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:

Single-cell and spatial atlases of spinal cord injury in the Tabulae Paralytica

Abstract

Here, we introduce the Tabulae Paralytica—a compilation of four atlases of spinal cord injury (SCI) comprising a single-nucleus transcriptome atlas of half a million cells, a multiome atlas pairing transcriptomic and epigenomic measurements within the same nuclei, and two spatial transcriptomic atlases of the injured spinal cord spanning four spatial and temporal dimensions. We integrated these atlases into a common framework to dissect the molecular logic that governs the responses to injury within the spinal cord1. The Tabulae Paralytica uncovered new biological principles that dictate the consequences of SCI, including conserved and divergent neuronal responses to injury; the priming of specific neuronal subpopulations to upregulate circuit-reorganizing programs after injury; an inverse relationship between neuronal stress responses and the activation of circuit reorganization programs; the necessity of re-establishing a tripartite neuroprotective barrier between immune-privileged and extra-neural environments after SCI and a failure to form this barrier in old mice. We leveraged the Tabulae Paralytica to develop a rejuvenative gene therapy that re-established this tripartite barrier, and restored the natural recovery of walking after paralysis in old mice. The Tabulae Paralytica provides a window into the pathobiology of SCI, while establishing a framework for integrating multimodal, genome-scale measurements in four dimensions to study biology and medicine.

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 of the Tabulae Paralytica and the snRNA-seq atlas.
Fig. 2: Cell types and subtypes of the uninjured and injured mouse spinal cord.
Fig. 3: Biological principles governing the response to SCI.
Fig. 4: Failure to re-establish a tripartite neuroprotective barrier in old mice.
Fig. 5: A multiomic atlas of SCI.
Fig. 6: A spatial transcriptomic atlas of SCI.
Fig. 7: A 4D spatiotemporal atlas of SCI.
Fig. 8: A rejuvenative gene therapy re-establishes the tripartite barrier to restore walking.

Similar content being viewed by others

Data availability

Sequencing data have been deposited to the Gene Expression Omnibus (GSE234774, snRNA-seq and spatial transcriptomics, and GSE230765, multiome). Source data are provided with this paper.

Code availability

Augur, Libra and Magellan are available from GitHub (https://github.com/neurorestore/Augur, https://github.com/neurorestore/Libra and https://github.com/neurorestore/Magellan).

References

  1. Sofroniew, M. V. Dissecting spinal cord regeneration. Nature 557, 343–350 (2018).

    ADS  CAS  PubMed  Google Scholar 

  2. Anderson, M. A. et al. Natural and targeted circuit reorganization after spinal cord injury. Nature Neurosci. 25, 1584–1596 (2022).

  3. Courtine, G. & Sofroniew, M. V. Spinal cord repair: advances in biology and technology. Nat. Med. 25, 898–908 (2019).

    CAS  PubMed  Google Scholar 

  4. Ahuja, C. S. et al. Traumatic spinal cord injury. Nat. Rev. Dis. Primers 3, 17018 (2017).

    PubMed  Google Scholar 

  5. Rowald, A. et al. Activity-dependent spinal cord neuromodulation rapidly restores trunk and leg motor functions after complete paralysis. Nat. Med. 28, 260–271 (2022).

    CAS  PubMed  Google Scholar 

  6. Squair, J. W. et al. Neuroprosthetic baroreflex controls haemodynamics after spinal cord injury. Nature 590, 308–314 (2021).

    ADS  CAS  PubMed  Google Scholar 

  7. Wagner, F. B. et al. Targeted neurotechnology restores walking in humans with spinal cord injury. Nature 563, 65–71 (2018).

    ADS  CAS  PubMed  Google Scholar 

  8. Kathe, C. et al. The neurons that restore walking after paralysis. Nature 611, 540–547 (2022).

  9. Popovich, P. G., Wei, P. & Stokes, B. T. Cellular inflammatory response after spinal cord injury in Sprague-Dawley and Lewis rats. J. Comp. Neurol. 377, 443–464 (1997).

    CAS  PubMed  Google Scholar 

  10. Anderson, M. A. et al. Astrocyte scar formation aids central nervous system axon regeneration. Nature 532, 195–200 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Squair, J. W., Gautier, M., Sofroniew, M. V., Courtine, G. & Anderson, M. A. Engineering spinal cord repair. Curr. Opin. Biotech. 72, 48–53 (2021).

