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.

Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues

Abstract

RNA-sequencing (RNA-seq) measures the quantitative change in gene expression over the whole transcriptome, but it lacks spatial context. In contrast, in situ hybridization provides the location of gene expression, but only for a small number of genes. Here we detail a protocol for genome-wide profiling of gene expression in situ in fixed cells and tissues, in which RNA is converted into cross-linked cDNA amplicons and sequenced manually on a confocal microscope. Unlike traditional RNA-seq, our method enriches for context-specific transcripts over housekeeping and/or structural RNA, and it preserves the tissue architecture for RNA localization studies. Our protocol is written for researchers experienced in cell microscopy with minimal computing skills. Library construction and sequencing can be completed within 14 d, with image analysis requiring an additional 2 d.

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: Schematic overview of FISSEQ library construction and sequencing.
Figure 2: Comparing single-cell RNA-seq with FISSEQ.
Figure 3: Counting resolution-limited amplicons using partition sequencing.
Figure 4: Schematic overview of the SOLiD color-coding and decoding scheme.
Figure 5: Example of image analysis, registration and sequence clustering.
Figure 6: Schematic overview of FISSEQ experimental and analysis steps.

References

  1. Rifai, N., Gillette, M.A. & Carr, S.A. Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat. Biotechnol. 24, 971–983 (2006).

    Article  CAS  Google Scholar 

  2. Jaitin, D.A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  Google Scholar 

  3. Battich, N., Stoeger, T. & Pelkmans, L. Image-based transcriptomics in thousands of single human cells at single-molecule resolution. Nat. Methods 10, 1127–1133 (2013).

    Article  CAS  Google Scholar 

  4. Lein, E.S. et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 445, 168–176 (2007).

    Article  CAS  Google Scholar 

  5. Zeng, H. et al. Large-scale cellular-resolution gene profiling in human neocortex reveals species-specific molecular signatures. Cell 149, 483–496 (2012).

    Article  CAS  Google Scholar 

  6. Diez-Roux, G. et al. A high-resolution anatomical atlas of the transcriptome in the mouse embryo. PLoS Biol. 9, e1000582 (2011).

    Article  CAS  Google Scholar 

  7. Femino, A.M., Fay, F.S., Fogarty, K. & Singer, R.H. Visualization of single RNA transcripts in situ. Science 280, 585–590 (1998).

    Article  CAS  Google Scholar 

  8. 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 

  9. Levsky, J.M., Shenoy, S.M., Pezo, R.C. & Singer, R.H. Single-cell gene expression profiling. Science 297, 836–840 (2002).

    Article  CAS  Google Scholar 

  10. Lubeck, E., Coskun, A.F., Zhiyentayev, T., Ahmad, M. & Cai, L. Single-cell in situ RNA profiling by sequential hybridization. Nat. Methods 11, 360–361 (2014).

    Article  CAS  Google Scholar 

  11. Choi, H.M. et al. Programmable in situ amplification for multiplexed imaging of mRNA expression. Nat. Biotechnol. 28, 1208–1212 (2011).

    Article  Google Scholar 

  12. Ke, R. et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods 10, 857–860 (2013).

    Article  CAS  Google Scholar 

  13. Larsson, C., Grundberg, I., Söderberg, O. & Nilsson, M. In situ detection and genotyping of individual mRNA molecules. Nat. Methods 7, 395–397 (2010).

    Article  CAS  Google Scholar 

  14. Larsson, C. et al. In situ genotyping individual DNA molecules by target-primed rolling-circle amplification of padlock probes. Nat. Methods 1, 227–232 (2004).

    Article  CAS  Google Scholar 

  15. Lagunavicius, A. et al. Novel application of Phi29 DNA polymerase: RNA detection and analysis in vitro and in situ by target RNA-primed RCA. RNA 15, 765–771 (2009).

    Article  CAS  Google Scholar 

  16. Merkiene, E., Gaidamaviciute, E., Riauba, L., Janulaitis, A. & Lagunavicius, A. Direct detection of RNA in vitro and in situ by target-primed RCA: the impact of E. coli RNase III on the detection efficiency of RNA sequences distanced far from the 3′-end. RNA 16, 1508–1515 (2010).

    Article  CAS  Google Scholar 

  17. Lee, J.H. et al. A robust approach to identifying tissue-specific gene expression regulatory variants using personalized human induced pluripotent stem cells. PLoS Genet. 5, e1000718 (2009).

    Article  Google Scholar 

  18. Shendure, J. et al. Accurate multiplex polony sequencing of an evolved bacterial genome. Science 309, 1728–1732 (2005).

    Article  CAS  Google Scholar 

  19. Drmanac, R. et al. Human genome sequencing using unchained base reads on self-assembling DNA nanoarrays. Science 327, 78–81 (2010).

    Article  CAS  Google Scholar 

  20. Grun, D., Kester, L. & van Oudenaarden, A. Validation of noise models for single-cell transcriptomics. Nat. Methods 11, 637–640 (2014).

    Article  Google Scholar 

  21. Shapiro, E., Biezuner, T. & Linnarsson, S. Single-cell sequencing-based technologies will revolutionize whole-organism science. Nat. Rev. Genet. 14, 618–630 (2013).

