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:

Decoder-seq enhances mRNA capture efficiency in spatial RNA sequencing

Abstract

Spatial transcriptomics technologies with high resolution often lack high sensitivity in mRNA detection. Here we report a dendrimeric DNA coordinate barcoding design for spatial RNA sequencing (Decoder-seq), which offers both high sensitivity and high resolution. Decoder-seq combines dendrimeric nanosubstrates with microfluidic coordinate barcoding to generate spatial arrays with a DNA density approximately ten times higher than previously reported methods while maintaining flexibility in resolution. We show that the high RNA capture efficiency of Decoder-seq improved the detection of lowly expressed olfactory receptor (Olfr) genes in mouse olfactory bulbs and contributed to the discovery of a unique layer enrichment pattern for two Olfr genes. The near-cellular resolution provided by Decoder-seq has enabled the construction of a spatial single-cell atlas of the mouse hippocampus, revealing dendrite-enriched mRNAs in neurons. When applying Decoder-seq to human renal cell carcinomas, we dissected the heterogeneous tumor microenvironment across different cancer subtypes and identified spatial gradient-expressed genes related to epithelial–mesenchymal transition with the potential to predict tumor prognosis and progression.

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: Decoder-seq workflow.
Fig. 2: Characterization of 3D dendrimeric DNA arrays.
Fig. 3: Spatially resolved transcriptome analysis of MOBs with Decoder-seq.
Fig. 4: Decoder-seq resolves the mouse hippocampus at near-cellular resolution.
Fig. 5: Resolving spatial heterogeneity in RCCs with Decoder-seq.
Fig. 6: Deciphering the potential spatial evolutionary trajectory of tumor invasion in RCCs.

Similar content being viewed by others

Data availability

Decoder-seq datasets (including raw sequences, expression matrices, Space Ranger output and image files), one 10x Visium dataset and ISS datasets have been deposited in NCBI’s Gene Expression Omnibus archive under accession number GSE235896 (ref. 66). DBiT data were obtained from NCBI (GSE137986), and Stereo-seq data of the MOB were downloaded from the China National GeneBank DataBase (https://db.cngb.org/stomics/mosta/download/). The MOB scRNA-seq data were downloaded from NCBI (GSE121891). The Ex-ST and Slide-seqV2 MOB datasets were downloaded from https://data.mendeley.com/datasets/nrbsxrk9mp/1 and https://singlecell.broadinstitute.org/single_cell/study/SCP354/slide-seq-study#study-download, respectively.

The Slide-seqV2 datasets for Olfr gene analysis were downloaded from NCBI (GSE169012). Two public 10x Visium MOB datasets were obtained from the 10x Genomics website (https://www.10xgenomics.com/resources/datasets/adult-mouse-olfactory-bulb-1-standard-1) and NCBI (GSE153859).

The single-cell mouse hippocampus reference was obtained from the DropViz website (http://dropviz.org/?_state_id_=2b493beb4e780071). The Stereo-seq and MERFISH mouse hippocampus datasets were obtained from https://db.cngb.org/stomics/mosta/download/ and https://download.brainimagelibrary.org/29/3c/293cc39ceea87f6d/coronal_2/220501_wb3_co2_15_5z18R_merfish5/, respectively. The scRNA-seq reference datasets for ccRCC and chRCC were obtained from the European Genome–Phenome Archive (EGAD00001008030) and NCBI (GSE159115 andGSE152938), respectively. Bulk RNA-seq data for ccRCC and chRCC were downloaded from TCGA (https://portal.gdc.cancer.gov). Source data are provided with this paper.

Code availability

Code related to this manuscript can be found at https://github.com/songjiajia2018/Decoder-seq.git, https://github.com/songjiajia2018/seg2knn.git and https://github.com/songjiajia2018/dynamicst.git.

References

  1. Palla, G., Fischer, D. S., Regev, A. & Theis, F. J. Spatial components of molecular tissue biology. Nat. Biotechnol. 40, 308–318 (2022).

    Article  CAS  PubMed  Google Scholar 

  2. Rao, A., Barkley, D., Franca, G. S. & Yanai, I. Exploring tissue architecture using spatial transcriptomics. Nature 596, 211–220 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chen, Y. et al. Mapping gene expression in the spatial dimension. Small Methods 5, e2100722 (2021).

