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:

Spatial transcriptomic analysis of cryosectioned tissue samples with Geo-seq

This article has been updated

Abstract

Conventional gene expression studies analyze multiple cells simultaneously or single cells, for which the exact in vivo or in situ position is unknown. Although cellular heterogeneity can be discerned when analyzing single cells, any spatially defined attributes that underpin the heterogeneous nature of the cells cannot be identified. Here, we describe how to use geographical position sequencing (Geo-seq), a method that combines laser capture microdissection (LCM) and single-cell RNA-seq technology. The combination of these two methods enables the elucidation of cellular heterogeneity and spatial variance simultaneously. The Geo-seq protocol allows the profiling of transcriptome information from only a small number cells and retains their native spatial information. This protocol has wide potential applications to address biological and pathological questions of cellular properties such as prospective cell fates, biological function and the gene regulatory network. Geo-seq has been applied to investigate the spatial transcriptome of mouse early embryo, mouse brain, and pathological liver and sperm tissues. The entire protocol from tissue collection and microdissection to sequencing requires 5 d, Data analysis takes another 1 or 2 weeks, depending on the amount of data and the speed of the processor.

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: Geo-seq workflow.
Figure 2: Inclusion of the decidual tissue improves tissue integrity of the embryo cryosections.
Figure 3: PFA fixation decreases RNA quality severely.
Figure 4: Comparison of different staining methods.
Figure 5: Comparison of RNA quality of samples prepared under different lysis conditions.
Figure 6: Effect of different bead/DNA ratios on cDNA purification.
Figure 7: Laser microdissection of cryosections of gastrula-stage mouse embryo (corresponding to Step 19).
Figure 8: Real-time PCR amplification plot of GAPDH in representative LCM samples (corresponding to Step 42).
Figure 9: Bioanalyzer electropherogram of cDNA libraries (corresponding to Step 57).
Figure 10: Examples of Geo-seq results showing high spatial resolution of gene expression pattern (displayed as corn plots) in the mid-gastrula-stage mouse embryo (corresponding to Step 77).

Similar content being viewed by others

Change history

  • 13 April 2017

    In the version of this article initially published, Shengbao Suo was not indicated as contributing equally to the work with Jun Chen; Jing-Dong J. Han was not indicated as a corresponding author; the expanded form of "Geo-seq" was not given in the abstract; in the heading prior to Step 77, it was not stated that the time indicated excludes the time required for the design of 2D corn plots and for zip-code mapping; "Corn plot visualization and zip-code mapping can be performed through the iTranscriptome portal at http://www.itranscriptome.org." was omitted from the end of Step 77; and some grant numbers were omitted from the Acknowledgments section. These omissions have been corrected in the HTML and PDF versions of the article.

References

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

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Cann, G.M. et al. mRNA-Seq of single prostate cancer circulating tumor cells reveals recapitulation of gene expression and pathways found in prostate cancer. PLoS One 7, e49144 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Shalek, A.K. et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature 498, 236–240 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. van Wolfswinkel, J.C., Wagner, D.E. & Reddien, P.W. Single-cell analysis reveals functionally distinct classes within the planarian stem cell compartment. Cell Stem Cell 15, 326–339 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Guo, G. et al. Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev. Cell 18, 675–685 (2010).

    CAS  PubMed  Google Scholar 

  7. Yan, L. et al. Single-cell RNA-Seq profiling of human preimplantation embryos and embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1131–1139 (2013).

    CAS  PubMed  Google Scholar 

  8. Durruthy-Durruthy, R. et al. Reconstruction of the mouse otocyst and early neuroblast lineage at single-cell resolution. Cell 157, 964–978 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Xue, Z. et al. Genetic programs in human and mouse early embryos revealed by single-cell RNA sequencing. Nature 500, 593–597 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Hebenstreit, D. Methods, challenges and potentials of single cell RNA-seq. Biology 1, 658–667 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Tang, F. et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009).