    CAS  PubMed  Google Scholar 

  12. Asboth, L. et al. Cortico-reticulo-spinal circuit reorganization enables functional recovery after severe spinal cord contusion. Nat. Neurosci. 21, 576–588 (2018).

    CAS  PubMed  Google Scholar 

  13. Scheff, S. W., Rabchevsky, A. G., Fugaccia, I., Main, J. A. & Lumpp, J. E. Experimental modeling of spinal cord injury: characterization of a force-defined injury device. J. Neurotraum. 20, 179–193 (2003).

    Google Scholar 

  14. Dusart, I. & Schwab, M. E. Secondary cell death and the inflammatory reaction after dorsal hemisection of the rat spinal cord. Eur. J. Neurosci. 6, 712–724 (1994).

    CAS  PubMed  Google Scholar 

  15. Zappia, L. & Oshlack, A. Clustering trees: a visualization for evaluating clusterings at multiple resolutions. Gigascience 7, giy083 (2018).

    PubMed  PubMed Central  Google Scholar 

  16. Milich, L. M. et al. Single-cell analysis of the cellular heterogeneity and interactions in the injured mouse spinal cord. J. Exp. Med. 218, e20210040 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Sofroniew, M. V. & Vinters, H. V. Astrocytes: biology and pathology. Acta Neuropathol. 119, 7–35 (2010).

    PubMed  Google Scholar 

  18. Faulkner, J. R. et al. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J. Neurosci. 24, 2143–2155 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Herrmann, J. E. et al. STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury. J. Neurosci. 28, 7231–7243 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wanner, I. B. et al. Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury. J. Neurosci. 33, 12870–12886 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zheng, S. C. et al. Universal prediction of cell-cycle position using transfer learning. Genome Biol. 23, 41 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Munji, R. N. et al. Profiling the mouse brain endothelial transcriptome in health and disease models reveals a core blood–brain barrier dysfunction module. Nat. Neurosci. 22, 1892–1902 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014.e22 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Yasuda, K. et al. Drug transporters on arachnoid barrier cells contribute to the blood–cerebrospinal fluid barrier. Drug Metab. Dispos. 41, 923–931 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Dewar, D., Underhill, S. M. & Goldberg, M. P. Oligodendrocytes and ischemic brain injury. J. Cereb. Blood Flow Metabol. 23, 263–274 (2002).

    Google Scholar 

  26. Petracca, Y. L. et al. The late and dual origin of cerebrospinal fluid-contacting neurons in the mouse spinal cord. Development 143, 880–891 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Vígh, B. et al. The system of cerebrospinal fluid-contacting neurons. Its supposed role in the nonsynaptic signal transmission of the brain. Histol. Histopathol. 19, 607–628 (2004).

    PubMed  Google Scholar 

  28. Squair, J. W. et al. Recovery of walking after paralysis by regenerating characterized neurons to their natural target region. Science 381, 1338–1345 (2023).

  29. Courtine, G. et al. Recovery of supraspinal control of stepping via indirect propriospinal relay connections after spinal cord injury. Nat. Med. 14, 69–74 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Skinnider, M. A. et al. Cell type prioritization in single-cell data. Nat. Biotechnol. 39, 30–34 (2021).

    CAS  PubMed  Google Scholar 

  31. Squair, J. W., Skinnider, M. A., Gautier, M., Foster, L. J. & Courtine, G. Prioritization of cell types responsive to biological perturbations in single-cell data with Augur. Nat. Protoc. 16, 3836–3873 (2021).

  32. Renthal, W. et al. Transcriptional reprogramming of distinct peripheral sensory neuron subtypes after axonal injury. Neuron 108, 128–144.e9 (2020).

  33. Nguyen, M. Q., Pichon, C. E. L. & Ryba, N. Stereotyped transcriptomic transformation of somatosensory neurons in response to injury. eLife 8, e49679 (2019).

    PubMed  PubMed Central  Google Scholar 

  34. Cajal, S. R. Y. & May, R. M. Cajal’s Degeneration and Regeneration of the Nervous System (Oxford Univ. Press, 1991).

  35. Sperry, R. W. Chemoaffinity in the orderly growth of nerve fiber patterns and connections. Proc. Natl Acad. Sci. USA 50, 703–710 (1963).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cajal, S. R. Y. Degeneration and Regeneration of the Nervous System (Oxford Univ. Press, 1928).