    Article  PubMed  Google Scholar 

  4. Moses, L. & Pachter, L. Museum of spatial transcriptomics. Nat. Methods 19, 534–546 (2022).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Alon, S. et al. Expansion sequencing: spatially precise in situ transcriptomics in intact biological systems. Science 371, eaax2656 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chang, T. et al. Rapid and signal crowdedness-robust in situ sequencing through hybrid block coding. Proc. Natl Acad. Sci. USA 120, e2309227120 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Moffitt, J. R., Lundberg, E. & Heyn, H. The emerging landscape of spatial profiling technologies. Nat. Rev. Genet. 23, 741–759 (2022).

    Article  CAS  PubMed  Google Scholar 

  9. Ståhl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science 353, 78–82 (2016).

    Article  PubMed  Google Scholar 

  10. Vickovic, S. et al. High-definition spatial transcriptomics for in situ tissue profiling. Nat. Methods 16, 987–990 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rodriques, S. G. et al. Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science 363, 1463–1467 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Stickels, R. R. et al. Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2. Nat. Biotechnol. 39, 313–319 (2021).

    Article  CAS  PubMed  Google Scholar 

  13. Cho, C. S. et al. Microscopic examination of spatial transcriptome using Seq-Scope. Cell 184, 3559–3572 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Chen, A. et al. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell 185, 1777–1792 (2022).

    Article  CAS  PubMed  Google Scholar 

  15. Fu, X. et al. Polony gels enable amplifiable DNA stamping and spatial transcriptomics of chronic pain. Cell 185, 4621–4633 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Longo, S. K., Guo, M. G., Ji, A. L. & Khavari, P. A. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat. Rev. Genet. 22, 627–644 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhu, K. W. et al. Decoding the olfactory map through targeted transcriptomics links murine olfactory receptors to glomeruli. Nat. Commun. 13, 5137 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang, I. H. et al. Spatial transcriptomic reconstruction of the mouse olfactory glomerular map suggests principles of odor processing. Nat. Neurosci. 25, 484–492 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kvastad, L. et al. The spatial RNA integrity number assay for in situ evaluation of transcriptome quality. Commun. Biol. 4, 57 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Liu, Y. et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell 183, 1665–1681 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Arima, H. & Keiichi, M. Recent findings concerning PAMAM dendrimer conjugates with cyclodextrins as carriers of DNA and RNA. Sensors 9, 6346–6361 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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  PubMed  Google Scholar 

  23. Rouhanifard, S. H. et al. ClampFISH detects individual nucleic acid molecules using click chemistry-based amplification. Nat. Biotechnol. 37, 84–89 (2019).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. Fan, Y. et al. Expansion spatial transcriptomics. Nat. Methods 20, 1179–1182 (2023).

  26. Chéret, J. et al. Olfactory receptor OR2AT4 regulates human hair growth. Nat. Commun. 9, 3624 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Littman, R. et al. Joint cell segmentation and cell type annotation for spatial transcriptomics. Mol. Syst. Biol. 17, e10108 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, M. et al. Molecularly defined and spatially resolved cell atlas of the whole mouse brain. Nature 624, 343–354 (2023).

  29. Yoon, Y. J. et al. Glutamate-induced RNA localization and translation in neurons. Proc. Natl Acad. Sci. USA 113, E6877–E6886 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Steward, O. & Worley, P. Local synthesis of proteins at synaptic sites on dendrites: role in synaptic plasticity and memory consolidation? Neurobiol. Learn. Mem. 78, 508–527 (2002).

