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:

Spatial multi-omics at subcellular resolution via high-throughput in situ pairwise sequencing

Abstract

Technology for spatial multi-omics aids the discovery of new insights into cellular functions and disease mechanisms. Here we report the development and applicability of multi-omics in situ pairwise sequencing (MiP-seq), a method for the simultaneous detection of DNAs, RNAs, proteins and biomolecules at subcellular resolution. Compared with other in situ sequencing methods, MiP-seq enhances decoding capacity and reduces sequencing and imaging costs while maintaining the efficacy of detection of gene mutations, allele-specific expression and RNA modifications. MiP-seq can be integrated with in vivo calcium imaging and Raman imaging, which enabled us to generate a spatial multi-omics atlas of mouse brain tissues and to correlate gene expression with neuronal activity and cellular biochemical fingerprints. We also report a sequential dilution strategy for resolving optically crowded signals during in situ sequencing. High-throughput in situ pairwise sequencing may facilitate the multidimensional analysis of molecular and functional maps of 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: Application of MiP-seq for the spatial profiling of biomolecules.
Fig. 2: A large-field-of-view 3D reconstruction and whole-mount spatial profiling of multiple genes.
Fig. 3: Spatial transcriptomic analysis of 100 genes in the nucleus of the HDB using MiP-seq.
Fig. 4: Detection of DNA, RNA, protein and neurotransmitter by MiP-seq.
Fig. 5: Combination of multiple imaging analysis with spatial gene profiles detected by MiP-seq.
Fig. 6: Sequential dilution strategy for resolving optically crowded signals.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. Raw and processed RNA-seq data are available from GEO (GSE190219). The source image data for this study are available via Zenodo at https://zenodo.org/records/10837475 (ref. 56). Publicly available datasets used in the study include GSE113576 and GSE74672. The data of conventional in situ hybridization used in this study were obtained from the Allen Institute of Brain Science (https://mouse.brain-map.org/search/index). Other cited data are all indicated with corresponding references in the paper.

Code availability

The custom written package and scripts used in this study are available in GitHub at https://github.com/GangCaoLab/InnerEye.

References

  1. Regev, A. et al. Science forum: the human cell atlas. eLife 6, e27041 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Sonawane, A. R. et al. Understanding tissue-specific gene regulation. Cell Rep. 21, 1077–1088 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Regev, A. et al. The human cell atlas white paper. Preprint at https://doi.org/10.48550/arXiv.1810.05192 (2018).

  4. 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 

  5. Echeverria, G. V. et al. High-resolution clonal mapping of multi-organ metastasis in triple negative breast cancer. Nat. Commun. 9, 5079 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Merino, D. et al. Barcoding reveals complex clonal behavior in patient-derived xenografts of metastatic triple negative breast cancer. Nat. Commun. 10, 766 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lan, X. et al. Fate mapping of human glioblastoma reveals an invariant stem cell hierarchy. Nature 549, 227–232 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lee, J. H. Quantitative approaches for investigating the spatial context of gene expression. Wiley Interdiscip. Rev. Syst. Biol. Med. 9, e1369 (2017).

    Article  PubMed  Google Scholar 

  9. 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 

  10. Eng, C.-H. L. et al. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH+. Nature 568, 235–239 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Takei, Y. et al. Integrated spatial genomics reveals global architecture of single nuclei. Nature 590, 344–350 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. Spatially resolved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  13. 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 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Qian, X. et al. Probabilistic cell typing enables fine mapping of closely related cell types in situ. Nat. Methods 17, 101–106 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Gyllborg, D. et al. Hybridization-based in situ sequencing (HybISS) for spatially resolved transcriptomics in human and mouse brain tissue. Nucleic Acids Res. 48, e112 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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 

  18. Sountoulidis, A. et al. SCRINSHOT enables spatial mapping of cell states in tissue sections with single-cell resolution. PLoS Biol. 18, e3000675 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lee, J. H. et al. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat. Protoc. 10, 442–458 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang, X. et al. Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science 361, eaat5691 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Fürth, D., Hatini, V. & Lee, J. H. In situ transcriptome accessibility sequencing (INSTA-seq). Preprint at bioRxiv https://doi.org/10.1101/722819 (2019).