  37. Anderson, M. A. et al. Required growth facilitators propel axon regeneration across complete spinal cord injury. Nature 561, 396–400 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Harel, N. Y. & Strittmatter, S. M. Can regenerating axons recapitulate developmental guidance during recovery from spinal cord injury?. Nat. Rev. Neurosci. 7, 603–616 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Mironova, Y. A. & Giger, R. J. Where no synapses go: gatekeepers of circuit remodeling and synaptic strength. Trends Neurosci. 36, 363–373 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lin, A. C. & Holt, C. E. Local translation and directional steering in axons. EMBO J. 26, 3729–3736 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Short, D., Masry, W. E. & Jones, P. High dose methylprednisolone in the management of acute spinal cord injury–a systematic review from a clinical perspective. Spinal Cord 38, 273–286 (2000).

    CAS  PubMed  Google Scholar 

  42. Bracken, M. B. Efficacy of methylprednisolone in acute spinal cord injury. JAMA 251, 45 (1984).

    CAS  PubMed  Google Scholar 

  43. Hurlbert, R. J. Methylprednisolone for acute spinal cord injury: an inappropriate standard of care. J. Neurosurg. Spine 93, 1–7 (2000).

    CAS  Google Scholar 

  44. Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).

    CAS  PubMed  Google Scholar 

  45. Gal-Oz, S. T. et al. ImmGen report: sexual dimorphism in the immune system transcriptome. Nat. Commun. 10, 4295 (2019).

    ADS  PubMed  PubMed Central  Google Scholar 

  46. Mattucci, S. et al. Basic biomechanics of spinal cord injury—how injuries happen in people and how animal models have informed our understanding. Clin. Biomech. 64, 58–68 (2018).

    Google Scholar 

  47. O’Shea, T. M. et al. Lesion environments direct transplanted neural progenitors towards a wound repair astroglial phenotype in mice. Nat. Commun. 13, 5702 (2022).

    ADS  PubMed  PubMed Central  Google Scholar 

  48. Squair, J. W. et al. Confronting false discoveries in single-cell differential expression. Nat. Commun. 12, 5692 (2021).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Granja, J. M. et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nat. Genet. 53, 403–411 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Kang, J. B. et al. Efficient and precise single-cell reference atlas mapping with Symphony. Nat. Commun. 12, 5890 (2021).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  51. Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Schwarzschild, M. A., Cole, R. L. & Hyman, S. E. Glutamate, but not dopamine, stimulates stress-activated protein kinase and AP-1-mediated transcription in striatal neurons. J. Neurosci. 17, 3455–3466 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Burda, J. E. et al. Divergent transcriptional regulation of astrocyte reactivity across disorders. Nature 606, 557–564 (2022).

  54. Pollard, K. S., Hubisz, M. J., Rosenbloom, K. R. & Siepel, A. Detection of nonneutral substitution rates on mammalian phylogenies. Genome Res. 20, 110–121 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Christian, C. A. et al. Endogenous positive allosteric modulation of GABAA receptors by diazepam binding inhibitor. Neuron 78, 1063–1074 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Chung, K. & Deisseroth, K. CLARITY for mapping the nervous system. Nat. Methods 10, 508–513 (2013).

    CAS  PubMed  Google Scholar 

  57. Olsson, A.-K., Dimberg, A., Kreuger, J. & Claesson-Welsh, L. VEGF receptor signalling? in control of vascular function. Nat. Rev. Mol. Cell Bio. 7, 359–371 (2006).

    CAS  Google Scholar 

  58. Li, Y. et al. Microglia-organized scar-free spinal cord repair in neonatal mice. Nature 587, 613–618 (2020).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  59. Li, C. et al. Temporal and spatial cellular and molecular pathological alterations with single-cell resolution in the adult spinal cord after injury. Signal Transduct. Target Ther. 7, 65 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Wells, J. E. A., Hurlbert, R. J., Fehlings, M. G. & Yong, V. W. Neuroprotection by minocycline facilitates significant recovery from spinal cord injury in mice. Brain 126, 1628–1637 (2003).

    PubMed  Google Scholar 

  61. Stirling, D. P. et al. Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improves functional outcome after spinal cord injury. J. Neurosci. 24, 2182–2190 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Festoff, B. W. et al. Minocycline neuroprotects, reduces microgliosis, and inhibits caspase protease expression early after spinal cord injury. J. Neurochem. 97, 1314–1326 (2006).

