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:

Variability within rare cell states enables multiple paths toward drug resistance

Abstract

Molecular differences between individual cells can lead to dramatic differences in cell fate, such as death versus survival of cancer cells upon drug treatment. These originating differences remain largely hidden due to difficulties in determining precisely what variable molecular features lead to which cellular fates. Thus, we developed Rewind, a methodology that combines genetic barcoding with RNA fluorescence in situ hybridization to directly capture rare cells that give rise to cellular behaviors of interest. Applying Rewind to BRAFV600E melanoma, we trace drug-resistant cell fates back to single-cell gene expression differences in their drug-naive precursors (initial frequency of ~1:1,000–1:10,000 cells) and relative persistence of MAP kinase signaling soon after drug treatment. Within this rare subpopulation, we uncover a rich substructure in which molecular differences among several distinct subpopulations predict future differences in phenotypic behavior, such as proliferative capacity of distinct resistant clones after drug treatment. Our results reveal hidden, rare-cell variability that underlies a range of latent phenotypic outcomes upon drug exposure.

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

Fig. 1: Rewind identifies rare cell states giving rise to vemurafenib-resistant colonies.
Fig. 2: A coordinated primed cell state characterized by high expression of multiple markers gives rise to vemurafenib resistance in WM989 A6-G3 cells.
Fig. 3: Resistance to vemurafenib is associated with single-cell variability in phosphorylated ERK levels 24 h after but not before treatment.
Fig. 4: Variation in gene expression among primed cells is associated with differences in resistant cell fate.
Fig. 5: Rewind identifies a distinct subpopulation of cells that require DOT1L inhibition to become vemurafenib resistant.
Fig. 6: DOT1Li inhibition enables a new subpopulation of cells to survive vemurafenib treatment without converting them into the known primed cell state.

Similar content being viewed by others

Data availability

All RNA sequencing data generated for this study are available at the Gene Expression Omnibus (accession no. GSE161300). Additional sequencing and imaging data are available on Dropbox at https://www.dropbox.com/sh/mmeg3mckrpridu3/AAALBaMLoJsJiQC2-lrVY0Cva?dl=0 and upon reasonable request to the corresponding author.

Code availability

Software used to segment cells and quantify RNA spots is available at https://github.com/arjunrajlaboratory/rajlabimagetools. Software used to stitch, segment and quantify scan images of resistant colonies is available at https://github.com/arjunrajlaboratory/colonycounting_v2. Additional custom image analysis scripts are available at https://github.com/arjunrajlaboratory/timemachineimageanalysis. The pipeline used for barcode sequencing analysis is available at https://github.com/arjunrajlaboratory/timemachine.

References

  1. Symmons, O. & Raj, A. What’s luck got to do with it: single cells, multiple fates, and biological nondeterminism. Mol. Cell 62, 788–802 (2016).

    Article  CAS  Google Scholar 

  2. Raj, A. & van Oudenaarden, A. Nature, nurture, or chance: stochastic gene expression and its consequences. Cell 135, 216–226 (2008).

    Article  CAS  Google Scholar 

  3. 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 

  4. Sharma, S. V. et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80 (2010).

    Article  CAS  Google Scholar 

  5. Roesch, A. et al. A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell 141, 583–594 (2010).

    Article  CAS  Google Scholar 

  6. Shaffer, S. M. et al. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature 546, 431–435 (2017).

    Article  CAS  Google Scholar 

  7. Shaffer, S. M. et al. Memory sequencing reveals heritable single-cell gene expression programs associated with distinct cellular behaviors. Cell 182, 947–959 (2020)

  8. Cohen, A. A. et al. Dynamic proteomics of individual cancer cells in response to a drug. Science 322, 1511–1516 (2008).

    Article  CAS  Google Scholar 

  9. Spencer, S., Gaudet, S., Albeck, J., Burke, J. & Sorger, P. Non-genetic origins of cell-to-cell variability in TRAIL-induced apoptosis. Nature 459, 428–432 (2009).