    Article  CAS  PubMed  Google Scholar 

  31. Kosik, K. S. Life at low copy number: how dendrites manage with so few mRNAs. Neuron 92, 1168–1180 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Tushev, G. et al. Alternative 3′ UTRs modify the localization, regulatory potential, stability, and plasticity of mRNAs in neuronal compartments. Neuron 98, 495–511 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Ainsley, J. A., Drane, L., Jacobs, J., Kittelberger, K. A. & Reijmers, L. G. Functionally diverse dendritic mRNAs rapidly associate with ribosomes following a novel experience. Nat. Commun. 5, 4510 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Nakayama, K. et al. RNG105/caprin1, an RNA granule protein for dendritic mRNA localization, is essential for long-term memory formation. eLife 6, e29677 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Fu, T. et al. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response. J. Hematol. Oncol. 14, 98 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Zhang, Y. et al. Single-cell analyses of renal cell cancers reveal insights into tumor microenvironment, cell of origin, and therapy response. Proc. Natl Acad. Sci. USA 118, e2103240118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Su, C. et al. Single-cell RNA sequencing in multiple pathologic types of renal cell carcinoma revealed novel potential tumor-specific markers. Front. Oncol. 11, 719564 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kansler, E. R. et al. Cytotoxic innate lymphoid cells sense cancer cell-expressed interleukin-15 to suppress human and murine malignancies. Nat. Immunol. 23, 904–915 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sanchez, D. J. & Simon, M. C. Genetic and metabolic hallmarks of clear cell renal cell carcinoma. Biochim. Biophys. Acta Rev. Cancer 1870, 23–31 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hsieh, J. J., Le, V., Cao, D., Cheng, E. H. & Creighton, C. J. Genomic classifications of renal cell carcinoma: a critical step towards the future application of personalized kidney cancer care with pan-omics precision. J. Pathol. 244, 525–537 (2018).

    Article  PubMed  Google Scholar 

  41. Certo, M. et al. Endothelial cell and T‐cell crosstalk: targeting metabolism as a therapeutic approach in chronic inflammation. Br. J. Pharmacol. 178, 2041–2059 (2021).

    Article  CAS  PubMed  Google Scholar 

  42. de Visser, K. E. & Joyce, J. A. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell 41, 374–403 (2023).

    Article  PubMed  Google Scholar 

  43. Kareva, I. Metabolism and gut microbiota in cancer immunoediting, CD8/Treg ratios, immune cell homeostasis, and cancer (immuno)therapy: concise review. Stem Cells 37, 1273–1280 (2019).

    Article  PubMed  Google Scholar 

  44. Lewis, S. M. et al. Spatial omics and multiplexed imaging to explore cancer biology. Nat. Methods 18, 997–1012 (2021).

    Article  CAS  PubMed  Google Scholar 

  45. Gulati, G. S. et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science 367, 405–411 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Szabo, P. M. et al. Cancer-associated fibroblasts are the main contributors to epithelial-to-mesenchymal signatures in the tumor microenvironment. Sci. Rep. 13, 3051 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jiang, F. et al. Simultaneous profiling of spatial gene expression and chromatin accessibility during mouse brain development. Nat. Methods 20, 1048–1057 (2023).

  48. Zhang, J. et al. DNA nanolithography enables a highly ordered recognition interface in a microfluidic chip for the efficient capture and release of circulating tumor cells. Angew. Chem. 132, 14219–14223 (2020).

    Article  Google Scholar 

  49. Vickovic, S. et al. SM-Omics is an automated platform for high-throughput spatial multi-omics. Nat. Commun. 13, 795 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Liu, Y. et al. High-plex protein and whole transcriptome co-mapping at cellular resolution with spatial CITE-seq. Nat. Biotechnol. 41, 1405–1409 (2023).

  51. Ben-Chetrit, N. et al. Integration of whole transcriptome spatial profiling with protein markers. Nat. Biotechnol. 41, 788–793 (2023).

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

    Article  CAS  PubMed  Google Scholar 

  53. Dries, R. et al. Giotto: a toolbox for integrative analysis and visualization of spatial expression data. Genome Biol. 22, 78 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Yang, M. et al. Spatiotemporal insight into early pregnancy governed by immune-featured stromal cells. Cell 186, 4271–4288 (2023).

  56. Wang, W. et al. Lymphatic endothelial transcription factor TBX1 promotes an immunosuppressive microenvironment to facilitate post-myocardial infarction repair. Immunity 56, 2342–2357 (2023).

