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:

Tracing DNA paths and RNA profiles in cultured cells and tissues with ORCA

Abstract

Chromatin conformation capture (3C) methods and fluorescent in situ hybridization (FISH) microscopy have been used to investigate the spatial organization of the genome. Although powerful, both techniques have limitations. Hi-C is challenging for low cell numbers and requires very deep sequencing to achieve its high resolution. In contrast, FISH can be done on small cell numbers and capture rare cell populations, but typically targets pairs of loci at a lower resolution. Here we detail a protocol for optical reconstruction of chromatin architecture (ORCA), a microscopy approach to trace the 3D DNA path within the nuclei of fixed tissues and cultured cells with a genomic resolution as fine as 2 kb and a throughput of ~10,000 cells per experiment. ORCA can identify structural features with comparable resolution to Hi-C while providing single-cell resolution and multimodal measurements characteristic of microscopy. We describe how to use this DNA labeling in parallel with multiplexed labeling of dozens of RNAs to relate chromatin structure and gene expression in the same cells. Oligopaint probe design, primary probe making, sample collection, cryosectioning and RNA/DNA primary probe hybridization can be completed in 1.5 weeks, while automated RNA/DNA barcode hybridization and RNA/DNA imaging typically takes 2–6 d for data collection and 2–7 d for the automated steps of image analysis.

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

Fig. 1: Optical reconstruction of chromatin architecture (ORCA) approach.
Fig. 2: Schematic overview of ORCA and RNA labeling experimental process and computational analysis.
Fig. 3: Comparison between ORCA and other methods.
Fig. 4: Fluidics system layout.
Fig. 5: Homebuilt microscope.
Fig. 6: Primary probe synthesis.
Fig. 7: Sample preparation for primary probe hybridization.
Fig. 8: Oligo setup for barcode labeling.
Fig. 9: Microscope and fluidics setup for an automated experiment.
Fig. 10: ChrTracer3 GUI.
Fig. 11: BuildMosaics GUI.
Fig. 12: AlignMosaics GUI.
Fig. 13: MosaicAnalyzer GUI.
Fig. 14: Data generated from a single RNA + ORCA labeling experiment.

Similar content being viewed by others

Data availability

We provide a mini dataset to help users become comfortable analyzing raw microscope data through our MATLAB GUIs: https://bit.ly/2S6eCjk. This 337 GB mini dataset contains RNA and DNA\ORCA images for a single embryo (4 FOVs) from Fig. 14. Images for an entire dataset are too large to append and, thus, image data is available upon request. All DNA paths from all cells are available upon request.

The ORCA data in Fig. 3 and Fig. 14 and the barcode detection efficiency data in Extended Data Fig. 2 are from our previous work25,46. The single-cell Hi-C and OligoSTORM data in Fig. 3 are from refs. 52,56, (Single-cell Hi-C) and9 (OligoSTORM).

Code availability

The “storm-control” software controls both the lasers and the camera. This software is a fork of the “storm-control” system written by Hazen Babcock and the Zhuang lab, with modifications specific to our application included on our fork available at: https://github.com/BoettigerLab/storm-control. The software for the fluidics handling is available at: https://github.com/BoettigerLab/fluidics-control.

Software, including the GUIs described above and some demo analysis scripts are available at https://github.com/BoettigerLab/ORCA-public. Demo data to test this software is also available (see ‘Data availability’. All software is open source and available under the CC-BY license. It may be freely edited, modified or shared. We ask only that users cite this work.

