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.

  • Protocol
  • Published:

Assessment of engineered cells using CellNet and RNA-seq

Abstract

CellNet is a computational platform designed to assess cell populations engineered by either directed differentiation of pluripotent stem cells (PSCs) or direct conversion, and to suggest specific hypotheses to improve cell fate engineering protocols. CellNet takes as input gene expression data and compares them with large data sets of normal expression profiles compiled from public sources, in regard to the extent to which cell- and tissue-specific gene regulatory networks are established. CellNet was originally designed to work with human or mouse microarray expression data for 21 cell or tissue (C/T) types. Here we describe how to apply CellNet to RNA-seq data and how to build a completely new CellNet platform applicable to, for example, other species or additional cell and tissue types. Once the raw data have been preprocessed, running CellNet takes only several minutes, whereas the time required to create a completely new CellNet is several hours.

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: Inputs and outputs of CellNet.
Figure 2: Outline of the PROCEDURE.
Figure 3: Classification heatmap of the example query data.
Figure 4: C/T-specific GRN status of fibroblasts as they are reprogrammed to pluripotency.
Figure 5: Network influence score (NIS).
Figure 6: Precision recall curves for each murine RNA-seq C/T classifier.
Figure 7: Expression of C/T-specific genes.

Similar content being viewed by others

References

  1. Murry, C.E. & Keller, G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 132, 661–680 (2008).

    Article  CAS  Google Scholar 

  2. Ieda, M. et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142, 375–386 (2010).

    Article  CAS  Google Scholar 

  3. Kyba, M., Perlingeiro, R.C.R. & Daley, G.Q. HoxB4 confers definitive lymphoid-myeloid engraftment potential on embryonic stem cell and yolk sac hematopoietic progenitors. Cell 109, 29–37 (2002).

    Article  CAS  Google Scholar 

  4. Bock, C. et al. Reference maps of human ES and iPS cell variation enable high-throughput characterization of pluripotent cell lines. Cell 144, 439–452 (2011).

    Article  CAS  Google Scholar 

  5. McKinney-Freeman, S. et al. The transcriptional landscape of hematopoietic stem cell ontogeny. Cell Stem Cell 11, 701–714 (2012).

    Article  CAS  Google Scholar 

  6. Hussein, S.M.I. et al. Genome-wide characterization of the routes to pluripotency. Nature 516, 198–206 (2015).

    Article  Google Scholar 

  7. Davidson, E.H. & Erwin, D.H. Gene regulatory networks and the evolution of animal body plans. Science 311, 796–800 (2006).

    Article  CAS  Google Scholar 

  8. Cahan, P. et al. CellNet: network biology applied to stem cell engineering. Cell 158, 903–915 (2014).

    Article  CAS  Google Scholar 

  9. Morris, S.A. et al. Dissecting engineered cell types and enhancing cell fate conversion via CellNet. Cell 158, 889–902 (2014).

    Article  CAS  Google Scholar 

  10. Berger, D.R., Ware, B.R., Davidson, M.D., Allsup, S.R. & Khetani, S.R. Enhancing the functional maturity of iPSC-derived human hepatocytes via controlled presentation of cell-cell interactions in vitro. Hepatology 61, 1370–1381 (2014).

    Article  Google Scholar 

  11. Godoy, P. et al. Gene networks and transcription factor motifs defining the differentiation of stem cells into hepatocyte-like cells. J. Hepatol. 63, 934–942 (2015).

    Article  CAS  Google Scholar 

  12. Song, G. et al. Direct reprogramming of hepatic myofibroblasts into hepatocytes in vivo attenuates liver fibrosis. Cell Stem Cell 18, 797–808 (2016).

    Article  CAS  Google Scholar 

  13. Cao, N. et al. Conversion of human fibroblasts into functional cardiomyocytes by small molecules. Science 352, 1216–1220 (2016).

    Article  CAS  Google Scholar 

  14. Uosaki, H. et al. Transcriptional landscape of cardiomyocyte maturation. Cell Rep. 13, 1705–1716 (2015).

    Article  CAS  Google Scholar 

  15. Lu, Y.-F. et al. Engineered murine HSCs reconstitute multi-lineage hematopoiesis and adaptive immunity. Cell Rep. 17, 3178–3192 (2016).

    Article  CAS  Google Scholar 

  16. Pavlidis, N. & Fizazi, K. Carcinoma of unknown primary (CUP). Crit. Rev. Oncol. Hematol. 69, 271–278 (2009).

    Article  Google Scholar 

  17. Bian, Q. & Cahan, P. Computational tools for stem cell biology. Trends Biotechnol. 34, 993–1009 (2016).

    Article  CAS  Google Scholar 

  18. Müller, F.J. et al. A bioinformatic assay for pluripotency in human cells. Nat. Methods 8, 315–317 (2011).

    Article  Google Scholar 

  19. Avior, Y., Biancotti, J.-C. & Benvenisty, N. TeratoScore: assessing the differentiation potential of human pluripotent stem cells by quantitative expression analysis of teratomas. Stem Cell Reports 4, 967–974 (2015).