  57. Wu, R. et al. Comprehensive analysis of spatial architecture in primary liver cancer. Sci. Adv. 7, eabg3750 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Aran, D., Hu, Z. & Butte, A. J. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 18, 220 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. He, Y., Jiang, Z., Chen, C. & Wang, X. Classification of triple-negative breast cancers based on immunogenomic profiling. J. Exp. Clin. Cancer Res. 37, 327 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Andreatta, M. & Carmona, S. J. UCell: robust and scalable single-cell gene signature scoring. Comput. Struct. Biotechnol. J. 19, 3796–3798 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Schubert, M. et al. Perturbation-response genes reveal signaling footprints in cancer gene expression. Nat. Commun. 9, 20 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Li, R. et al. Mapping single-cell transcriptomes in the intra-tumoral and associated territories of kidney cancer. Cancer Cell 40, 1583–1599 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kuppe, C. et al. Spatial multi-omic map of human myocardial infarction. Nature 608, 766–777 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kent, L. N. & Leone, G. The broken cycle: E2F dysfunction in cancer. Nat. Rev. Cancer 19, 326–338 (2019).

    Article  CAS  PubMed  Google Scholar 

  65. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Cao, J. et al. Decoder-seq enhances mRNA capture efficiency in spatial RNA sequencing. Gene Expression Omnibus https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE235896 (2023).

Download references

Acknowledgements

We thank D. Yang and S. Yang for their assistance in AFM characterization, X. Xu for early discussions in experimental design, X. Lin and Z. Wu for their assistance in bioinformatics analysis and W. Shi, J. Feng and S. Huang for the preparation of microfluidic chips. C.Y. was supported by the National Natural Science Foundation of China (22293031 and 21927806), Innovative Research Team of High-Level Local Universities in Shanghai (SHSMU-ZLCX20212601) and the Fundamental Research Funds for the Central Universities (20720210001 and 20720220005). L.W. was supported by the National Natural Science Foundation of China (22004083) and Shanghai Rising-Star Program (23QA1408200). J.S. was supported by the National Natural Science Foundation of China (22104080). J.C. was supported by the China Postdoctoral Science Foundation (2021M702167).

Author information

Authors and Affiliations

Authors

Contributions

C.Y. conceived the project. C.Y., L.W. and J.C. designed the experiments. J.S. designed the bioinformatics analysis. J.C., Z.Z. and T.L. performed the experiments. J.C., L.W., J.S., Z.Z., D.S. and R.C. analyzed the data. J.C., X.C. and Y.A. fabricated microfluidic devices. T.L. prepared the microfluidic chip. Z.Z. collected the biopsy samples. L.W. and J.C. wrote the paper. C.Y., L.W., J.S. and J.Z. revised the paper and supervised the project.

Corresponding authors

Correspondence to Junhua Zheng, Jia Song, Lingling Wu or Chaoyong Yang.

Ethics declarations

Competing interests

C.Y. is a co-founder of Dynamic Biosystems. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Qing Nie and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Spatially-localized cDNA synthesis.

(a) Fluorescence image of a MOB tissue section stained with nucleic acid dyes (SYBR Green). (b) Fluorescence image of cDNA containing Cy3-labeled dCTP after permeabilization, RT and tissue removal. (c) Magnified view of the section outlined in white dashed box in (a). (d) Magnified view of the section outlined in white dashed box in (b). (e–g) Magnified views of the sections outlined in white dashed box in (c). (e’-g’) Magnified views of the sections outlined in white dashed box in (d). The distribution of fluorescent cDNAs showed a similar pattern to that of the nucleic acid-stained tissue section after tissue removal, indicating that the cDNAs were localized to the areas under the cells and there was negligible diffusion. White triangle arrowheads point to the nuclei.

Source data

Extended Data Fig. 2 Quality evaluation and comparison of MOB data.

(a) Distribution of cDNA amplicon sizes. (b) Distribution of DNA sequencing library sizes. (c) Gene consistency of results generated from two replicate MOB sections 50-μm-spot Decoder-seq (Spearman’s correlation coefficient R = 0.96, p < 0.001). (d) Distribution of detected UMIs. UMIs(left) and genes (right) per spot from two replicate MOB sections with 50-μm-spot Decoder-seq. (e-f) Average UMI (left) and gene (right) number per spot (e) and per μm2 (f) for 50-μm-spot Decoder-seq and 55-μm-spot 10× Visium in varying sequencing reads. (g) Distribution of detected UMIs (left) and genes (right) from two replicate MOB sections with 15-μm-spot Decoder-seq.

Source data

Extended Data Fig. 3 Spatial gene expression patterns of region-specific marker genes in MOBs.