  22. 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 

  23. Chen, F. et al. Nanoscale imaging of RNA with expansion microscopy. Nat. Methods 13, 679–684 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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 

  25. 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 

  26. 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 

  27. Liu, Y. et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell 183, 1665–1681.e18 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Close, J. L., Long, B. R. & Zeng, H. Spatially resolved transcriptomics in neuroscience. Nat. Methods 18, 23–25 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Burgess, D. J. Spatial transcriptomics coming of age. Nat. Rev. Genet. 20, 317 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Asp, M., Bergenstråhle, J. & Lundeberg, J. Spatially resolved transcriptomes—next generation tools for tissue exploration. BioEssays 42, 1900221 (2020).

    Article  Google Scholar 

  31. Jin, J., Vaud, S., Zhelkovsky, A. M., Posfai, J. & McReynolds, L. A. Sensitive and specific miRNA detection method using SplintR Ligase. Nucleic Acids Res. 44, e116 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Krzywkowski, T. & Nilsson, M. Fidelity of RNA templated end-joining by chlorella virus DNA ligase and a novel iLock assay with improved direct RNA detection accuracy. Nucleic Acids Res. 45, e161 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liu, S. et al. Barcoded oligonucleotides ligated on RNA amplified for multiplexed and parallel in situ analyses. Nucleic Acids Res. 49, e58 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lohman, G. J., Zhang, Y., Zhelkovsky, A. M., Cantor, E. J. & Evans, T. C. Jr Efficient DNA ligation in DNA—RNA hybrid helices by Chlorella virus DNA ligase. Nucleic Acids Res. 42, 1831–1844 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Krzywkowski, T., Kühnemund, M. & Nilsson, M. Chimeric padlock and iLock probes for increased efficiency of targeted RNA detection. RNA 25, 82–89 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Choi, H. M. T. et al. Third-generation in situ hybridization chain reaction: multiplexed, quantitative, sensitive, versatile, robust. Development 145, dev165753 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yap, E. L. et al. Bidirectional perisomatic inhibitory plasticity of a Fos neuronal network. Nature 590, 115–121 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Lim, S. H. et al. Nanoplasmonic immunosensor for the detection of SCG2, a candidate serum biomarker for the early diagnosis of neurodevelopmental disorder. Sci. Rep. 11, 22764 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Chen, X. et al. Huntingtin-associated protein 1 in mouse hypothalamus stabilizes glucocorticoid receptor in stress response. Front. Cell Neurosci. 14, 125 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mackenzie, K. D. et al. Huntingtin-associated protein-1 (HAP1) regulates endocytosis and interacts with multiple trafficking-related proteins. Cell Signal. 35, 176–187 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Kang, Z. et al. Molecular and genetic evidence for the PDGFRα-independent population of oligodendrocyte progenitor cells in the developing mouse brain. J. Neurosci. 38, 9505 (2018).

    Article  Google Scholar 

  42. Goltsev, Y. et al. Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Cell 174, 968–981.e15 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bi, Y. et al. Near-resonance enhanced label-free stimulated Raman scattering microscopy with spatial resolution near 130 nm. Light Sci. Appl. 7, 81 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Asp, M., Bergenstråhle, J. & Lundeberg, J. Spatially resolved transcriptomes—next generation tools for tissue exploration. Bioessays 42, e1900221 (2020).

    Article  PubMed  Google Scholar 

  45. Muster, B., Rapp, A. & Cardoso, M. C. Systematic analysis of DNA damage induction and DNA repair pathway activation by continuous wave visible light laser micro-irradiation. AIMS Genet. 4, 47–68 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Strell, C. et al. Placing RNA in context and space—methods for spatially resolved transcriptomics. FEBS J. 286, 1468–1481 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. Träutlein, D., Deibler, M., Leitenstorfer, A. & Ferrando-May, E. Specific local induction of DNA strand breaks by infrared multi-photon absorption. Nucleic Acids Res. 38, e14 (2010).