References

  1. Amano, T. et al. Chromosomal dynamics at the Shh locus: limb bud-specific differential regulation of competence and active transcription. Dev. Cell 16, 47–57 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Fabre, P. J. et al. Nanoscale spatial organization of the HoxD gene cluster in distinct transcriptional states. Proc. Natl Acad. Sci. USA 112, 13964–13969 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Williamson, I., Lettice, L. A., Hill, R. E. & Bickmore, W. A. Shh and ZRS enhancer colocalisation is specific to the zone of polarising activity. Development 143, 2994–3001 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Barbieri, M. et al. Active and poised promoter states drive folding of the extended HoxB locus in mouse embryonic stem cells. Nat. Struct. Mol. Biol. 24, 515–524 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Fraser, J., Williamson, I., Bickmore, W. A. & Dostie, J. An overview of genome organization and how we got there: from FISH to Hi-C. Microbiol. Mol. Biol. Rev. 79, 347–372 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Rust, M. J., Bates, M. & Zhuang, X. Sub-diffraction-limit imaging by stochastic optical reconstruction microscopy (STORM). Nat. Methods 3, 793–795 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Huang, B., Wang, W., Bates, M. & Zhuang, X. Three-dimensional super-resolution imaging by stochastic optical reconstruction microscopy. Science 319, 810–813 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Beliveau, B. J. et al. Single-molecule super-resolution imaging of chromosomes and in situ haplotype visualization using Oligopaint FISH probes. Nat. Commun. 6, 7147 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Boettiger, A. N. et al. Super-resolution imaging reveals distinct chromatin folding for different epigenetic states. Nature 529, 418–422 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kundu, S. et al. Polycomb repressive complex 1 generates discrete compacted domains that change during differentiation. Mol. Cell 65, 432–446.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Szabo, Q. et al. TADs are 3D structural units of higher-order chromosome organization in Drosophila. Sci. Adv. 4, eaar8082 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Kieffer-Kwon, K.-R. et al. Myc regulates chromatin decompaction and nuclear architecture during B cell activation. Mol. Cell 67, 566–578.e10 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320.e24 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Huang, P. et al. Comparative analysis of three-dimensional chromosomal architecture identifies a novel fetal hemoglobin regulatory element. Genes Devel. 31, 1704–1713 (2017).

    Article  CAS  PubMed  Google Scholar 

  15. Hsu, S. C. et al. The BET protein BRD2 cooperates with CTCF to enforce transcriptional and architectural boundaries. Mol. Cell 66, 102–116.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Brown, J. M. et al. A tissue-specific self-interacting chromatin domain forms independently of enhancer-promoter interactions. Nat. Commun. 9, 3849 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  17. van Bemmel, J. G. et al. The bipartite TAD organization of the X-inactivation center ensures opposing developmental regulation of Tsix and Xist. Nat. Genet. 51, 1024–1034 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Williamson, I. et al. Developmentally regulated Shh expression is robust to TAD perturbations. Development https://doi.org/10.1242/dev.179523 (2019).

  19. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nora, E. P. et al. Targeted degradation of CTCF decouples local insulation of chromosome domains from genomic compartmentalization. Cell 169, 930–944.e22 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schwarzer, W. et al. Two independent modes of chromatin organization revealed by cohesin removal. Nature 551, 51–56 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Symmons, O. et al. The Shh topological domain facilitates the action of remote enhancers by reducing the effects of genomic distances. Dev. Cell 39, 529–543 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Finn, E. H. & Misteli, T. Molecular basis and biological function of variability in spatial genome organization. Science 365, eaaw9498 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Mateo, L. J. et al. Visualizing DNA folding and RNA in embryos at single-cell resolution. Nature 568, 49–54 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lubeck, E. & Cai, L. Single-cell systems biology by super-resolution imaging and combinatorial labeling. Nat. Methods 9, 743–748 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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  PubMed  PubMed Central  Google Scholar 

  28. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. Spatially resolved, highly multiplexed RNA profiling in single cells. Science 348, 412 (2015).

    Article  CAS  Google Scholar 

  29. Fazal, F. M. et al. Atlas of subcellular RNA localization revealed by APEX-Seq. Cell 178, 473–490.e26 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  31. Levesque, M. J. & Raj, A. Single-chromosome transcriptional profiling reveals chromosomal gene expression regulation. Nat. Methods 10, 246–248 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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  PubMed  PubMed Central  Google Scholar 