    Article  CAS  Google Scholar 

  20. Roost, M.S. et al. KeyGenes, a tool to probe tissue differentiation using a human fetal transcriptional atlas. Stem Cell Reports 4, 1112–1124 (2015).

    Article  CAS  Google Scholar 

  21. Rackham, O.J.L. et al. A predictive computational framework for direct reprogramming between human cell types. Nat. Genet. 48, 331–335 (2016).

    Article  CAS  Google Scholar 

  22. D'Alessio, A.C. et al. A systematic approach to identify candidate transcription factors that control cell identity. Stem Cell Reports 5, 763–775 (2015).

    Article  CAS  Google Scholar 

  23. Cieply, B. et al. Multiphasic and dynamic changes in alternative splicing during induction of pluripotency are coordinated by numerous RNA-binding proteins. Cell Rep. 15, 247–255 (2016).

    Article  CAS  Google Scholar 

  24. Mertens, J. et al. Differential responses to lithium in hyperexcitable neurons from patients with bipolar disorder. Nature 527, 95–99 (2015).

    Article  CAS  Google Scholar 

  25. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J. 17, 10–12 (2011).

    Article  Google Scholar 

  26. Patro, R., Duggal, G., Love, M.I., Irizarry, R.A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    Article  CAS  Google Scholar 

  27. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  Google Scholar 

  28. Mouse ENCODE Consortium. An encyclopedia of mouse DNA elements (Mouse ENCODE). Genome Biol. 13, 418 (2012).

  29. Xu, H. et al. ESCAPE: database for integrating high-content published data collected from human and mouse embryonic stem cells. Database 2013, bat045 (2013).

    PubMed  PubMed Central  Google Scholar 

  30. Correa-Cerro, L.S. et al. Generation of mouse ES cell lines engineered for the forced induction of transcription factors. Sci. Rep. 1, 167 (2011).

    Article  Google Scholar 

  31. Margolin, A.A. et al. Reverse engineering cellular networks. Nat. Protoc. 1, 662–671 (2006).

    Article  CAS  Google Scholar 

  32. Margolin, A.A. & Califano, A. Theory and limitations of genetic network inference from microarray data. Ann. N. Y. Acad. Sci. 1115, 51–72 (2007).

    Article  CAS  Google Scholar 

  33. Faith, J.J. et al. Large-scale mapping and validation of Escherichia coli transcriptional regulation from a compendium of expression profiles. PLoS Biol. 5, e8 (2007).

    Article  Google Scholar 

  34. Rosvall, M. & Bergstrom, C.T. Maps of random walks on complex networks reveal community structures. Proc. Natl. Acad. Sci. USA 105, 1118–1123 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

P.C. is supported by the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK; grant no. K01DK096013). We thank E. Appleton for helpful comments on the protocol.

Author information

Authors and Affiliations

Authors

Contributions

A.H.R. wrote code, performed analysis, and wrote the manuscript. R.M.S. wrote code and performed analysis. Y.T. analyzed data, debugged code, and edited the manuscript. J.K. debugged code and analyzed data. E.K.W.L. analyzed data and edited the manuscript. P.C. devised the method, wrote code, analyzed data, wrote the manuscript, and oversaw the project.

Corresponding author

Correspondence to Patrick Cahan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Comparison of GRN performance based on either total counts normalization or DESeq.

X-axis represents the Z-score for the predicted transcription factor- to target genes interactions. The Y-axis represents the area under the precision recall curve relative to randomly generated GRNs. AUPR was calculated as described previously8 using three sets of TF-to-target gene annotations as gold standards. The first gold standard is derived from lists of genes whose promoters are bound by transcription factors as determined by Chip-Seq data produced as part of the mouse ENCODE project28. The second gold standard is the Escape database, which is a compilation of genes whose promoters are bound by transcription factors in mouse embryonic stem cells defined by Chip-Chip or Chip-Seq data29. The third gold standard is derived from the determination of genes that are differentially expressed upon acute induction of one of 94 transcription factors ('Ko': named after the surname of the senior author of the associated study30).

Supplementary information

Supplementary Figures and Text

Supplementary Figure 1 and Supplementary Tables 1 and 2. (PDF 725 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Radley, A., Schwab, R., Tan, Y. et al. Assessment of engineered cells using CellNet and RNA-seq. Nat Protoc 12, 1089–1102 (2017). https://doi.org/10.1038/nprot.2017.022

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2017.022

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 AI and Robotics

Sign up for the Nature Briefing: AI and Robotics newsletter — what matters in AI and robotics research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: AI and Robotics