  10. Sigal, A. et al. Variability and memory of protein levels in human cells. Nature 444, 643–646 (2006).

    Article  CAS  Google Scholar 

  11. Frieda, K. L. et al. Synthetic recording and in situ readout of lineage information in single cells. Nature 541, 107–111 (2016).

  12. Raj, B. et al. Simultaneous single-cell profiling of lineages and cell types in the vertebrate brain. Nat. Biotechnol. 36, 442–450 (2018).

    Article  CAS  Google Scholar 

  13. Alemany, A., Florescu, M., Baron, C. S., Peterson-Maduro, J. & van Oudenaarden, A. Whole-organism clone tracing using single-cell sequencing. Nature 556, 108–112 (2018).

    Article  CAS  Google Scholar 

  14. McKenna, A. et al. Whole organism lineage tracing by combinatorial and cumulative genome editing. Science 353, aaf7907 (2016).

    Article  Google Scholar 

  15. Kalhor, R. et al. Developmental barcoding of whole mouse via homing CRISPR. Science 361, eaat9804 (2018).

    Article  Google Scholar 

  16. Quinn, J. J. et al. Single-cell lineages reveal the rates, routes, and drivers of metastasis in cancer xenografts. Science https://doi.org/10.1126/science.abc1944 (2020).

  17. Biddy, B. A. et al. Single-cell mapping of lineage and identity in direct reprogramming. Nature 564, 219–224 (2018).

    Article  CAS  Google Scholar 

  18. Weinreb, C., Rodriguez-Fraticelli, A., Camargo, F. D. & Klein, A. M. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science 367, eaaw3381 (2020).

    Article  CAS  Google Scholar 

  19. Al’Khafaji, A. et al. Expressed barcodes enable clonal characterization of chemotherapeutic responses in chronic lymphocytic leukemia. Preprint at bioRxiv https://doi.org/10.1101/761981 (2019).

  20. Hurley, K. et al. Reconstructed single-cell fate trajectories define lineage plasticity windows during differentiation of human PSC-derived distal lung progenitors. Cell Stem Cell 26, 593–608 (2020).

  21. Al’Khafaji, A. M., Deatherage, D. & Brock, A. Control of lineage-specific gene expression by functionalized gRNA barcodes. ACS Synth. Biol. 7, 2468–2474 (2018).

    Article  Google Scholar 

  22. Feldman, D. et al. CloneSifter: enrichment of rare clones from heterogeneous cell populations. BMC Biol. 18, 177 (2020).

  23. Rebbeck, C. et al. SmartCodes: functionalized barcodes that enable targeted retrieval of clonal lineages from a heterogeneous population. Preprint at bioRxiv https://doi.org/10.1101/352617 (2018).

  24. Umkehrer, C. et al. Isolating live cell clones from barcoded populations using CRISPRa-inducible reports. Nat. Biotechnol. https://doi.org/10.1038/s41587-020-0614-0 (2020).

  25. Torre, E. A. et al. Genetic screening for single-cell variability modulators driving therapy resistance. Nat. Genet. 53, 76–85 (2021).

    Article  CAS  Google Scholar 

  26. Müller, J. et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat. Commun. 5, 5712 (2014).

    Article  Google Scholar 

  27. Gerosa, L. et al. Receptor-driven ERK pulses reconfigure MAPK signaling and enable persistence of drug-adapted BRAF-mutant melanoma cells. Cell Syst. 11, 478–494.e9 (2020).

    Article  CAS  Google Scholar 

  28. Schuh, L. et al. Gene networks with transcriptional bursting recapitulate rare transient coordinated high expression states in cancer. Cell Syst. 10, 363–378 (2020).

    Article  CAS  Google Scholar 

  29. Sun, C. et al. Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature 508, 118–122 (2014).

    Article  CAS  Google Scholar 

  30. Corcoran, R. B. et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2, 227–235 (2012).

    Article  CAS  Google Scholar 

  31. Ji, Z. et al. MITF modulates therapeutic resistance through EGFR signaling. J. Invest. Dermatol. 135, 1863–1872 (2015).

    Article  CAS  Google Scholar 

  32. Caron, A., Briscoe, D. M., Richard, D. & Laplante, M. DEPTOR at the nexus of cancer, metabolism, and immunity. Physiol. Rev. 98, 1765–1803 (2018).

