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:

Deterministic scRNA-seq captures variation in intestinal crypt and organoid composition

Abstract

Single-cell RNA sequencing (scRNA-seq) approaches have transformed our ability to resolve cellular properties across systems, but are currently tailored toward large cell inputs (>1,000 cells). This renders them inefficient and costly when processing small, individual tissue samples, a problem that tends to be resolved by loading bulk samples, yielding confounded mosaic cell population read-outs. Here, we developed a deterministic, mRNA-capture bead and cell co-encapsulation dropleting system, DisCo, aimed at processing low-input samples (<500 cells). We demonstrate that DisCo enables precise particle and cell positioning and droplet sorting control through combined machine-vision and multilayer microfluidics, enabling continuous processing of low-input single-cell suspensions at high capture efficiency (>70%) and at speeds up to 350 cells per hour. To underscore DisCo’s unique capabilities, we analyzed 31 individual intestinal organoids at varying developmental stages. This revealed extensive organoid heterogeneity, identifying distinct subtypes including a regenerative fetal-like Ly6a+ stem cell population that persists as symmetrical cysts, or spheroids, even under differentiation conditions, and an uncharacterized ‘gobloid’ subtype consisting predominantly of precursor and mature (Muc2+) goblet cells. To complement this dataset and to demonstrate DisCo’s capacity to process low-input, in vivo-derived tissues, we also analyzed individual mouse intestinal crypts. This revealed the existence of crypts with a compositional similarity to spheroids, which consisted predominantly of regenerative stem cells, suggesting the existence of regenerating crypts in the homeostatic intestine. These findings demonstrate the unique power of DisCo in providing high-resolution snapshots of cellular heterogeneity in small, individual tissues.

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: Overview and critical feature assessment of the DisCo system.
Fig. 2: Use of DisCo to map intestinal organoid cell identities.
Fig. 3: Use of DisCo to map intestinal organoid cell identity along development.
Fig. 4: Cell type distribution and marker gene expression across individual intestinal organoids.
Fig. 5: Cell type distribution across individual intestinal crypts.

Similar content being viewed by others

Data availability

The GEO (Gene Expression Omnibus) accession number for scRNA-seq data reported in this paper is GSE148093. The raw data and count matrices for Fig. 1h and Extended Data Fig. 2c are stored under the access code GSM4454017. The raw data and count matrices for Fig. 1i and Extended Data Fig. 2a are available under the access code GSM4454017. The raw data and count matrices for Fig. 1j are stored under the access codes GSM4454012GSM4454016. The raw data and count matrices for Extended Data Fig. 2e,f are stored under the access codes GSM5567775GSM5567779. The raw data and count matrices for Extended Data Fig. 2g are stored under the access codes GSM5567571GSM5567730. The raw data and count matrices for Extended Data Fig. 4 are stored under the access codes GSM5567845GSM5567854. The raw data for intestinal organoids embedded in Figs. 2 and 3, Extended Data Figs. 35, Figs. 4a,e and 5d,e and Extended Data Figs. 7, 9 and 10 are stored under access codes GSM4453981GSM4454011. The raw data and count matrices for intestinal crypts embedded in Fig. 5 and Extended Data Figs. 810 are stored under the access codes GSM5567818GSM5567844. Additionally, dataset GSM1544799 and data from ref. 23 (https://doi.org/10.1039/C9LC00014C, data available on request) were used for Fig. 1i and Extended Data Fig. 2a. In this study the following reference genomes were used: hg38 (GCF_000001405.26), mm10 (GCF_000001635.20) and mixed reference genome (GSE63269) of hg19 combined with mm10.

Code availability

This technology has been developed as an open source platform, therefore all required information for its implementation is publicly available. The source code for the machine-vision software is available on github (https://github.com/DeplanckeLab/DisCo_source) and the barcode merging script is supplied as Supplementary Software 1.

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Klein, A. M. et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gierahn, T. M. et al. RNA sequencing of single cells at high throughput. Nat. Methods 14, 395–398 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Han, X. et al. Mapping the mouse cell atlas by Microwell-Seq. Cell 172, 1091–1107 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Rosenberg, A. B. et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 360, 176–182 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Li, H. et al. Fly cell atlas: a single-cell transcriptomic atlas of the adult fruit fly. Preprint at bioRxiv https://doi.org/10.1101/2021.07.04.451050 (2021).