(a) Spatial gene expression (scaled) patterns of region-specific marker genes as determined with 15-μm-spot Decoder-seq. The marker genes and corresponding regions were Pcp4 (GCL), Cdhr1 and Eomes (MCL), Kit (EPL), Nrsn1 (GL), and Ptn and Omp (ONL). (b) Spatial gene expression (scaled) patterns of region-specific marker genes in MOBs as determined with 50-μm-spot Decoder-seq. The marker genes and corresponding regions were Sox11 (RMS), Cdhr1 and Eomes (MCL), Kit (EPL), Nrsn1 (GL), and Ptn and Omp (ONL). (c) Unsupervised clustering of the MOB section detected by Decoder-seq at 50 μm spatial resolution (left), and magnified views of RMS regions (right, black dashed box).

Extended Data Fig. 4 An integration and comparison of Decoder-seq data with single-cell data and other similar spatial methods.

(a, b) Integration of scRNA-seq data with Ex-ST data and Slide-seqV2 data, respectively. Cell types represented by different colors belong to scRNA-seq and black spots belong to Ex-ST data or Slide-seqV2. (c, d) Spatial distribution of spots annotated by cell types from scRNA-seq in Ex-ST (c) and Slide-seqV2 (d). The Ex-ST was merged with its corresponding DAPI image (blue). Slide-seqV2 only displays a partial region for easy comparison. (e) Comparison of relative abundances of major cell types detected by scRNA-seq, Decoder-seq, Ex-ST, and Slide-seqV2. The cell-type abundances revealed by these four methods are almost similar, with slight discrepancies that may arise from partial release and capture of tissue mRNA using SRT method and cell losses during tissue cell dissociation in scRNA-seq. (f) Comparison of the Spearman’s correlation coefficient, using two-tailed, two-sample t-test, among the degree of similarity between the average feature expression profiles of the single-cell reference and the average feature expression profiles of each cell type of scRNA-seq-based-annotation in Decoder-seq, Ex-ST, or Slide-seqV2, respectively.

Source data

Extended Data Fig. 5 Analysis of the Olfr gene in the MOBs.

(a) GO enrichment analysis of upregulated genes of glomeruli spots comparing to other spots in GL and ONL. (b) Statistical analysis of the disparities in distribution of layer-enriched spots, namely LC, with Olfr1033 or Olfr32 expression across ONL, GL, and other layers of the MOB (chi-squared test). (c) Spatial expression pattern of Olfr1033 and Olfr32 in the MOB derived from ISH data. The ISH data were subdivided into 15 regions from interior to exterior, and the Olfr1033 or Olfr32 gene expression in each region was normalized according to the region area and scaled from 0 to 1. (d, e) Spatial visualization of the raw count of Olfr1033 or Olfr32 genes in Decoder-seq (15 μm, n = 2) and Decoder-seq (50 μm, n = 2) (d), and 10x Visium (55 μm), Slide-seqV2 (10 μm), and Stereo-seq (bin21, 15 μm) (e). In Slide-seqV2, the white arrows point to the spot expressing marker genes of GL (low-magnification on left), and the Olfr1033 or Olfr32 (high-magnification on right). In Stereo-seq, magnified views of a portion within the white dashed boxes on the corresponding left. (f) Correlation matrix of UCell score-based LC-enriched GO terms (categorized as micro-programs) based on profiles across ONL and GL spots. The black dashed box represents the GO meta-programs; the black arrow indicates an LC-specific meta-program containing terms enriched in both LG and LO. The black dashed arrow shows the major function of LC-specific meta-program. (g) Spatial visualization of LC-specific meta-programs of MOBs in 15 μm-spot Decoder-seq (n = 3). (h) Violin plots with statistical analysis using two-tailed, two-sample t-test of the UCell score of LC-specific meta-programs in LC comparing to non-LC layers across the ONL and GL layers. LC: layer cluster; LG: LC compared to non-LC in GL; LO: LC compared to non-LC in ONL.

Source data

Extended Data Fig. 6 Comparison between Decoder-seq, MERFISH, and Stereo-seq data.