    Article  PubMed  Google Scholar 

  48. van der Walt, S. et al. scikit-image: image processing in Python. PeerJ 2, e453 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Marstal, K., Berendsen, F., Staring, M. & Klein, S. SimpleElastix: a user-friendly, multi-lingual library for medical image registration. In 2016 IEEE Conference on Computer Vision and Pattern Recognition Workshops (CVPRW) 574–582 (IEEE, 2016).

  50. Stringer, C., Wang, T., Michaelos, M. & Pachitariu, M. Cellpose: a generalist algorithm for cellular segmentation. Nat. Methods 18, 100–106 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  52. 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 

  53. Pham, D. et al. Robust mapping of spatiotemporal trajectories and cell–cell interactions in healthy and diseased tissues. Nat. Commun. 14, 7739 (2023).

  54. Csárdi, G. & Nepusz, T. The Igraph Software Package for Complex Network Research. InterJournal Complex Syst. 1695 (2006).

  55. Villanueva, R. A. M. & Chen, Z. J. ggplot2: elegant graphics for data analysis (2nd ed.). Measurement 17, 160–167 (2019).

    Google Scholar 

  56. Wu, X. et al. Development of multiomics in situ pairwise sequencing (MIP-seq) for single-cell resolution multidimensional spatial omics Version 4. Zenodo https://doi.org/10.1101/2023.01.07.523058 (2023).

Download references

Acknowledgements

We thank F. Yang (Huazhong Agricultural University) for helping with tissue processing; Z. Hu, Y. Li and C. Shi in the Core Facility at HZAU for technical support in imaging; Z. Zhang (Spatial FISH Co., Ltd) for technical support in FISH; M. Liang for helping with early experimental design; X. Li and J. Li for helping with RCA; Xiaojian Cao for helping with statistical analysis on differential gene expression in pathogen infection assay; R Tyagi for proofreading; J. He, H. Zhang and M. Jin (State Key Laboratory of Neuroscience, Institute of Neuroscience, Centre for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences) for providing zebrafish brain and related help; X. Li (Hubei Cancer Hospital) for providing human diffuse large B cell lymphoma samples; Z. Huang (Huazhong University of Science and Technology) for helping with SRS imaging; Wuhan Keqian Biology Company for providing PCV2 and CSFV virus. This work was supported by the National Natural Science Foundation of China (Grant No. 32221005,32171022, U21A20259, 31872470 and 81827901).

Author information

Authors and Affiliations

Authors

Contributions

G.C., J.D. and X.W. conceived and designed the project. X.W. performed probe-library design and preparation, cell and tissue experiment processing and sample preparation, in situ sequencing experiments, imaging, and data analysis. W.X. performed image-processing-pipeline development, in situ image processing, and analysis of the spatial patterns of cells and genes. W.X., C.W. and Y.Z. performed probe-design-software development, spatial-gene-decoding-pipeline design, and analysis. L.D. performed the spatial trajectory and spatial Delaunay network analysis. Y.L. performed C6 glioma-model establishment, Raman imaging analysis and the vascular-imaging experiment. Z.W. performed gene in situ verification in brain tissue, the parental orthogonal experiment, methylation in situ detection and in situ hybridization of the Raman tissue slice experiment. L.S. and G.D. performed conjugation of nucleic acid to antibody and in situ detection of proteins. A.G. performed MiP-seq and HCR3.0 efficiency comparison experiments. H.W. performed MiP-seq in situ DNA detection. X.Y. performed in vivo calcium imaging and analysis. K.Y. performed RNA-seq library sequencing for C6 cells and offspring brain tissue for the parental orthogonal experiment. L.Z. and L.Y. performed neuron culture and efficiency verification. Z.L. and D.L. performed bioinformatic analysis. J.Y. performed pathological diagnosis of DLBCL samples. G.C., J.D., Y.H., Z.F., P.W., X.W., W.X. and L.D. wrote the paper. All authors read and approved the final paper.