  33. Beliveau, B. J. et al. Versatile design and synthesis platform for visualizing genomes with Oligopaint FISH probes. Proc. Natl Acad. Sci. USA 109, 21301–21306 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Moffitt, J. R. et al. High-throughput single-cell gene-expression profiling with multiplexed error-robust fluorescence in situ hybridization. Proc. Natl Acad. Sci. USA 113, 11046–11051 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Beliveau, B. J. et al. In situ super-resolution imaging of genomic DNA with OligoSTORM and OligoDNA-PAINT. Methods Mol. Biol. 1663, 231–252 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Beliveau, B. J. et al. OligoMiner provides a rapid, flexible environment for the design of genome-scale oligonucleotide in situ hybridization probes. Proc. Natl Acad. Sci. USA 115, E2183–E2192 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Gelali, E. et al. iFISH is a publically available resource enabling versatile DNA FISH to study genome architecture. Nat. Commun. 10, 1636 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Boettiger, A. & Murphy, S. Advances in chromatin imaging at kilobase-scale resolution. Trends Genet. 36, 273–287 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. 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  PubMed  PubMed Central  Google Scholar 

  40. Zenklusen, D., Larson, D. R. & Singer, R. H. Single-RNA counting reveals alternative modes of gene expression in yeast. Nat. Struct. Mol. Biol. 15, 1263–1271 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Paré, A. et al. Visualization of individual Scr mRNAs during Drosophila embryogenesis yields evidence for transcriptional bursting. Curr. Biol. 19, 2037–2042 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Boettiger, A. N. & Levine, M. Rapid transcription fosters coordinate snail expression in the Drosophila embryo. Cell Rep 3, 8–15 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Trcek, T. et al. Single-mRNA counting using fluorescent in situ hybridization in budding yeast. Nat. Protoc. 7, 408–419 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rouhanifard, S. H. et al. ClampFISH detects individual nucleic acid molecules using click chemistry-based amplification. Nat. Biotechnol. https://doi.org/10.1038/nbt.4286 (2018).

  45. Lyubimova, A. et al. Single-molecule mRNA detection and counting in mammalian tissue. Nat. Protoc. 8, 1743–1758 (2013).

    Article  PubMed  Google Scholar 

  46. Bintu, B. et al. Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 362, eaau1783 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Weber, C. M., Hafner, A., Braun, S. M. G., Boettiger, A. N. & Crabtree, G. R. mSWI/SNF promotes distal repression by titrating polycomb dosage. Preprint at bioRxiv https://doi.org/10.1101/2020.01.29.925586 (2020).

  48. Quinodoz, S. A. et al. Higher-order inter-chromosomal hubs shape 3D genome organization in the nucleus. Cell 174, P744–757.E24 (2018).

    Article  Google Scholar 

  49. Beagrie, R. A. et al. Complex multi-enhancer contacts captured by genome architecture mapping. Nature https://doi.org/10.1038/nature21411 (2017).

  50. McCord, R. P., Kaplan, N. & Giorgetti, L. Chromosome Conformation Capture and Beyond: Toward an Integrative View of Chromosome Structure and Function. Mol. Cell 77, 688–708 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Nagano, T. et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502, 59–64 (2013).

    Article  CAS  PubMed  Google Scholar 

  52. Stevens, T. J. et al. 3D structures of individual mammalian genomes studied by single-cell Hi-C. Nature 544, 59–64 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Flyamer, I. M. et al. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 544, 110–114 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gassler, J. et al. A mechanism of cohesin-dependent loop extrusion organizes zygotic genome architecture. EMBO J. 36, 3600–3618 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Tan, L., Xing, D., Chang, C.-H., Li, H. & Xie, X. S. Three-dimensional genome structures of single diploid human cells. Science 361, 924–928 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ramani, V. et al. Massively multiplex single-cell Hi-C. Nat. Methods 14, 263–266 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Nir, G. et al. Walking along chromosomes with super-resolution imaging, contact maps, and integrative modeling. PLoS Genet. 14, e1007872 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Beliveau, B. J. et al. In situ super-resolution imaging of genomic DNA with OligoSTORM and OligoDNA-PAINT. Methods Mol. Biol. 1663, 231–252 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Jungmann, R. et al. Multiplexed 3D cellular super-resolution imaging with DNA-PAINT and Exchange-PAINT. Nat. Methods https://doi.org/10.1038/nmeth.2835 (2014).