(a) A spatial single-cell atlas of mouse hippocampus derived from Stereo-seq data (left) and MERFISH data (right). (b) Comparison of relative abundances of major cell types detected by Decoder-seq, Stereo-seq, and MERFISH. Slight variations in the relative abundance of certain cell populations might be attributed to technological differences between imaging-based and sequencing-based methodologies, as well as their use of different sample sections. For example, neurogenesis appears more abundant in both Decoder-seq and Stereo-seq datasets, while being relatively rare in the MERFISH dataset. Neurogenesis mainly occurs in SGZ of the hippocampal dentate gyrus, where various cell types such as astrocytes, interneurons, endothelial cells, dentate, oligodendrocytes, cajal-retzius, and others are densely stacked together. For MERFISH, assigning mRNAs based on a small number of marker genes (1147 genes for 123 subclass markers) to cell types, especially in tissue regions of high-density cell populations, may result in a loss in certain cell types. Accurately assigning bins to cells also remains computationally challenging in Stereo-seq. However, since Stereo-seq is essentially a spatially barcoding array-based sequencing method, our Decoder-seq exhibits greater similarity in spatial distribution of cells to Stereo-seq than MERFISH. Overall, Decoder-seq with 15 μm spots demonstrated near cellular resolution and potential for finer-scale spatial analysis. SGZ, sub-granular zone.

Source data

Extended Data Fig. 7 Resolving the spatial heterogeneity in RCCs with Decoder-seq, related to Fig. 5.

(a) Unsupervised clustering of Decoder-seq RCC data (upper panel); the proportion of each cluster in each sample (lower panel). (b) The spatial pie plot of CARD-inferred cell type composition.   (c) CN unsupervised clustering of the shared cluster. (d) H&E images of ccRCC-2-L, ccRCC-3-L, and chRCC-1-L. Distinct categories by pathological annotation for morphological regions, including normal kidney (dark blue), stroma (green), blood vessel (red), and RCC (black). (e) Bubble plot showing differential analysis of CN 1 and CN 2 based on UCell scoring with identified marker genes. (f) Comparison of Treg abundance in CN 1 and CN 2. (g) Comparison of CIBERSORTx-inferred Treg abundance in ccRCC and chRCC subtype samples based on data from TCGA. (h) Spatial projection of the angiogenic pathway in each tissue section.

Source data

Extended Data Fig. 8 The Decoder-seq data with a resolution of 10 μm.

(a) AutoCAD representations of PDMS chips with 10 μm width barcoding channels. Upper: 50 channels of 10 μm in width; Lower: 96 channels of 10 μm in width. (b) Fluorescence images of 3D dendrimeric DNA arrays consisting of 10 μm spots. Alexa 488-labeled DNA barcode Y was ligated to the immobilized DNA barcode X at the intersections of the first set and the second set of flows, generating square fluorescent spots. (c) An H&E image of the MOB and its corresponding spatial UMI map by RNA captures from a single-cell resolution. The spatial UMI count map derived from 10 μm-spot Decoder-seq exhibited fine structures with MOB morphology. (d) Spatial raw expression of Ptgds, Fabp7, Doc2g, and Pcp4 in MOB section.

Supplementary information

Supplementary Information

Supplementary Figs. 1–16, Tables 1–5, 8, 9, 14 and 15, Methods and Decoder-seq protocol.

Reporting Summary

Supplementary Tables 6, 7 and 10–13.

Supplementary Table 6. List of all Olfr genes identified in 10x Visium data and Decoder-seq data. Supplementary Table 7a. Dendrite-enriched genes identified by Decoder-seq along with fold change enrichment and false discovery rate-corrected q value (P < 0.05, log (fold change) > 0.8). Supplementary Table 7b. Soma-enriched genes identified by Decoder-seq along with fold change enrichment and false discovery rate-corrected q value (P < 0.05, log (fold change) < −0.8). Supplementary Table 7c. List of genes that overlap with Ainsley et al.33, Tushev et al.32, Nakayama et al.34 and Slide-seqV2. Supplementary Table 10. List of cell signatures for cell scoring. Supplementary Table 11. List of spatially upregulated genes. Supplementary Table 12. GO enrichment analysis of spatially upregulated genes. Supplementary Table 13. List of dEMT genes.

Supplementary Data

Statistical source data.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cao, J., Zheng, Z., Sun, D. et al. Decoder-seq enhances mRNA capture efficiency in spatial RNA sequencing. Nat Biotechnol (2024). https://doi.org/10.1038/s41587-023-02086-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41587-023-02086-y

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