Corresponding authors

Correspondence to Jinxia Dai or Gang Cao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Rong Fan, Ankur Sharma and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 In situ detection of gene expression in brain slices via MiP-seq.

a, b, The expression patterns of Cck, Penk, Crh, Oxt, Avp, Nov, Gad1, Npy, Cux2, Rorb, and Sst genes detected by MiP-seq were comparable to that by conventional in situ hybridization (from Allen Institute of Brain Science). c, In situ co-detection of ten genes (Kif5a, Snhg11, Tau, Gja1, Nr4a1, Rbfox3, Dpf1, Map2, Thy1, and Eef2) in mice brain slices based on dual barcode sequencing. Different pseudo-colours indicate the corresponding gene transcripts of ten genes, which display distinct subcellular localization (Kif5a in cytoplasm; Snhg11 in nucleus). The subcellular localization of Kif5a, Snhg11, Tau, and Eef2 genes transcripts are presented in the lower panels respectively. The detection of these genes’ mRNA in a-c was replicated at least three times. Scale bars, 50 µm in left two panels of a, 1000 µm in right panel of a and middle panel of b, 200 µm in left two panels and right two panels of b, 10 µm in c.

Extended Data Fig. 2 Parallel comparison of in situ detection for 77 genes expression in preoptic area of mice between MiP-seq and MERFISH.

a, Detection of 77 genes in preoptic area (POA) by MiP-seq, which is for parallel comparison to their expression in coronal section of POA via MERFISH (data from the work of Moffitt et al., 2018, Science). The grey signal for visualizing the entire cell morphology were the whole cell mRNA signal detected by oligo-dT-Cy3. The DAPI image for the coronal section of POA were illustrated in the right-lower panel, and the white boxed area indicated the area for MiP-seq image. b, Comparison of 77 genes expression in POA detected by MiP-seq (n= 913 cells) with MERFISH (n= 841 cells). The Y-axis represents the values of the expression normalized by respective standards for each of the two methods. The detection of 77 genes expression in preoptic area of mice was replicated at least three times. Scale bars, 50 µm in upper image and 100 µm in lower image of a.

Extended Data Fig. 3 In situ detection of gene transcripts with single-base variance via MiP-seq.

a, Detection of single-base variance in human and mouse β-actin gene transcripts in cocultured HBMEC (human origin) cells and Bend3 (mouse origin) cells. Red and green dot signals indicate human β-actin gene (ACTB) and mouse β-actin gene (Actb) mRNA sequence, respectively. The detection of ACTB and Actb mRNA was replicated at least three times. b, In situ sequencing for allele-specific gene expression of Fam171b, Rpl15, Hba-a2, and Rps11 in hippocampus of the offspring mice from C57BL/6 female mice crossed with BALB/c male mice. Cell morphology stained with Oligo-dT(grey) were overlaid with corresponding gene expression profiles. The expression level of Fam171b, Rps11, Hba-a2, and Rpl15 alleles for each cell (RCPs per cell) in hippocampus were presented in the upper panels. RH, right hippocampus; LH, left hippocampus. Numbers indicate different mice, and the presented data were from three different mice. n=5412 cells in 1-RH detection, n=5979 cells in 2-LH detection, n=4538 cells in 3-LH detection. c, In situ sequencing for allele-specific gene expression of Fam171b, Rpl15, Hba-a2, and Rps11 in PVN of the offspring mice from C57BL/6 female mice crossed with BALB/c male mice. Cell morphology stained with Oligo-dT(grey) were overlaid with corresponding gene expression profiles. The quantification of Fam171b, Rps11, Hba-a2, and Rpl115 alleles expression for each cell (RCPs per cell) in PVN were presented in the upper panels. Numbers indicate different mice, and the data from three different mice were presented here. n=2471 cells in 1-PVN detection, n=2510 cells in 3-PVN detection, n=2418 cells in 4-PVN detection. d, The correlation of alleles expression in hippocampus and PVN from different mice. Numbers indicate different mice. RH, right hippocampus; LH, left hippocampus. e, In situ co-detection of mutations for multiple genes (Eno1, Nptxr, Spp1, Tnc, and Tuba1a) in glioma area of rat brain from C6 glioma model, and the box plot for the expression levels of wild-type and mutant variances was presented. The detection of allele-specific gene expression and the detection of Eno1, Nptxr, Spp1, Tnc, and Tuba1a mRNA was replicated at least three times. Scale bars, 10 µm in a, 50 µm in b and c, 30 µm in e.