    Article  Google Scholar 

  33. Lito, P. et al. Relief of profound feedback inhibition of mitogenic signaling by RAF inhibitors attenuates their activity in BRAFV600E melanomas. Cancer Cell 22, 668–682 (2012).

    Article  CAS  Google Scholar 

  34. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  Google Scholar 

  35. Hsu, M.-Y., Elder, D. E. & Herlyn, M. Melanoma: the Wistar melanoma (WM) cell lines. In: Human Cell Culture 259–274 (Springer, 2002).

  36. Zorita, E., Cuscó, P. & Filion, G. J. Starcode: sequence clustering based on all-pairs search. Bioinformatics 31, 1913–1919 (2015).

    Article  CAS  Google Scholar 

  37. Choi, H. M. T. et al. Third-generation in situ hybridization chain reaction: multiplexed, quantitative, sensitive, versatile, robust. Development 145, dev165753 (2018).

    Article  Google Scholar 

  38. Rouhanifard, S. H. et al. ClampFISH detects individual nucleic acid molecules using click chemistry-based amplification. Nat. Biotechnol. 37, 84–89 (2018).

  39. Raj, A., van den Bogaard, P., Rifkin, S. A., van Oudenaarden, A. & Tyagi, S. Imaging individual mRNA molecules using multiple singly labeled probes. Nat. Methods 5, 877–879 (2008).

    Article  CAS  Google Scholar 

  40. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  Google Scholar 

  41. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  Google Scholar 

  42. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2012).

    Article  Google Scholar 

  43. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  Google Scholar 

  44. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

We thank C. Bartman, A. Anguierra, J. Murray, N. Zhang, L. Cai, B. Stanger, A. Coté, K. Kiani, E. Sanford and Y. Goyal, along with other members of the Raj laboratory, for many useful suggestions. We thank the Flow Cytometry Core Laboratory at the Children’s Hospital of Philadelphia Research Institute for assistance in designing and performing FACS, including F. Tuluc for several helpful discussions. B.L.E. acknowledges support from NIH training grants F30 CA236129, T32 GM007170 and T32 HG000046; E.A.T. acknowledges support from R01 CA238237; I.P.D. acknowledges support from NIH 4DN U01 HL129998 and the NIH Center for Photogenomics (RM1 HG007743); C.L.J. acknowledges support from NIH T32 DK007780 and F30 HG010822; N.J. acknowledges support from NIH F30 HD103378; S.M.S. acknowledges support from DP5 OD028144; and A.R. acknowledges support from R01 CA238237, NIH Director’s Transformative Research Award R01 GM137425, R01 CA232256, NSF CAREER 1350601, P30 CA016520, SPORE P50 CA174523, NIH U01 CA227550, NIH 4DN U01 HL129998, the NIH Center for Photogenomics (RM1 HG007743) and the Tara Miller Foundation.

Author information

Authors and Affiliations

Authors

Contributions

B.L.E. and A.R. conceived and designed the project with input from E.A.T. and S.M.S. B.L.E. performed experiments and analyses. I.P.D., C.L.J. and N.J. assisted with optimizing barcode RNA FISH protocols. C.J.C. assisted with cell and colony segmentation. A.R. supervised the project. B.L.E. and A.R. wrote the manuscript with feedback from S.M.S., C.L.J., N.J. and other members of the Raj laboratory.

Corresponding author

Correspondence to Arjun Raj.

Ethics declarations

Competing interests

A.R. receives consulting income, and A.R. and S.M.S. receive royalties, related to Stellaris RNA FISH probes.

Additional information

Peer review information Nature Biotechnology thanks Leor Weinberger 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.

Supplementary information

Supplementary Information

Supplementary Figs. 1–16.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–8

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Emert, B.L., Cote, C.J., Torre, E.A. et al. Variability within rare cell states enables multiple paths toward drug resistance. Nat Biotechnol 39, 865–876 (2021). https://doi.org/10.1038/s41587-021-00837-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-021-00837-3

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