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.

Intracellular autofluorescence: a biomarker for epithelial cancer stem cells

Abstract

Cancer stem cells (CSCs) are thought to drive tumor growth, metastasis and chemoresistance. Although surface markers such as CD133 and CD44 have been successfully used to isolate CSCs, their expression is not exclusively linked to the CSC phenotype and is prone to environmental alteration. We identified cells with an autofluorescent subcellular compartment that exclusively showed CSC features across different human tumor types. Primary tumor–derived autofluorescent cells did not overlap with side-population (SP) cells, were enriched in sphere culture and during chemotherapy, strongly expressed pluripotency-associated genes, were highly metastatic and showed long-term in vivo tumorigenicity, even at the single-cell level. Autofluorescence was due to riboflavin accumulation in membrane-bounded cytoplasmic structures bearing ATP-dependent ABCG2 transporters. In summary, we identified and characterized an intrinsic autofluorescent phenotype in CSCs of diverse epithelial cancers and used this marker to isolate and characterize these cells.

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: Identification of autofluorescent cancer stem cells.
Figure 2: In vitro and in vivo characterization of autofluorescent cells.
Figure 3: Invasiveness of autofluorescent cells.
Figure 4: Autofluorescent cells are resistant to chemotherapy.
Figure 5: Mechanism of autofluorescence in cancer stem cells.
Figure 6: Source of autofluorescence in cancer stem cells.

Similar content being viewed by others

References

  1. Clarke, M.F. et al. Cancer stem cells—perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 66, 9339–9344 (2006).

    Article  CAS  Google Scholar 

  2. Campbell, L.L. & Polyak, K. Breast tumor heterogeneity: cancer stem cells or clonal evolution? Cell Cycle 6, 2332–2338 (2007).

    Article  CAS  Google Scholar 

  3. Balic, A., Dorado, J., Alonso-Gómez, M. & Heeschen, C. Stem cells as the root of pancreatic ductal adenocarcinoma. Exp. Cell Res. 318, 691–704 (2012).

    Article  CAS  Google Scholar 

  4. Hermann, P.C., Bhaskar, S., Cioffi, M. & Heeschen, C. Cancer stem cells in solid tumors. Semin. Cancer Biol. 20, 77–84 (2010).

    Article  CAS  Google Scholar 

  5. Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A., Morrison, S. & Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 100, 3983–3988 (2003).

    Article  CAS  Google Scholar 

  6. Gallmeier, E. et al. Inhibition of ataxia telangiectasia- and Rad3-related function abrogates the in vitro and in vivo tumorigenicity of human colon cancer cells through depletion of the CD133+ tumor-initiating cell fraction. Stem Cells 29, 418–429 (2011).

    Article  CAS  Google Scholar 

  7. Wicha, M.S., Liu, S. & Dontu, G. Cancer stem cells: an old idea—a paradigm shift. Cancer Res. 66, 1883–1890 (2006).

    Article  CAS  Google Scholar 

  8. Beier, D. et al. CD133+ and CD133 glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 67, 4010–4015 (2007).

    Article  CAS  Google Scholar 

  9. Joo, K.M. et al. Clinical and biological implications of CD133-positive and CD133-negative cells in glioblastomas. Lab. Invest. 88, 808–815 (2008).

    Article  CAS  Google Scholar 

  10. Ogden, A.T. et al. Identification of A2B5+CD133 tumor-initiating cells in adult human gliomas. Neurosurgery 62, 505–514 (2008).

    Article  Google Scholar 

  11. Wang, J. et al. CD133 negative glioma cells form tumors in nude rats and give rise to CD133 positive cells. Int. J. Cancer 122, 761–768 (2008).

    Article  CAS  Google Scholar 

  12. Hermann, P.C. et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 1, 313–323 (2007).

    Article  CAS  Google Scholar 

  13. Zhang, S.N., Huang, F.T., Huang, Y.J., Zhong, W. & Yu, Z. Characterization of a cancer stem cell-like side population derived from human pancreatic adenocarcinoma cells. Tumori 96, 985–992 (2010).