    CAS  PubMed  Google Scholar 

  12. Tang, F. et al. RNA-Seq analysis to capture the transcriptome landscape of a single cell. Nat. Protoc. 5, 516–535 (2010).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  14. Islam, S. et al. Highly multiplexed and strand-specific single-cell RNA 5′ end sequencing. Nat. Protoc. 7, 813–828 (2012).

    CAS  PubMed  Google Scholar 

  15. Goetz, J.J. & Trimarchi, J.M. Transcriptome sequencing of single cells with Smart-Seq. Nat. Biotechnol. 30, 763–765 (2012).

    CAS  PubMed  Google Scholar 

  16. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    CAS  PubMed  Google Scholar 

  17. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    CAS  PubMed  Google Scholar 

  18. Wen, L. & Tang, F. Single-cell sequencing in stem cell biology. Genome Biol. 17, 71 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  20. Liu, A. Laser capture microdissection in the tissue biorepository. J. Biomol. Tech. 21, 120–125 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Datta, S. et al. Laser capture microdissection: big data from small samples. Histol. Histopathol. 30, 1255–1269 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Canas, R.A., Canales, J., Gomez-Maldonado, J., Avila, C. & Canovas, F.M. Transcriptome analysis in maritime pine using laser capture microdissection and 454 pyrosequencing. Tree Physiol. 34, 1278–1288 (2014).

    CAS  PubMed  Google Scholar 

  23. Erickson, H.S. et al. Quantitative RT-PCR gene expression analysis of laser microdissected tissue samples. Nat. Protoc. 4, 902–922 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Morrison, J.A., Bailey, C.M. & Kulesa, P.M. Gene profiling in the avian embryo using laser capture microdissection and RT-qPCR. Cold Spring Harbor Protoc. 2012, 1249–1262 (2012).

    Google Scholar 

  25. Grover, P.K., Cummins, A.G., Price, T.J., Roberts-Thomson, I.C. & Hardingham, J.E. A simple, cost-effective and flexible method for processing of snap-frozen tissue to prepare large amounts of intact RNA using laser microdissection. Biochimie 94, 2491–2497 (2012).

    CAS  PubMed  Google Scholar 

  26. Zechel, S., Zajac, P., Lonnerberg, P., Ibanez, C.F. & Linnarsson, S. Topographical transcriptome mapping of the mouse medial ganglionic eminence by spatially resolved RNA-seq. Genome Biol. 15, 486 (2014).

    PubMed  PubMed Central  Google Scholar 

  27. Bandyopadhyay, U., Fenton, W.A., Horwich, A.L. & Nagy, M. Production of RNA for transcriptomic analysis from mouse spinal cord motor neuron cell bodies by laser capture microdissection. J. Vis. Exp. 83, e51168 (2014).

    Google Scholar 

  28. Peng, G. et al. Spatial transcriptome for the molecular annotation of lineage fates and cell identity in mid-gastrula mouse embryo. Dev. Cell 36, 681–697 (2016).

    CAS  PubMed  Google Scholar 

  29. Cox, M.L. et al. Assessment of fixatives, fixation, and tissue processing on morphology and RNA integrity. Exp. Mol. Pathol. 80, 183–191 (2006).

    CAS  PubMed  Google Scholar 

  30. Goldsworthy, S.M., Stockton, P.S., Trempus, C.S., Foley, J.F. & Maronpot, R.R. Effects of fixation on RNA extraction and amplification from laser capture microdissected tissue. Mol. Carcinog. 25, 86–91 (1999).

    CAS  PubMed  Google Scholar 

  31. Wang, W.Z., Oeschger, F.M., Lee, S. & Molnar, Z. High quality RNA from multiple brain regions simultaneously acquired by laser capture microdissection. BMC Mol. Biol. 10, 69 (2009).

    PubMed  PubMed Central  Google Scholar 

  32. Parlato, R. et al. A preservation method that allows recovery of intact RNA from tissues dissected by laser capture microdissection. Anal. Biochem. 300, 139–145 (2002).

    CAS  PubMed  Google Scholar 

  33. Wang, H. et al. Histological staining methods preparatory to laser capture microdissection significantly affect the integrity of the cellular RNA. BMC Genomics 7, 97 (2006).