Extended Data Fig. 4 Spatial profiling of multiple genes in mice brain at a large-field of view and 3D reconstruction of the spatial profiling of specific marker genes in the whole cerebellum.

a, Expression of Plp1, Neurod1, Gad1, and Calb1 in mice brain detected by MiP-seq is consistent with conventional in situ hybridization results from Allen Institute of Brain Science. b, The Left panel is the enlarged view of rectangle region in Fig. 2a for ten genes codetection in hippocampus. The gene expression patterns in hippocampus detected by MiP-seq are consistent with the results from Allen Institute of Brain Science (middle and right panels). c, 3D view of Calb1, Gad1, Neurod1, and Plp1 gene expression in the whole cerebellum are illustrated, respectively. The detection of Calb1, Gad1, Neurod1, and Plp1 gene expression in cerebellum was replicated at least three times. Scale bars, 1000 µm in a, 300 µm in b, 500 µm in c.

Extended Data Fig. 5 Image processing pipeline for spatial transcriptome analysis by MiP-seq.

a, Diagram of image processing pipeline for spatial transcriptome analysis by MiP-seq. The raw images from multiple rounds sequencing are first matched to the first-round image by affine transformations that allow rotation, translation, scaling, and shear transformations. The candidate points are selected according to the aligned images, and are decoded according to the intensity of each channel in each round at its location. The cell contour is calculated by cellpose based on the merged images from three sequencing rounds after alignment. Finally, the decoded gene points are allocated to neighbouring cells according to the principle of proximity, and the expression profile corresponding to each cell is obtained. b, The image (i) before registration. Three rounds of the channel merged images are represented in cyan, yellow, and magenta, respectively. c, The image (ii) after registration. d, Image and candidate spots in a local area. (i) Gray scale image of the combined channel images after alignment. (ii) Candidate spots are marked as white points on the image. (iii) Decoded spots are marked as points with different colours. The 'general channel' of each round is calculated by averaging the images from different signal channels. e, The cell mask is calculated from the averaged general channel image from three sequencing rounds by cellpose. Different cells are indicated by different colours. f, The results of gene assignment in this field of view. Different coloured dots on the graph represent different genes. The red connecting line represents the gene to the corresponding cell. Scale bars, 30 µm in left image of b and c, 10 µm in right image of b and c, 10 µm in d, 30 µm in e and f.

Extended Data Fig. 6 Spatial transcriptomic analysis of 100 genes in HDB by MiP-seq and gene co-expression between cell clusters.

a, 3D projection image for in situ detection of 100 genes mRNAs in the first sequencing round by MiP-seq in HDB. b, The images of three round sequencing by MiP-seq. The signals lined by the rectangle box and circle are further illustrated in the middle and lower panel respectively. c, The spatial distribution of neurotransmitter-related and peptide hormone genes (blue) and their receptor genes (yellow). When the counts of the corresponding gene in each cell is greater than 10, the whole cell is marked by the colour of the corresponding gene. d, Spatial expression analysis of Hap1 and Pdgfra genes in HDB. The unit for the X and Y axes is pixel. e, The spatial distance of Hap1 (green curve) and Pdgfra (red curve) gene to the top 50 expression genes. The 'mean min distance' refers to the average minimal distance (measured by mean pixels) from the signal points of Hap1 or Pdgfra gene transcripts to the nearest signal points for each of top 50 expressed genes. The unit for the Y axis is pixel. f, Cells of HDB are clustered into 6 groups (0–5) based on the expression profile of single cells detected by MiP-seq. g, UMAP of featured genes expression in certain cell clusters and the corresponding spatial expression pattern in HDB. n=521 cells for data analysis of d-g. Scale bars, 50 µm in a, 10 µm in b.

Extended Data Fig. 7 Spatial transcriptomic alteration of PVN upon Mycobacterium tuberculosis (M.tb) infection.

a, Detection of 217 genes including cell markers, hormones, receptors, as well as immune related genes in mice PVN upon Mycobacterium tuberculosis (M.tb) infection and saline control treatment by MiP-seq. b, Expression of Ly6h and Gpr165 were up-regulated while C1qb and Pgm2l1 were down-regulated in response to M.tb infection. c, Heatmap of the expression of cell marker genes. d, Differentially expressed genes in endothelial cells, astrocytes, microglia, and neurons. Genes with significantly changes (P value ≤0.05 and log(fold change) ≥0.3) were labelled in green. n= 491 cells in saline control, n= 533 cells in M.tb infection group. The spatial transcriptomic detection in PVN upon M.tb infection via MiP-seq was replicated at least three times. Statistical significance was measured by Student’s t-test (Two-tailed). The raw data of d can be found in Supplementary Table 3. Scale bars, 100 µm in DAPI images of a, 10 µm in multiplexed genes detection image of a and in b.