    Article  Google Scholar 

  14. Zhou, J. et al. Persistence of side population cells with high drug efflux capacity in pancreatic cancer. World J. Gastroenterol. 14, 925–930 (2008).

    Article  Google Scholar 

  15. Kabashima, A. et al. Side population of pancreatic cancer cells predominates in TGF-β-mediated epithelial to mesenchymal transition and invasion. Int. J. Cancer 124, 2771–2779 (2009).

    Article  CAS  Google Scholar 

  16. Broadley, K.W. et al. Side population is not necessary or sufficient for a cancer stem cell phenotype in glioblastoma multiforme. Stem Cells 29, 452–461 (2011).

    Article  CAS  Google Scholar 

  17. Burkert, J., Otto, W.R. & Wright, N.A. Side populations of gastrointestinal cancers are not enriched in stem cells. J. Pathol. 214, 564–573 (2008).

    Article  CAS  Google Scholar 

  18. Clément, V. et al. Retraction: Marker-independent identification of glioma-initiating cells. Nat. Methods 10, 1035 (2013).

    Article  Google Scholar 

  19. Lonardo, E. et al. Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy. Cell Stem Cell 9, 433–446 (2011).

    Article  CAS  Google Scholar 

  20. Iliopoulos, D., Hirsch, H.A., Wang, G. & Struhl, K. Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion. Proc. Natl. Acad. Sci. USA 108, 1397–1402 (2011).

    Article  CAS  Google Scholar 

  21. Hermann, P.C. & Heeschen, C. Metastatic cancer stem cells—quo vadis? Clin. Chem. 59, 1268–1269 (2013).

    Article  CAS  Google Scholar 

  22. Hermann, P.C. et al. Multimodal treatment eliminates cancer stem cells and leads to long-term survival in primary human pancreatic cancer tissue xenografts. PLoS ONE 8, e66371 (2013).

    Article  CAS  Google Scholar 

  23. Cicalese, A. et al. The tumor suppressor p53 regulates polarity of self-renewing divisions in mammary stem cells. Cell 138, 1083–1095 (2009).

    Article  CAS  Google Scholar 

  24. Santini, D. et al. Prognostic role of human equilibrative transporter 1 (hENT1) in patients with resected gastric cancer. J. Cell. Physiol. 223, 384–388 (2010).

    CAS  PubMed  Google Scholar 

  25. Rabindran, S.K., Ross, D.D., Doyle, L.A., Yang, W. & Greenberger, L.M. Fumitremorgin C reverses multidrug resistance in cells transfected with the breast cancer resistance protein. Cancer Res. 60, 47–50 (2000).

    CAS  PubMed  Google Scholar 

  26. Bell, D.H. Characterization of the fluorescence of the antitumor agent, mitoxantrone. Biochim. Biophys. Acta 949, 132–137 (1988).

    Article  CAS  Google Scholar 

  27. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 12, 401–410 (2012).

    Article  CAS  Google Scholar 

  28. van Herwaarden, A.E. et al. Multidrug transporter ABCG2/breast cancer resistance protein secretes riboflavin (vitamin B2) into milk. Mol. Cell. Biol. 27, 1247–1253 (2007).

    Article  CAS  Google Scholar 

  29. Jonker, J.W. et al. The breast cancer resistance protein protects against a major chlorophyll-derived dietary phototoxin and protoporphyria. Proc. Natl. Acad. Sci. USA 99, 15649–15654 (2002).

    Article  CAS  Google Scholar 

  30. van Herwaarden, A.E. & Schinkel, A.H. The function of breast cancer resistance protein in epithelial barriers, stem cells and milk secretion of drugs and xenotoxins. Trends Pharmacol. Sci. 27, 10–16 (2006).