  8. Tabula Muris Consortium Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Article  Google Scholar 

  9. Han, X. et al. Construction of a human cell landscape at single-cell level. Nature 581, 303–309 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Stoeckius, M. et al. Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. McGinnis, C. S. et al. MULTI-seq: sample multiplexing for single-cell RNA sequencing using lipid-tagged indices. Nat. Methods 16, 619–626 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gehring, J., Hwee Park, J., Chen, S., Thomson, M. & Pachter, L. Highly multiplexed single-cell RNA-seq by DNA oligonucleotide tagging of cellular proteins. Nat. Biotechnol. 38, 35–38 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Hwang, B., Lee, J. H. & Bang, D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp. Mol. Med. 50, 1–14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 10X Genomics. Chromium Single Cell 3ʹ Reagent Kits User Guide (v3 Chemistry) (CG000183 Rev C) (2018).

  15. DeLaughter, D. M. The use of the Fluidigm C1 for RNA expression analyses of single cells. Curr. Protoc. Mol. Biol. 122, e55 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Wagner, D. E. et al. Single-cell mapping of gene expression landscapes and lineage in the zebrafish embryo. Science 360, 981–987 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Packer, J. S. et al. A lineage-resolved molecular atlas of C. elegans embryogenesis at single-cell resolution. Science 365, eaax1971 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Serra, D. et al. Self-organization and symmetry breaking in intestinal organoid development. Nature 562, 66–72 (2019).

    Article  Google Scholar 

  19. Grün, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–255 (2015).

    Article  PubMed  Google Scholar 

  20. Lukonin, I. et al. Phenotypic landscape of intestinal organoid regeneration. Nature 586, 275–280 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tirier, S. M. et al. Pheno-seq: linking visual features and gene expression in 3D cell culture systems. Sci. Rep. 9, 12367 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Unger, M. A., Chou, H. P., Thorsen, T., Scherer, A. & Quake, S. R. Monolithic microfabricated valves and pumps by multilayer soft lithography. Science 288, 113–116 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Biocanin, M., Bues, J., Dainese, R., Amstad, E. & Deplancke, B. Simplified Drop-seq workflow with minimized bead loss using a bead capture and processing microfluidic chip. Lab Chip 19, 1610–1620 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Sato, T. et al. Single Lgr5 stem cells build crypt–villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Haber, A. L. et al. A single-cell survey of the small intestinal epithelium. Nature 551, 333–339 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yin, X. et al. Niche-independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny. Nat. Methods 11, 106–112 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Gregorieff, A., Liu, Y., Inanlou, M. R., Khomchuk, Y. & Wrana, J. L. Yap-dependent reprogramming of Lgr5+ stem cells drives intestinal regeneration and cancer. Nature 526, 715–718 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Ayyaz, A. et al. Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell. Nature 569, 121–125 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Roulis, M. et al. Paracrine orchestration of intestinal tumorigenesis by a mesenchymal niche. Nature 580, 524–529 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Battich, N. et al. Sequencing metabolically labeled transcripts in single cells reveals mRNA turnover strategies. Science 367, 1151–1156 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Birchenough, G., Johansson, M., Gustafsson, J., Bergstrom, J. & Hansson, G. C. New developments in goblet cell mucus secretion and function. Mucosal Immunol. 8, 712–719 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yui, S. et al. YAP/TAZ-dependent reprogramming of colonic epithelium links ECM remodeling to tissue regeneration. Cell Stem Cell 22, 35–49 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Macnair, W. & Claassen, M. psupertime: supervised pseudotime inference for single cell RNA-seq data with sequential labels. Preprint at bioRxiv https://doi.org/10.1101/622001 (2019).

  35. Lareau, C. A., Ma, S., Duarte, F. M. & Buenrostro, J. D. Inference and effects of barcode multiplets in droplet-based single-cell assays. Nat. Commun. 11, 866 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Chung, M., Núñez, D., Cai, D. & Kurabayashi, K. Deterministic droplet-based co-encapsulation and pairing of microparticles via active sorting and downstream merging. Lab Chip 17, 3664–3671 (2017).