Extended Data Fig. 8 In situ detection of multiple genomic loci via MiP-seq.

a, The remaining data for co-detection of Rbfox3 and Nr4a1 genes in mice tissue by MiP-seq. Different cells are numbered. The data for #1 cell are shown in Fig. 4b. b, The intranuclear location of Rbfox3 and Nr4a1 loci for 20 cells in mice brain. c, The histogram of Euclidean distance between two Rbfox3 loci, two Nr4a1 loci, and the two loci to nuclear edge, respectively. n= 20 cells in each group. d, More images for in situ sequencing of Nptxr point mutation located in different chromosomes in C6 cell. Different cells are numbered. The data for #1 cell are shown in Fig. 4c. e, The intranuclear location of wildtype and mutant Nptxr loci for 20 C6 cells. f, The histogram of Euclidean distance between wildtype and mutant Nptxr loci, and between their locus to nuclear edge. n= 20 cells in each group. Data was presented as mean ± SD in c and f. The raw data can be found in Supplementary Table 3. The detection of these genomic loci was replicated at least three times. Scale bars, 2 µm in a, b and d, e.

Extended Data Fig. 9 In situ detection of neuropeptide and multiple proteins via MiP-seq.

a, Nearly identical signals for neuropeptide orexin detected by MiP-seq (Cy3 red fluorescence) and classic immunostaining (Alexa fluor 488 green fluorescence) were observed in lateral hypothalamus (LH) region of mice brain. For orexin detection by MiP-seq, the targeting oligonucleotides was conjugated with secondary antibody. b, In situ co-detection of four proteins (E2 of CSFV, Cap of PCV2, flag for IFNG, and Pol II) conjugated with distinct DNA oligonucleotides via MiP-seq. The comparison experiment between MiP-seq and classic immunostaining in Orexin detection and the codetection of four proteins (E2 of CSFV, Cap of PCV2, flag for IFNG, and Pol II) by MiP-seq was replicated at least three times. Scale bar, 50 µm in a, 20 µm in b.

Extended Data Fig. 10 Co-detection of proteins and mRNAs in tumour samples via MiP-seq.

a, Co-detection of 30 immune related proteins with 6 diagnostic marker genes mRNAs in human diffuse large B cell lymphoma (DLBCL) sections. The images for DAPI staining and oligo-dT-CY3 hybridization were illustrated in the right lower panel which are used for cell segmentation. b, UMAP for 16 cell clusters in DLBCL based on the analysis of expression profile of single cells (n=2597 cells) in a. c, The spatial map of 16 cell clusters. d, The heatmap of spatial interaction frequency among 16 cell clusters. e, The spatial map of three cell clusters showing high interaction frequency. f, The normalized spatial expression heatmap of detected proteins and mRNAs in DLBCL. g, The spatial expression of CD27, CD45RA, CCR7 and CXCR6 in DLBCL sections. The codetection of 30 proteins and 6 mRNAs in human DLBCL was replicated at least three times. Scale bar, 50 μm in a.

Supplementary information

Main Supplementary Information

Supplementary figures.

Reporting Summary

Peer Review File

Supplementary Video

3D gene-expression architecture for the whole cerebellum.

Supplemantary Data

Probe sequences.

Supplemantary Data 2

Raw data for efficiency comparison between different methods.

Supplemantary Data 3

Raw data for statistical analysis, and cell cluster genes list.

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

Wu, X., Xu, W., Deng, L. et al. Spatial multi-omics at subcellular resolution via high-throughput in situ pairwise sequencing. Nat. Biomed. Eng (2024). https://doi.org/10.1038/s41551-024-01205-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-024-01205-7

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