    Article  CAS  Google Scholar 

  31. Ifergan, I., Goler-Baron, V. & Assaraf, Y.G. Riboflavin concentration within ABCG2-rich extracellular vesicles is a novel marker for multidrug resistance in malignant cells. Biochem. Biophys. Res. Commun. 380, 5–10 (2009).

    Article  CAS  Google Scholar 

  32. Mueller, M.T. et al. Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer. Gastroenterology 137, 1102–1113 (2009).

    Article  CAS  Google Scholar 

  33. Qin, D., Xia, Y. & Whitesides, G.M. Soft lithography for micro- and nanoscale patterning. Nat. Protoc. 5, 491–502 (2010).

    Article  CAS  Google Scholar 

  34. Sainz, B. Jr. et al. Identification of the Niemann-Pick C1-like 1 cholesterol absorption receptor as a new hepatitis C virus entry factor. Nat. Med. 18, 281–285 (2012).

    Article  CAS  Google Scholar 

  35. Sainz, B. Jr., Barretto, N. & Uprichard, S.L. Hepatitis C virus infection in phenotypically distinct Huh7 cell lines. PLoS ONE 4, e6561 (2009).

    Article  Google Scholar 

  36. Greenwood, M. & Yule, G.U. On the statistical interpretation of some bacteriological methods employed in water analysis. J. Hyg. (Lond.) 16, 36–54 (1917).

    Article  CAS  Google Scholar 

  37. Taswell, C. Limiting dilution assays for the determination of immunocompetent cell frequencies. I. Data analysis. J. Immunol. 126, 1614–1619 (1981).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. González-Neira and L. Moreno from the CNIO Human Genotyping-CEGEN Unit for performing the TaqMan OpenArray SNAP analysis, and S.M. Trabulo and M. Tatari for their excellent in vivo technical assistance. The research was supported by the ERC Advanced Investigator Grant (Pa-CSC 233460), the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 256974 (EPC-TM-NET) and no. 602783 (CAM-PaC), the Associazione Italiana Ricerca Cancro (AIRC grant no. 12182 to A.S.), the Italian Cancer Genome Project Ministry of University and Research (FIRB RBAP10AHJB to A.S.), the FIMP-Italian Ministry of Health (CUP_J33G13000210001), the Subdirección General de Evaluación y Fomento de la Investigación, Fondo de Investigación Sanitaria (PS09/02129 & PI12/02643) and the Programa Nacional de Internacionalización de la I+D, Subprogramma: FCCI 2009 (PLE2009-0105; both Ministerio de Economía y Competitividad (es), Spain). M.C. was supported by the La Caixa Predoctoral Fellowship Program.

Author information

Authors and Affiliations

Authors

Contributions

I.M.-L. acquired, analyzed and interpreted data as well as developed the study concept and drafted the manuscript; J.D. and A.B. performed the in vivo experiments; E.L., S.A., M.C., A.G.S. and S.Z. acquired and analyzed in vitro data; J.C.-T. designed the microchip-based single-cell assay; D.M. characterized the autofluorescence signal by confocal microscopy and developed the protocol for automated analysis of autofluorescence; M.H., M.E., J.K. and A.S. provided extensively characterized PDAC samples; B.S. and C.H. developed the study concept, obtained funding, interpreted the data and wrote the manuscript.

Corresponding authors

Correspondence to Bruno Sainz Jr or Christopher Heeschen.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Screening for cancer stem cell marker(s)

(a) Flow cytometry analysis of CD133 at increasing times to analysis. Cells were trypsinized, stained, transferred to RPMI medium and then flow cytometry analysis was performed with 0, 10, 20, or 60 min delay. (b) Flow cytometry analysis of CD44+CD133+ and CD44+cMet+ in 185 (left panel) and 215 cells cultured as adherent cells or spheres. (c) Representative in vivo tumorigenicity of cells sorted for different surface marker (combinations). * For statistical analysis, we used limiting dilution analysis (LDA; http://bioinf.wehi.edu.au/software/elda/). LDA is based on the Poisson single-hit model, which assumes that the number of biological active cells in each group varies according to a Poisson distribution, and a single biologically active cell is sufficient for inducing tumor formation. n.s., not significant.

