cells from indicated lines of CCO. Both GFP+ (top) and GFP cells (bottom) formed colonies. c, LGR5–GFP cells gave rise to organoids containing de novo LGR5–GFP+ cells. d, e, The number of colonies was counted for LGR5–GFP+ and LGR5–GFP cells (d) or KRT20–GFP+ and KRT20–GFP cells (e). Scale bars, 25 μm. *P = 0.018 (d), P = 0.0002 (e, left), P < 0.0001 (e, right), two-tailed Student’s t-test. Colony numbers for four to six wells were counted for each analysis.

Source data

Extended Data Figure 10 Combined effect of chemotherapeutics and CSC targeting.

ac, LGR5 mRNA expression in CCO7 (b) and CCO25 (c) xenografts treated with vehicle, cetuximab (CTX) or oxaliplatin (OX) (a). Relative LGR5 expression to ACTB is shown. dh, Combination therapy by anti-cancer agents and CSC ablation. Time course of chemotherapeutic and/or dimerizer treatments of mice with for LGR5-iCT CCO xenografts (d). Effects of vehicle (V), chemotherapeutics (CTX) or chemotherapeutics + dimerizer (CTX + D) on CCO7 (e) and CCO25 (f) xenografts. CCOs were xenografted subcutaneously and monitored by bioluminescence or caliper measurements. Relative tumour size based on day 0 volume is plotted (e, f). g, h, Relative tumour size 3 weeks after treatment with the indicated therapeutics (V, vehicle; CTX, cetuximab; D, dimerizer; CTX + D, cetuximab + dimerizer) for CCO7 (g) and CCO25 (h). Values from the CTX and CTX + D groups (e, f) were compared to those from the V and D groups aggregated from four (g) or three (h) individual experiments. Each grey plot represents the size of each tumour and tumours with extensive regression (>95% tumour reduction) are highlighted in red. Note that >95% tumour reduction was only achieved by CTX + D treatment. When a tumour was not detectable, the size was set to 0.01 to avoid arithmetic error during log transformation. For each treatment group, number of the analysed tumours is indicated.

Source data

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-2 and Supplementary Figure 1, the uncropped blots. (PDF 371 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

Shimokawa, M., Ohta, Y., Nishikori, S. et al. Visualization and targeting of LGR5+ human colon cancer stem cells. Nature 545, 187–192 (2017). https://doi.org/10.1038/nature22081

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature22081

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