    Article  CAS  PubMed  Google Scholar 

  38. Cheng, Y. H. et al. Hydro-Seq enables contamination-free high-throughput single-cell RNA-sequencing for circulating tumor cells. Nat. Commun. 10, 2163 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Zhang, M. et al. Highly parallel and efficient single cell mRNA sequencing with paired picoliter chambers. Nat. Commun. 11, 2118 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pollen, A. A. et al. Low-coverage single-cell mRNA sequencing reveals cellular heterogeneity and activated signaling pathways in developing cerebral cortex. Nat. Biotechnol. 32, 1053–1058 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Dura, B. et al. Profiling lymphocyte interactions at the single-cell level by microfluidic cell pairing. Nat. Commun. 6, 5940 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Gérard, A. et al. High-throughput single-cell activity-based screening and sequencing of antibodies using droplet microfluidics. Nat. Biotechnol. 38, 715–721 (2020).

    Article  PubMed  Google Scholar 

  43. Maier, G. L. et al. Multimodal and multisensory coding in the Drosophila larval peripheral gustatory center. Preprint at bioRxiv https://doi.org/10.1101/2020.05.21.109959 (2020).

  44. Haque, A., Engel, J., Teichmann, S. A. & Lönnberg, T. A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med. 9, 75 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Denisenko, E. et al. Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows. Genome Biol. 21, 130 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mustata, R. C. et al. Identification of Lgr5-independent spheroid-generating progenitors of the mouse fetal intestinal epithelium. Cell Rep. 5, 421–432 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  48. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2016).

    Article  Google Scholar 

  49. Zhang, X. et al. Comparative analysis of droplet-based ultra-high-throughput single-cell RNA-seq systems. Mol. Cell 73, 130–142 (2019).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  51. Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 17, 77 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Goldstein, L. D. et al. Massively parallel nanowell-based single-cell gene expression profiling. BMC Genomics 18, 519 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Wang, Y. et al. Comparative analysis of commercially available single-cell RNA sequencing platforms for their performance in complex human tissues. Preprint at bioRxiv https://doi.org/10.1101/541433 (2019).

  54. Ding, J. et al. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat. Biotechnol. 38, 737–746 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Gjorevski, N. et al. Designer matrices for intestinal stem cell and organoid culture. Nature 539, 560–564 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Bas, T. & Augenlicht, L. H. Real time analysis of metabolic profile in ex vivo mouse intestinal crypt organoid cultures. J. Vis. Exp. 93, e52026 (2014).

  57. Macosko, E., Goldman, M. & McCarroll, S. Drop-Seq Laboratory Protocol version 3.1. http://mccarrolllab.org/download/905/ (2015).

  58. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. 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  PubMed  Google Scholar 

  61. Saelens, W., Cannoodt, R., Todorov, H. & Saeys, Y. A comparison of single-cell trajectory inference methods. Nat. Biotechnol. 37, 547–554 (2019).

    Article  CAS  PubMed  Google Scholar 

  62. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank the former members of the Deplancke laboratory (EPFL) for their support: W. Chen and P.C. Schwalie for constructive discussions, and V. Braman for help in establishing the intestinal organoid culture. Furthermore, the authors thank G. Sorrentino from K. Schoonjans’ laboratory (EPFL) for valuable advice and support during organoid culture establishment; L. Aeberli and G. Muller from SEED Biosciences for cell sorting support; and the EPFL CMi, GECF, BIOP, FCCF, Histology Core Facility, SCITAS, and UNIL VITAL-IT for device fabrication, sequencing, imaging, sorting, histology, and computational support, respectively. The authors also thank J. Sordet-Dessimoz from the Histology Core Facility for her support with the RNAscope assay. This research was supported by the Swiss National Science Foundation Grant (IZLIZ3_156815) and a Precision Health and Related Technologies (PHRT-502) grant (to B.D.), the Swiss National Science Foundation SPARK initiative (CRSK-3_190627) and the EuroTech PostDoc Programme co-funded by the European Commission under its framework program Horizon 2020 (754462, to J.P.), as well as by the EPFL SV Interdisciplinary PhD Funding Program (to B.D. and E.A.). Y.S. is an ISAC Marylou Ingram scholar.

Author information

Authors and Affiliations

Authors

Contributions

B.D., J.B. and M.B. designed the study. B.D., J.B., M.B. and J.P. wrote the manuscript. J.B. and R.D. designed and fabricated the microfluidic chips. J.B. developed the machine-vision integration for DisCo. J.B. and M.B. benchmarked the system and performed all single-cell RNA-seq experiments. J.P., J.B., M.B., W.S., V.G. and R.G. performed data analysis related to single organoid and crypt scRNA-seq experiments. J.B., S.R. and M.B. performed all organoid and cell culture assays. J.B., A.C., J.R. and R.S. performed all imaging assays. M.B. and J.R. isolated intestinal crypts, and M.B. picked and dissociated crypts. Y.S. supervised data analysis aspects and reviewed the manuscript. E.A. provided critical comments regarding microfluidic chip design and fabrication. M.C. provided critical comments on intestinal organoid scRNA-seq data analysis. M.P.L. provided critical comments regarding intestinal organoid scRNA-seq data and the design of critical confirmation experiments. All authors read, discussed and approved the final manuscript.