    CAS  PubMed  Google Scholar 

  63. Rabchevsky, A. G., Fugaccia, I., Sullivan, P. G., Blades, D. A. & Scheff, S. W. Efficacy of methylprednisolone therapy for the injured rat spinal cord. J. Neurosci. Res. 68, 7–18 (2002).

    CAS  PubMed  Google Scholar 

  64. James, N. D. et al. Chondroitinase gene therapy improves upper limb function following cervical contusion injury. Exp. Neurol. 271, 131–135 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Muir, E. M. et al. Modification of N-glycosylation sites allows secretion of bacterial chondroitinase ABC from mammalian cells. J. Biotechnol. 145, 103–110 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Bartus, K. et al. Large-scale chondroitin sulfate proteoglycan digestion with chondroitinase gene therapy leads to reduced pathology and modulates macrophage phenotype following spinal cord contusion injury. J. Neurosci. 34, 4822–4836 (2014).

    PubMed  PubMed Central  Google Scholar 

  67. Esposito, M. S., Capelli, P. & Arber, S. Brainstem nucleus MdV mediates skilled forelimb motor tasks. Nature 508, 351–356 (2014).

    ADS  CAS  PubMed  Google Scholar 

  68. Kathe, C. et al. Wireless closed-loop optogenetics across the entire spinal cord in ecological environments. Nat. Biotechnol. 40, 198–208 (2022).

    CAS  PubMed  Google Scholar 

  69. Takeoka, A., Vollenweider, I., Courtine, G. & Arber, S. Muscle spindle feedback directs locomotor recovery and circuit reorganization after spinal cord injury. Cell 159, 1626–1639 (2014).

    CAS  PubMed  Google Scholar 

  70. Mathis, A. et al. DeepLabCut: markerless pose estimation of user-defined body parts with deep learning. Nat. Neurosci. 21, 1281–1289 (2018).

    CAS  PubMed  Google Scholar 

  71. Chung, K. et al. Structural and molecular interrogation of intact biological systems. Nature 497, 332–337 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  72. Tomer, R., Ye, L., Hsueh, B. & Deisseroth, K. Advanced CLARITY for rapid and high-resolution imaging of intact tissues. Nat. Protoc. 9, 1682–1697 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Voigt, F. F. et al. The mesoSPIM initiative: open-source light-sheet microscopes for imaging cleared tissue. Nat. Methods 16, 1105–1108 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  75. Caglayan, E., Liu, Y. & Konopka, G. Neuronal ambient RNA contamination causes misinterpreted and masked cell types in brain single-nuclei datasets. Neuron 110, 4043–4056.e5 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Fleming, S. J. et al. Unsupervised removal of systematic background noise from droplet-based single-cell experiments using CellBender. Nat. Methods 20, 1323–1335 (2023).

  77. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst 8, 329–337.e4 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Germain, P.-L., Lun, A., Meixide, C. G., Macnair, W. & Robinson, M. D. Doublet identification in single-cell sequencing data using scDblFinder. F1000Res. 10, 979 (2021).

    PubMed  Google Scholar 

  80. Bais, A. S. & Kostka, D. scds: computational annotation of doublets in single-cell RNA sequencing data. Bioinformatics 36, 1150–1158 (2019).

    PubMed Central  Google Scholar 

  81. Xi, N. M. & Li, J. J. Benchmarking computational doublet-detection methods for single-cell RNA sequencing data. Cell Syst. 12, 176–194.e6 (2021).

    CAS  PubMed  Google Scholar 

  82. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Osseward, P. J. et al. Conserved genetic signatures parcellate cardinal spinal neuron classes into local and projection subsets. Science 372, 385–393 (2021).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  84. Häring, M. et al. Neuronal atlas of the dorsal horn defines its architecture and links sensory input to transcriptional cell types. Nat. Neurosci. 21, 869–880 (2018).

    PubMed  Google Scholar 

  85. Alkaslasi, M. R. et al. Single nucleus RNA-sequencing defines unexpected diversity of cholinergic neuron types in the adult mouse spinal cord. Nat. Commun. 12, 2471 (2021).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  86. Blum, J. A. et al. Single-cell transcriptomic analysis of the adult mouse spinal cord reveals molecular diversity of autonomic and skeletal motor neurons. Nat. Neurosci. 24, 572–583 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Delile, J. et al. Single cell transcriptomics reveals spatial and temporal dynamics of gene expression in the developing mouse spinal cord. Development 146, dev173807 (2019).