Supplementary Figure 2 Screening for cancer stem cell marker(s)

(a) Representative cytometry plots for side population (SP) and non-side population (Non-SP) cells derived from primary PDAC tissue (untreated or treated with FTC) (left panel). Representative images of sphere formation for SP versus non-SP cells and subsequent quantification (right panel). (b) In vivo tumorigenicity of SP versus non-SP cells (top row, left panel), of aldehyde dehydrogenases (ALDH) negative versus ALDH positive (top row, right panel), adherent versus sphere-derived cells (bottom row, left panel), and Fluo versus Fluo+ cells (bottom row, right panel). * For statistical analysis, we used limiting dilution analysis (LDA; http://bioinf.wehi.edu.au/software/elda/). n.s., not significant.

Supplementary Figure 3 Screening for cancer stem cell marker(s)

In vivo tumorigenicity of primary cells sorted for various CSC markers. * For statistical analysis, we used limiting dilution analysis (LDA; http://bioinf.wehi.edu.au/software/elda/). n.s., not significant.

Supplementary Figure 4 Identification of autofluorescent cancer stem cells

(a) Representative cytometry plots showing no excitation of autofluorescence with 561 nm yellow-green laser (left panel) and with 640 nm red laser (right panel) using the indicated filters. (b) Emission spectra for GFP and autofluorescence, respectively. (c) RTqPCR analysis of GFP mRNA expression. Control values for each sample were compared to a standard curve comprised of serially diluted GFP DNA (left panel). Western blot analysis of GFP protein expression in indicated samples (right panel). (d) Flow cytometry of cell size for Fluo+ and Fluo cells, respectively.

Supplementary Figure 5 Identification of autofluorescent cancer stem cells

(a) Genotyping comparison for unsorted, sorted Fluo+ and sorted Fluo from 3 different PDX samples. Sample groups shared a 100% similar genotype profile (upper table). Graph sample for the SNP rs11199914 showing the same genotype profile between each patient tumor sample (lower panel). (b) Flow cytometry analysis of autofluorescent content in a freshly digested patient sample (left panel), gated for EPCAM+ cells in Fluo+ population (upper right) and Fluo- (lower panel). (c) Flow cytometry analysis of stroma and epithelial cells in a freshly digested patient tumor stained for EpCAM. EpCAM- cells (stroma) were gated to analyze the autofluorescent content.

Supplementary Figure 6 Identification of autofluorescent cancer stem cells

(a) Representative flow cytometry plots illustrating the gating strategy used for autofluorescence analyses. (b) Representative flow cytometry plots for AnnexinV staining in Fluo+ and Fluo cells.

Supplementary Figure 7 Characterization of autofluorescent cancer stem cells

(a) Flow cytometry analysis of autofluorescence content in CRC-014 and CRC-010 tumors (left panel). RT-qPCR analysis of pluripotency-associated gene expression in sorted Fluo+ and Fluo cells from CRC-014 and CRC-010 (n=2, performed in triplicate). Data are normalized for ß-actin expression (right panel). (b) Flow cytometry analysis of autofluorescence content in HCC-6 cells (left panel), RT-qPCR analysis of pluripotency-associated gene expression in primary HCC sorted for Fluo+ and Fluo cells. Data are normalized for ß-actin expression and performed in triplicate (right panel). (c) Flow cytometry analysis of autofluorescence content in Lung-005 tumors (left panel). RT-qPCR analysis of pluripotency-associated gene expression in sorted NSCLC Fluo+ and Fluo cells. Data are normalized for ß-actin expression and performed in triplicate (right panel). (d) Autofluorescent content in different primary patient and PDX tumors. Statistical significance was assessed by Mann-Whitney test.