  60. Georgieva, M. et al. Nanometer resolved single-molecule colocalization of nuclear factors by two-color super resolution microscopy imaging. Methods 105, 44–55 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Wang, S. et al. Spatial organization of chromatin domains and compartments in single chromosomes. Science 353, 598–602 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Finn, E. H. et al. Extensive heterogeneity and intrinsic variation in spatial genome organization. Cell 176, 1502–1515.e10 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Cardozo Gizzi, A. M. et al. Microscopy-based chromosome conformation capture enables simultaneous visualization of genome organization and transcription in intact organisms. Mol. Cell 74, 212–222.e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  64. Cardozo Gizzi, A. M. et al. Direct and simultaneous observation of transcription and chromosome architecture in single cells with Hi-M. Nat. Protoc. 15, 840–876 (2020).

    Article  CAS  PubMed  Google Scholar 

  65. Moffitt, J. R. et al. High-performance multiplexed fluorescence in situ hybridization in culture and tissue with matrix imprinting and clearing. Proc. Natl Acad. Sci. USA 113, 14456–14461 (2016).

    Article  CAS  PubMed  Google Scholar 

  66. Eng, C.-H. L. et al. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH. Nature https://doi.org/10.1038/s41586-019-1049-y (2019).

  67. Magaraki, A., Loda, A., Gribnau, J. & Baarends, W. M. Simultaneous RNA-DNA FISH in mouse preimplantation embryos. Methods Mol. Biol. 1861, 131–147 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. Lai, L.-T., Meng, Z., Shao, F. & Zhang, L.-F. in Long Non-Coding RNAs: Methods and Protocols (eds. Feng, Y. & Zhang, L.) 135–145 (Springer, 2016).

  69. Lanzuolo, C., Roure, V., Dekker, J., Bantignies, F. & Orlando, V. Polycomb response elements mediate the formation of chromosome higher-order structures in the bithorax complex. Nat. Cell Biol. 9, 1167–1174 (2007).

    Article  CAS  PubMed  Google Scholar 

  70. Lee, J. T., Strauss, W. M., Dausman, J. A. & Jaenisch, R. A 450 kb transgene displays properties of the mammalian X-inactivation center. Cell 86, 83–94 (1996).

    Article  CAS  PubMed  Google Scholar 

  71. Deng, W., Shi, X., Tjian, R., Lionnet, T. & Singer, R. H. CASFISH: CRISPR/Cas9-mediated in situ labeling of genomic loci in fixed cells. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.1515692112 (2015).

  72. Solovei, I. et al. Spatial preservation of nuclear chromatin architecture during three-dimensional fluorescence in situ hybridization (3D-FISH). Exp. Cell Res. 276, 10–23 (2002).

    Article  CAS  PubMed  Google Scholar 

  73. Su, J.-H., Zheng, P., Kinrot, S. S., Bintu, B. & Zhuang, X. Genome-scale imaging of the 3D organization and transcriptional activity of chromatin. Cell https://doi.org/10.1016/j.cell.2020.07.032 (2020).

  74. Rasnik, I., McKinney, S. A. & Ha, T. Nonblinking and long-lasting single-molecule fluorescence imaging. Nat. Methods 3, 891–893 (2006).

    Article  CAS  PubMed  Google Scholar 

  75. Shi, X., Lim, J. & Ha, T. Acidification of the oxygen scavenging system in single-molecule fluorescence studies: in situ sensing with a ratiometric dual-emission probe. Anal. Chem. 82, 6132–6138 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Tokunaga, M., Imamoto, N. & Sakata-Sogawa, K. Highly inclined thin illumination enables clear single-molecule imaging in cells. Nat. Methods 5, 159–161 (2008).

    Article  CAS  PubMed  Google Scholar 

  77. Lim, D., Ford, T. N., Chu, K. K. & Mertz, J. Optically sectioned in vivo imaging with speckle illumination HiLo microscopy. J. Biomed. Opt. 16, 016014 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Babcock, H. et al. ZhuangLab/storm-control: v2019.06.28 release. https://doi.org/10.5281/zenodo.3264857 (2019).