Corresponding author

Correspondence to Bart Deplancke.

Ethics declarations

Competing interests

B.D., J.B., M.B. and R.D. have filed a patent application for the deterministic co-encapsulation system (patent no. US20190240664A1). All other authors have no competing interests.

Peer review

Peer review information

Nature Methods thanks Jarrett Camp and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Lei Tang was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 DisCo device features and their performance.

(a) Schematic of the DisCo device design (blue: flow layer, green: control layer). 1. oil valve, 2. oil inlet, 3. cell inlet, 4. bead inlet, 5. cell valve, 6. dropleting valve, 7. bead valve, 8. sample valve, 9. waste valve, 10. sample outlet, 11. waste outlet. (b) Visualization of real-time image processing for particle detection. (c) Particle positioning by valve oscillation. Approaching particles are detected in the detection zone. Once a particle is detected, the channel valve is oscillated to induce discrete movements of particles. Oscillation is terminated once correct placement of a particle is achieved. (d) Stopping accuracy in a defined window. Beads (n = 744) were positioned using valve oscillation, their position was manually determined within the stopping area. Scale was approximated from channel width. (e) Volume-defined droplet on-demand generation by valve pressurization. Droplets (n = 68, ~8 per condition, 1 experiment) were produced by pressurizing the dropleting valve at different pressures. Size was determined by imaging the dropleting process. Volumes were calculated from the imaging data based on droplet length and channel geometry. Thus, they should be considered an approximation. Points represent mean values, error bars +/-SD. The channel width of displayed images is 250 μm.

Extended Data Fig. 2 Quality assessment of DisCo scRNA-seq data.

(a) Cumulative reads per barcode (n = 500) for DisCo and two Drop-seq experiments2,23. (b) Hamming distances between all 12 nt barcodes of a Drop-seq experiment and generated 12 nt random barcode sequences representing the probability density for each set of barcodes. (c) Species purity (bars) and doublet ratio (dots) for unmerged (n = 949) and merged barcodes (n = 274). Data represent mean values, error bars standard deviation. (d-e) HEK 293T cells were processed with DisCo at 22 °C after 20, 40 or 60 min or stored on ice for 120 or 180 min and subsequently processed. (d) UMAP embedding of all profiled HEK 293T cells from the five sampling time points, color-coded by sampling time. (e) Violin plots showing the percentage of UMIs per cell of heat-shock-protein (HSP), mitochondrial protein-coding (MT), or ribosomal protein-coding (RPL) genes. (f) Correlation of the number of manually counted cells by fluorescence microscopy and the number of cells quantified by the DISPENCELL platform. (g) A quantified number of HEK 293T cells was processed with the Fluidigm C1 system. Processing efficiency was calculated as the percentage of cells retrieved from the sequencing data respective to the quantified number of input cells. The red line represents 100% efficiency, and samples were colored according to the recovery efficiency after sequencing.

Extended Data Fig. 3 DisCo performance on individual intestinal organoids and data analysis.

(a) Representative bright-field image of a differentiated organoid culture from single LGR5+ cells, as performed for experiments shown in Fig. 2. (b) Correlation of encapsulated cells on-chip with the number of cells detected after sequencing (cells passing QC, filtered above 800 genes/cell). (c) UMAP embedding colored by the number of detected genes (nFeature) per cell, the number of detected UMIs (nCount) per cell, the percentage of mitochondrial (mt) reads per cell, and the percentage of reads mapping to genes coding for respectively heat-shock proteins (Hsp), and ribosomal proteins (Rpl) per cell. (d) UMAP embedding colored by expression of selected marker genes (Clu, Anxa1, Spink4, ChgB, ChgA, Agr2, Clca1, and Fcgbp). (e) UMAP embedding for each of the three independent experimental batches colored by cluster annotation.

Extended Data Fig. 4 Batch effect assessment on organoids by split organoid experiment.