Supplementary Figure 8 Characterization of autofluorescent cancer stem cells

(a) Polydimethyl-siloxane post-arrays containing several thousand nano-volume wells (left panel). Representative images of single Fluo+ cells giving rise to either (i) two Fluo+ cells or (ii) one Fluo and one Fluo+ cell. Arrows indicate Fluo+ cells (upper right panels). Representative images of a Fluo cells giving rise to two Fluo cells (lower right panel). (b) In vivo serial passaging of Fluo and Fluo+ cells derived from respective tumors originally generated from 103 cells. (c) Flow cytometry plot for autofluorescent content in PDAC PDX single cell-derived tumor (left panel). RT-qPCR analysis of pluripotency-associated gene expression in sorted Fluo+ and Fluo cells obtained from PDAC PDX single cell-derived tumor (right panel). Data are normalized for ß-actin expression and performed in triplicate. Error bars (c) s.d. Statistical significance was assessed by Mann-Whitney test. For statistical analysis, we used limiting dilution analysis (LDA; http://bioinf.wehi.edu.au/software/elda/).

Supplementary Figure 9 Characterization of autofluorescent cancer stem cells

(a) RT-qPCR analysis of pluripotency-associated gene expression in sorted Fluo and Fluo+ cells derived from PDAC-Tumor-02, freshly digested and without culture or supplementation with Riboflavin. Data are normalized using ß-actin expression and performed in triplicate. (b) In vivo tumorigenicity of serially diluted sorted Fluo and Fluo+ cells derived from freshly resected PDX tumors and primary tumors, respectively, without any culturing or supplementation with Riboflavin (right panel). (c) Unsorted cells were treated with Gemcitabine for 12 days. Following treatment, cells were sorted for autofluorescence and injected into mice. Shown are the tumorigenicity results of long-term Gemcitabine treated Fluo and Fluo+ sorted cells after two months. Error bars (a) s.d. Statistical significance was assessed by Mann-Whitney test. For statistical analysis of tumorigenicity, we used limiting dilution analysis (LDA; http://bioinf.wehi.edu.au/software/elda/).

Supplementary Figure 10 Origin and mechanism of autofluorescence

(a) Intracellular ATP content in sorted Fluo+ and Fluo cells and unsorted cells. (b) RT-qPCR analysis of ATG12 gene expression in sorted Fluo+ and Fluo cells of different primary PDAC PDX in vitro cultures (n = 2, each performed in duplicate) (upper panel). Western blot analysis of LC3 protein expression in sorted Fluo+ and Fluo primary PDAC PDX in vitro cells (lower panel). (c) Representative flow cytometry analysis for autofluorescence content using specific autophagy inhibitors (E64D [10 μM] plus Pepstatin-A [1 μg/ml]) or the autophagy inducer Rapamycin (100 ng/ml). (d) Representative flow cytometry plots illustrating the recovery of autofluorescence following the addition of various vitamins. Error bars (a) s.d. of two technical replicates.

Supplementary Figure 11 Source of autofluorescence in cancer stem cells

(a) RT-qPCR analysis of pluripotency-associated gene expression in sorted Fluo and Fluo+ PDAC PDX in vitro cultured (185) cells either untreated or pretreated with 30 μM riboflavin for 24 h. Data are normalized for β-actin expression (left panel). Tumorigenicity of serially diluted sorted Fluo+ and Fluo 185 cells pre-treated with riboflavin 30 μM (right panel). (b) Panc01 and implanted subcutaneously (upper panel) and orthotopically (lower panel), and treated with or without riboflavin prior to analysis. Error bars (a) s.d. of two technical replicates. For statistical analysis of tumorigenicity, we used limiting dilution analysis (LDA; http://bioinf.wehi.edu.au/software/elda/).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1-–11 and Supplementary Tables 1 and 2 (PDF 4511 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Miranda-Lorenzo, I., Dorado, J., Lonardo, E. et al. Intracellular autofluorescence: a biomarker for epithelial cancer stem cells. Nat Methods 11, 1161–1169 (2014). https://doi.org/10.1038/nmeth.3112

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.3112

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