  79. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Murphy, T. Hafner, M. Park, and A. Rajpurkar for critical reading of the manuscript. This work was supported by the Searle Scholars Program, Burroughs Wellcome Careers at the Scientific Interface Award, Beckman Young Investigator Program, NIH New Innovator Award DP2 (DGM132935A), and the Packard Fellows Program (A.N.B.). L.J.M. was supported by the Howard Hughes Medical Institute (HHMI) Gilliam Fellowship. N.S.-A. was supported by a Stanford Graduate Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

L.J.M. and A.N.B. designed the experiments. L.J.M. collected the microscopy data. N.A.S.-A. assembled and programmed the fluidics robot, which was modified from a commercial CNC mill. L.J.M. and A.N.B. analysed the data and wrote the manuscript.

Corresponding author

Correspondence to Alistair N. Boettiger.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks Jan Ellenberg, Kazuhiro Maeshima, Lothar Schermelleh 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.

Related links

Key references using this protocol

Mateo, L. et al. Nature 568, 49–54 (2019): https://doi.org/10.1038/s41586-019-1035-4

Bintu, B. et al. Science 362, eaau1783 (2018): https://doi.org/10.1126/science.aau1783

Extended data

Extended Data Fig. 1 Distinct intronic RNA patterns in cryosectioned tissue.

A zoomed-in image of the most posterior body segments of a 10-12 hpf Drosophila embryo. With multiplex RNA labeling, we targeted the following RNA introns: Antp (green), abd-A (yellow), Abd-B (red), inv (blue). Note that these introns have distinct RNA patterns.

Extended Data Fig. 2 Barcode detection efficiency.

Figure modified from previous publication25. a, The average detection efficiency of each barcode is shown for probe sets with the resolution and cell type indicated. Error bars represent standard error of the mean. n = 1,979 cells, n = 8,801 cells, n = 94,300 cells, and n = 15,230 cells for the 30-kb, 10-kb, 3-kb, and 2-kb probesets, respectively. b, Box-and-whisker plots comparing the distribution of detection efficiencies across barcodes between two different embryos. N.S. indicates not statistically significant (two-sided Wilcoxon test, p-values indicated). For the boxplots in (b-e), the red line marks the median, the upper and lower limits of the box mark the interquartile range, and the whiskers extend to the furthest datum within 1.5 times the interquartile range. Notches denote the 95% confidence interval around the median. Sample size (n) is indicated. c, Comparison of the distribution of detection efficiencies between cells of different embryos. d, Comparison of the detection efficiency across barcodes between anterior and posterior cells. e, Comparisons of the detection efficiency across all cells between anterior and posterior cells. Results are shown for the 10-kb, 3-kb and 2-kb probesets as indicated. f, Correlation between normalized distance data from different cell types and a modified dataset in which 50% of detected barcodes were removed at random to simulate a high missed detection rate. The correlation coefficients are indicated (Pearson), computed using all unique combinations of 52 barcodes. g, Bar graph comparing the mean correlation between matched segments in the original and downsampled data shown in (f) compared to the mean inter-segment correlation. The correlation coefficients are indicated (Pearson), computed using all unique combinations of 52 barcodes. All 15 matched segment pairs shown in f are plotted compared to all 210 inter-segment pairs. This illustrates that the difference between the substantially downsampled data and the original measurements are much more similar than the typical differences between segments, demonstrating the robustness of the segment-specific findings to missing data. Error bars denote standard error of the mean. h, Histogram showing the detection efficiency per cell for each probeset.

Extended Data Fig. 3 Setting up the flow chamber.