    PubMed  PubMed Central  Google Scholar 

  34. Sonne, S.B. et al. Optimizing staining protocols for laser microdissection of specific cell types from the testis including carcinoma in situ. PLoS One 4, e5536 (2009).

    PubMed  PubMed Central  Google Scholar 

  35. Clement-Ziza, M., Munnich, A., Lyonnet, S., Jaubert, F. & Besmond, C. Stabilization of RNA during laser capture microdissection by performing experiments under argon atmosphere or using ethanol as a solvent in staining solutions. RNA 14, 2698–2704 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Chomczynski, P. & Sacchi, N. The single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction: twenty-something years on. Nat. Protoc. 1, 581–585 (2006).

    CAS  PubMed  Google Scholar 

  37. Bengtsson, M., Hemberg, M., Rorsman, P. & Stahlberg, A. Quantification of mRNA in single cells and modelling of RT-qPCR induced noise. BMC Mol. Biol. 9, 63 (2008).

    PubMed  PubMed Central  Google Scholar 

  38. Trombetta, J.J. et al. Preparation of single-cell RNA-seq libraries for next generation sequencing. Curr. Protoc. Mol. Biol. 107, 4.22.1–4.22.17 (2014).

    Google Scholar 

  39. Miller, D.F. et al. A new method for stranded whole transcriptome RNA-seq. Methods 63, 126–134 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Achim, K. et al. High-throughput spatial mapping of single-cell RNA-seq data to tissue of origin. Nat. Biotechnol. 33, 503–509 (2015).

    CAS  PubMed  Google Scholar 

  41. Satija, R., Farrell, J.A., Gennert, D., Schier, A.F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Durruthy-Durruthy, R., Gottlieb, A. & Heller, S. 3D computational reconstruction of tissues with hollow spherical morphologies using single-cell gene expression data. Nat. Protoc. 10, 459–474 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Junker, J.P. et al. Genome-wide RNA tomography in the zebrafish embryo. Cell 159, 662–675 (2014).

    CAS  PubMed  Google Scholar 

  44. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  45. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Huber, W., von Heydebreck, A., Sultmann, H., Poustka, A. & Vingron, M. Variance stabilization applied to microarray data calibration and to the quantification of differential expression. Bioinformatics 18, S96–104 (2002).

    PubMed  Google Scholar 

  47. Zhang, W. et al. Integrating genomic, epigenomic, and transcriptomic features reveals modular signatures underlying poor prognosis in ovarian cancer. Cell Rep. 4, 542–553 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to Q. Zhou (Institute of Zoology, Chinese Academy of Sciences) for helpful suggestions, and to Y. Qian of the Jing laboratory for technical support. The sequencing was performed on an Illumina HiSeq2500 system by BerryGenomics (Beijing, China). This work was supported in part by the 'Strategic Priority Research Program' of the Chinese Academy of Sciences (XDA01010201 to N.J., XDB19020301 and XDA01010303 to J.-D.J.H.), the National Key Basic Research and Development Program of China (2014CB964804 and 2015CB964500 to N.J., and 2015CB964803 and 2016YFE0108700 to J.-D.J.H.) and the National Natural Science Foundation of China (31430058, 31571513, 31630043, 91519314 to N.J., and 91329302, 31210103916, 91519330 to J.-D.J.H.).

Author information

Authors and Affiliations

Authors

Contributions

N.J. supervised the project. N.J. and G.P. designed the experiment. J.C. and G.P. performed the laser microdissection and RNA-seq. J.C. and S.S. wrote the manuscript. G.P., P.P.L.T., J.-D.J.H. and N.J. edited the manuscript.

Corresponding authors

Correspondence to Jing-Dong J Han, Guangdun Peng or Naihe Jing.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Methods

Supplementary Methods (PDF 421 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, J., Suo, S., Tam, P. et al. Spatial transcriptomic analysis of cryosectioned tissue samples with Geo-seq. Nat Protoc 12, 566–580 (2017). https://doi.org/10.1038/nprot.2017.003

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2017.003

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