(a) UMAP embedding of cells collected from nine additional individual organoids (under maintenance conditions) for the purpose of evaluating batch effects. Left: All 748 processed cells clustered with k-means clustering, after which clusters were annotated according to marker gene expression. Right: Expression dot plot of selected marker genes. (b) Projection of cells (colored by cell type) derived from one organoid that was split into two independent samples (split organoid) on the reference UMAP shown in a). Organoid ‘S2_2’ was split into two batches, which were processed subsequently, with a one-hour delay, during which the second batch was stored at 4 °C.

Extended Data Fig. 5 Marker genes and YAP1 target gene activity of intestinal organoid cells.

(a) Heatmap of top DE genes per annotated cluster. (b) YAP1 target gene activity on a UMAP embedding. The expression of genes that are positively regulated by YAP127 was calculated as the cumulative Z-score and projected on the UMAP embedding of all sequenced cells.

Extended Data Fig. 6 Control images of DisCo- and RNAscope-processed organoids.

(a) Selected organoids from Fig. 4, imaged in microwell plates before dissociation to single cells. Scale bar: 50 μm. (b) RNAscope controls for organoids shown in Fig. 4b,f. Positive control (PpiB), and negative control (Duplex negative). Scale bar: 50 μm.

Extended Data Fig. 7 Marker gene expression per individual organoid.

(a) Violin plots showing marker gene expression (Fabp1, Muc2, Sox9, Olfm4, Reg3b, Ly6a) per organoid. (b) Violin plots showing the expression of selected genes (Defa24, Gip, Vnn1, Zg16) identified via psupertime analysis per individual organoid.

Extended Data Fig. 8 DisCo performance on individual intestinal crypts and data analysis.

(a) Processing efficiency of DisCo for individual and bulk intestinal crypts. All cells processed with DisCo were manually counted during the experiment, and compared to cell numbers after quality filtering (>500 UMIs). The red line represents 100% efficiency, and samples are colored according to sample type. (b) Expression dot plot of marker genes for clusters shown in Fig. 5a. (c) Gene activity represented as the cumulative Z-score and projected on the UMAP embedding of all sequenced cells using the expression of Top: Paneth cell-associated genes encompassing Lyz1, Defa17, Defa24 and Ang4 and Bottom: genes that are positively regulated by YAP127. (d) Projection of cell types onto the reference UMAP of cells derived from the 21 individual crypts. Cells per single crypt were colored according to their global clustering and highlighted on the UMAP embedding of all sequenced cells. Enterocytes (Entero), PIC (Potential intermediate cells), RegStem, (Regenerative Stem), TA (Transit amplifying cells; G1: G1/S and G2: G2/M cell cycle phase).

Extended Data Fig. 9 Analysis of combined intestinal organoid and crypt data.

(a) Combined UMAP embedding (as shown in Fig. 5d) stratified by the five individual batches of intestinal crypt samples and the three independent experimental batches of intestinal organoid differentiation samples, collectively embedded and colored by cluster annotation. (b) UMAP-based visualization of the expression of specific markers that were used for cluster annotation. (c) Bar graph depicting the cumulative Z-score of the expression of genes that are indicated within the respective bar graph. CanStem: canonical stem cell, RegStem: regenerative stem cell.

Extended Data Fig. 10 Individual organoids and crypts mapped onto the denominator UMAP.

Projection of cell types onto the reference UMAP of the ex vivo cell preparation for the 21 individual intestinal crypts and bulk samples embedded together with the 31 individual intestinal organoids. Cells per single crypt or organoid are colored according to their global clustering and highlighted on the UMAP embedding of all sequenced cells.

Supplementary information

Supplementary Information

Supplementary Fig. 1

Reporting Summary

Supplementary Video 1

Video showing on-demand droplet production with varying dropleting valve pressures.

Supplementary Video 2

Annotated video showing the operational DisCo system.

Supplementary Data 1

CAD file of the microfluidic chip control layer and CAD file of the microfluidic chip flow layer.

Supplementary Software 1

R-script for cell barcode merging and R-script used for scRNA-seq data analysis.

Supplementary Tables 1–3

Supplementary Table 1: Cellular yield per intestinal organoid and intestinal crypt. Supplementary Table 2: DE genes for cell clusters (as shown in Fig. 2B). Supplementary Table 3: Buffer-dependent-dissociation efficiencies for intestinal crypts.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bues, J., Biočanin, M., Pezoldt, J. et al. Deterministic scRNA-seq captures variation in intestinal crypt and organoid composition. Nat Methods 19, 323–330 (2022). https://doi.org/10.1038/s41592-021-01391-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-021-01391-1

This article is cited by

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