a, Layout of all the components that are needed to assemble the flow chamber. Before using, clean all components with a kimwipe that is slightly damp with ultra-pure water. Note that the 400 mm coverslip is marked with an alcohol-resistant pen. The marks indicate the location of the embryos, which are cryosectioned between the two lines in the center of the coverslip. This space is determined by the middle area of gasket 2. We also used the pen to label that our sample is WT. We find that labeling the slide does not harm the sample. b, Invert the plastic chamber. Then take gasket 1 and place its two holes over the two metal aqueduct ports of the plastic chamber. c, Carefully, place gasket 2 on top of the coverslip. You can use the pen labeled markers as an indicator of where to place the gasket. Make sure to not place gasket 2 directly on top of your sample. Any sample under the gasket will not be accessible for imaging. Also, make sure to not accidentally slide the gasket on top of your sample as this may result in your sample rubbing off the coverslip. d, Place the miroaqueuct glass slide on top of gasket 2. Carefully, place the microaqueduct slide so that its wells on the side (pointed by arrows) are perpendicular to gasket 2. e, Invert the sandwiched components - including the 40-mm coverslip, gasket 2, and the microaqueduct slide - and place the two holes of the microaqueduct slide on top of the plastic chamber’s aqueduct ports. f, Place the plastic chamber on a flat surface. Invert the metal mount and place it on top of the plastic chamber. When placing it on top, it is useful to make sure that the clamps are aligned to the plastic chamber holes as shown here. g, Invert all the components. During this flip, make sure to carefully hold onto both the metal chamber and plastic chamber. We find it useful to hold the metal part with our index/middle finger(s) and to have our thumbs holding onto the tubes of the plastic chamber. h, Tighten the metal clamps by moving the chamber counterclockwise to lock it. Make sure to tighten this as hard as you can, and without breaking the glass components inside. Afterward, connect the “In” and “Out” fluidics tube to the flow chamber’s metal tubes (shown with arrows).

Supplementary information

Supplementary Information

Supplementary Data 1–8 legends.

Reporting Summary

Supplementary Data 1

Primary probe synthesis costs. An EXCEL file that contains the information of all the components - including vendor, product number, unit, etc - needed for primary probe synthesis. Importantly, we have included the total cost to make a single primary probe.

Supplementary Data 2

Cost of readout probe hybridization. An EXCEL file listing the reagents needed for each hybridization round. The user can edit this file to calculate the total cost per each experiment. The cost depends upon the number of readout probes, fluorescent oligos, and buffer used. The user can choose which fluorescent oligos to use; however, we advise that these oligos should be imaged in a different channel from the fiducial. See Supplementary Data Legends for full description.

Supplementary Data 3

Length of imaging experiment and ChrTracer3 analysis. An editable EXCEL file that is useful to estimate the amount of time it will take to complete an imaging experiment and analyze the raw data in ChrTracer3. We have listed all the parameters that would influence the length of time for both of these. Please note that in ‘Imaging Time’ we have listed the ‘Number of readouts per hybridization’ to be ‘2’, which means we are showing an example of a three-color experiment Details of a three-color experiment can be found in the legend of Supplementary Data 2.

Supplementary Data 4

Raw data storage. An editable EXCEL file to help the user calculate the amount of storage space needed for an imaging experiment. This will also calculate the cost to store the data.

Supplementary Data 5

Components for our homebuilt microscope. An EXCEL file containing detailed information - including the supplier(s), part number(s), quantity, units, and total costs - of each component used for our microscope. The total cost of the homebuilt microscope is also included in this table.

Supplementary Data 6

RNA Table. An example of a RNAtable.xlsx file that is used for downstream analysis, such as in BuildMosaicsGUI. See Supplementary Data Legends for full description.

Supplementary Data 7

DNA Table. An example of ExperimentalLayout.xlsx that is used for ChrTracer3 and downstream GUIs. The columns are the same as that of RNAtable.xlsx (see legend for Supplementary Data 6), with two exceptions. 1) For DNA labeling, we typically relabel some of the barcodes at the experiment for quantification purposes. Thus, you will see that at the bottom of the ‘DataType’ column, the last 5 rows are labeled as ‘R’, indicating that these are the barcodes that were relabeled. 2) There is no ‘rnaNames’ column as this is a DNA table.

Supplementary Data 8

ChrTracer3 output file. An example of a fov001_AllFits.csv table that is generated at the end of ChrTracer3 analysis. See Supplementary Data Legends for full description.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mateo, L.J., Sinnott-Armstrong, N. & Boettiger, A.N. Tracing DNA paths and RNA profiles in cultured cells and tissues with ORCA. Nat Protoc 16, 1647–1713 (2021). https://doi.org/10.1038/s41596-020-00478-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-020-00478-x

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