    Article  CAS  Google Scholar 

  22. Islam, S. et al. Quantitative single-cell RNA-seq with unique molecular identifiers. Nat. Methods 11, 163–166 (2014).

    Article  CAS  Google Scholar 

  23. Lee, J.H. et al. Highly multiplexed subcellular RNA sequencing in situ. Science 343, 1360–1363 (2014).

    Article  CAS  Google Scholar 

  24. Adiconis, X. et al. Comparative analysis of RNA sequencing methods for degraded or low-input samples. Nat. Methods 10, 623–629 (2013).

    Article  CAS  Google Scholar 

  25. Yachida, S. et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467, 1114–1117 (2010).

    Article  CAS  Google Scholar 

  26. Frumkin, D. et al. Amplification of multiple genomic loci from single cells isolated by laser micro-dissection of tissues. BMC Biotechnol. 8, 17 (2008).

    Article  Google Scholar 

  27. Lovatt, D. et al. Transcriptome in vivo analysis (TIVA) of spatially defined single cells in live tissue. Nat. Methods 11, 190–196 (2014).

    Article  CAS  Google Scholar 

  28. Schmid, M.W. et al. A powerful method for transcriptional profiling of specific cell types in eukaryotes: laser-assisted microdissection and RNA sequencing. PLoS ONE 7, e29685 (2012).

    Article  CAS  Google Scholar 

  29. Islam, S. et al. Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq. Genome Res. 21, 1160–1167 (2011).

    Article  CAS  Google Scholar 

  30. Ramskold, D. et al. Full-length mRNA-seq from single-cell levels of RNA and individual circulating tumor cells. Nat. Biotechnol. 30, 777–782 (2012).

    Article  Google Scholar 

  31. Hashimshony, T., Wagner, F., Sher, N. & Yanai, I. CEL-seq: single-cell RNA-seq by multiplexed linear amplification. Cell Rep. 2, 666–673 (2012).

    Article  CAS  Google Scholar 

  32. Avital, G., Hashimshony, T. & Yanai, I. Seeing is believing: new methods for in situ single-cell transcriptomics. Genome Biol. 15, 110 (2014).

    Article  Google Scholar 

  33. Buxbaum, A.R., Wu, B. & Singer, R.H. Single β-actin mRNA detection in neurons reveals a mechanism for regulating its translatability. Science 343, 419–422 (2014).

    Article  CAS  Google Scholar 

  34. Hanna, J. et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature 462, 595–601 (2009).

    Article  CAS  Google Scholar 

  35. Buganim, Y. et al. Single-cell expression analyses during cellular reprogramming reveal an early stochastic and a late hierarchic phase. Cell 150, 1209–1222 (2012).

    Article  CAS  Google Scholar 

  36. Itzkovitz, S., Blat, I.C., Jacks, T., Clevers, H. & van Oudenaarden, A. Optimality in the development of intestinal crypts. Cell 148, 608–619 (2012).

    Article  CAS  Google Scholar 

  37. Hansen, C.H. & van Oudenaarden, A. Allele-specific detection of single mRNA molecules in situ. Nat. Methods 10, 869–871 (2013).

    Article  CAS  Google Scholar 

  38. Porreca, G.J. et al. Multiplex amplification of large sets of human exons. Nat. Methods 4, 931–936 (2007).

    Article  CAS  Google Scholar 

  39. Kosuri, S. et al. Composability of regulatory sequences controlling transcription and translation in Escherichia coli. Proc. Natl. Acad. Sci. USA 110, 14024–14029 (2013).

    Article  CAS  Google Scholar 

  40. Li, J.B. et al. Genome-wide identification of human RNA editing sites by parallel DNA capturing and sequencing. Science 324, 1210–1213 (2009).

    Article  CAS  Google Scholar 

  41. Zhang, K. et al. Digital RNA allelotyping reveals tissue-specific and allele-specific gene expression in human. Nat. Methods 6, 613–618 (2009).

    Article  CAS  Google Scholar 

  42. Michael, W.M., Choi, M. & Dreyfuss, G. A nuclear export signal in hnRNP A1: a signal-mediated, temperature-dependent nuclear protein export pathway. Cell 83, 415–422 (1995).

    Article  CAS  Google Scholar 

  43. Kaposi-Novak, P., Lee, J.S., Mikaelyan, A., Patel, V. & Thorgeirsson, S.S. Oligonucleotide microarray analysis of aminoallyl-labeled cDNA targets from linear RNA amplification. Biotechniques 37, 580, 582–586, 588 (2004).

    Article  Google Scholar 

  44. Nanda, J.S. & Lorsch, J.R. Labeling a protein with fluorophores using NHS ester derivitization. Methods Enzymol. 536, 87–94 (2014).

    Article  CAS  Google Scholar 

  45. Mardis, E.R. Next-generation DNA sequencing methods. Annu. Rev. Genomics Hum. Genet. 9, 387–402 (2008).

    Article  CAS  Google Scholar 

  46. Massingham, T. & Goldman, N. Error-correcting properties of the SOLiD Exact Call Chemistry. BMC Bioinformatics 13, 145 (2012).

    Article  Google Scholar 

  47. Applied Biosystems. SOLiD System Accuracy with the Exact Call Chemistry Module (https://www3.appliedbiosystems.com/cms/groups/global_marketing_group/documents/generaldocuments/cms_091372.pdf).

  48. Itzkovitz, S. & van Oudenaarden, A. Validating transcripts with probes and imaging technology. Nat. Methods 8, S12–S19 (2011).

    Article  CAS  Google Scholar 

  49. Eliceiri, K.W. et al. Biological imaging software tools. Nat. Methods 9, 697–710 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  51. Kankaanpaa, P. et al. BioImageXD: an open, general-purpose and high-throughput image-processing platform. Nat. Methods 9, 683–689 (2012).

    Article  Google Scholar 

  52. Pawley, J.B. Handbook of Biological Confocal Microscopy 3rd edn. (Springer, 2006).

Download references

Acknowledgements

This study was funded by US National Institutes of Health (NIH) Centers of Excellence in Genomic Sciences (CEGS) grant no. P50 HG005550. J.H.L. and co-workers were funded by National Heart, Blood and Lung Institute (NHBLI) grant no. RC2HL102815, by the Allen Institute for Brain Science and by National Institute of Mental Health (NIMH) grant no. MH098977. E.R.D. was funded by NIH grant no. GM080177 and by National Science Foundation (NSF) Graduate Research Fellowship grant no. DGE1144152.

Author information

Authors and Affiliations

Authors

Contributions

J.H.L. and E.R.D. conceived FISSEQ library construction, sequencing, image analysis and bioinformatics. J.S., R.K., J.L.Y., B.M.T., H.S.L. and J.A. provided key feedbacks during the FISSEQ method development. R.T. and T.C.F. assisted with automated microscopy and image analysis. K.Z. and G.M.C. oversaw the project. J.H.L. wrote the paper, and E.R.D. wrote the FISSEQ software.

Corresponding authors

Correspondence to Je Hyuk Lee or George M Church.

Ethics declarations

Competing interests

Potential conflicts of interest for G.M.C. are listed on http://arep.med.harvard.edu/gmc/tech.html. Other authors have no conflicts of interest.

Integrated supplementary information

Supplementary Figure 1 The altered distribution of poly A-associated transcripts at different fixation temperatures.

(a) Primary fibroblasts were fixed at room temperature or at 37°C for 10 minutes. The cells were then hybridized to the Cy3 Poly dT(50) or 18S rRNA probe. At room temperature a large fraction of the mRNA is retained in the nucleus. (b) The cold shock-induced nuclear retention of poly A-associated transcripts is cell type-specific. Highly transformed cell lines and iPS cell lines do not appear to be affected by the fixation temperature.

Supplementary Figure 2 Acid-treatment improves cell permeabilization for FISSEQ.

(a) Low amplicon counts are frequently due to poor permeabilization, which can be observed in the axial view of human iPS cells. (b) A brief treatment with 0.1 HCl can significantly improve the uniformity of FISSEQ amplicons throughout the cell, especially in iPS cells.

Supplementary Figure 3 Degradation of residual RNA is essential for FISSEQ amplicon generation.

Far more amplicons are observed after the RNase-treatment before circularization in numerous human iPS cell colonies (labeled with mCherry; bar: 1 μm).

Supplementary Figure 4 Image deconvolution of FISSEQ amplicons in fibroblasts.

(a) A high quality confocal image requires minimal image deconvolution, but doing so can dramatically increase the signal to noise ratio by removing many spurious pixels; however, over-deconvolution can actually lead to more background noise or artifacts (bar: 1 μm). (b) A low quality epifluorescence image can contain a significant amount of out of focus light. Using 3D deconvolution to remove such signal can dramatically improve the image and speed up base calling (bar: 1 μm).

Supplementary Figure 5 Image alignment of FISSEQ amplicons in fibroblasts.

(a) One of the adjustable parameter is the block size for local alignment. For large images (i.e. 4000-by-4000 pixels) dividing images into a 10-by-10 grid is essential. Even for small images one can appreciate the effect of increasing the block number. (b) When using local alignment it is important to specify the degree of block overlap for image stitching. A large image correction with insufficient or no overlap will result in image fragmentation. (c) In general sub-pixel registration accuracy will not affect alignment significantly; however, increasing the upsampling factor can lead to better registration in certain instances.

Supplementary Figure 6 Data analysis using RStudio.

(a) RStudio provides a straightforward GUI for importing, analyzing and visualizing FISSEQ data. We provide several sample datasets and R sessions for FISSEQ data analysis. One can view the history window and click through commands to reproduce the following plots. (b) Correlation plot of gene expression frequency in region 1 and region 2. (c) Frequency histogram of RNA class instances. (d) Distribution of cluster sizes. (e) Area plot of non-ribosomal reads and their location from 5 different wound healing regions.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, J., Daugharthy, E., Scheiman, J. et al. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat Protoc 10, 442–458 (2015). https://doi.org/10.1038/nprot.2014.191

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2014.191

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

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

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