  88. Hamel, R. et al. Time-resolved single-cell RNAseq profiling identifies a novel Fabp5-expressing subpopulation of inflammatory myeloid cells in chronic spinal cord injury. Preprint at bioRxiv https://doi.org/10.1101/2020.10.21.346635 (2020).

  89. Hayashi, M. et al. Graded arrays of spinal and supraspinal v2a interneuron subtypes underlie forelimb and hindlimb motor control. Neuron 97, 869–884.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. 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).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  91. Sathyamurthy, A. et al. Massively parallel single nucleus transcriptional profiling defines spinal cord neurons and their activity during behavior. Cell Rep. 22, 2216–2225 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Wahane, S. et al. Diversified transcriptional responses of myeloid and glial cells in spinal cord injury shaped by HDAC3 activity. Sci. Adv. 7, eabd8811 (2021).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  93. Baek, M., Menon, V., Jessell, T. M., Hantman, A. W. & Dasen, J. S. Molecular logic of spinocerebellar tract neuron diversity and connectivity. Cell Rep. 27, 2620–2635.e4 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 554, 475–480 (2018).

    ADS  CAS  PubMed  Google Scholar 

  95. Marques, S. et al. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352, 1326–1329 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  96. O′Shea, T. M. et al. Border-forming wound repair astrocytes. Preprint at bioRxiv, https://doi.org/10.1101/2023.08.25.554857 (2023).

  97. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).

  98. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

  99. Hou, W. & Ji, Z. Palo: spatially aware color palette optimization for single-cell and spatial data. Bioinformatics 38, 3654–3656 (2022).

  100. Koopmans, F. et al. SynGO: an evidence-based, expert-curated knowledge base for the synapse. Neuron 103, 217–234.e4 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Büttner, M., Ostner, J., Müller, C. L., Theis, F. J. & Schubert, B. scCODA is a Bayesian model for compositional single-cell data analysis. Nat. Commun. 12, 6876 (2021).

    ADS  PubMed  PubMed Central  Google Scholar 

  102. Phipson, B. et al. propeller: testing for differences in cell type proportions in single cell data. Bioinformatics 38, 4720–4726 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Simmons, S. Cell type composition analysis: comparison of statistical methods. Preprint at bioRxiv https://doi.org/10.1101/2022.02.04.479123 (2022).

  104. Zimmerman, K. D., Espeland, M. A. & Langefeld, C. D. A practical solution to pseudoreplication bias in single-cell studies. Nat. Commun. 12, 738 (2021).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  105. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  106. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  107. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    PubMed  PubMed Central  Google Scholar 

  108. Fornes, O. et al. JASPAR 2020: update of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 48, D87–D92 (2020).

    CAS  PubMed  Google Scholar 

  109. Cable, D. M. et al. Robust decomposition of cell type mixtures in spatial transcriptomics. Nat. Biotechnol. https://doi.org/10.1038/s41587-021-00830-w (2021).

  110. McKenzie, A. T., Katsyv, I., Song, W.-M., Wang, M. & Zhang, B. DGCA: a comprehensive R package for differential gene correlation analysis. BMC Syst. Biol. 10, 106 (2016).

    PubMed  PubMed Central  Google Scholar 

  111. Biancalani, T. et al. Deep learning and alignment of spatially resolved single-cell transcriptomes with Tangram. Nat. Methods 18, 1352–1362 (2021).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the Swiss National Science Foundation (grant nos. 310030_192558 to G.C., PZ00P3_185728 to M.A.A. and PZ00P3_208988 to J.W.S.); the ALARME Foundation (to M.A.A. and G.C.); the Dr Miriam and Sheldon G. Adelson Medical Foundation (to M.V.S.); Wings for Life (to M.V.S. and M.A.S.); the Wyss Center for Bio and Neuroengineering (to M.A.A. and G.C.); and a Human Frontiers in Science Program long-term fellowship (grant no. LT001278/2017-L to C.K.). We are grateful to J. Ravier and F. Merlos for the illustrations; the Advanced Lightsheet Imaging Center (ALICe) at the Wyss Center for Bio and Neuroengineering, Geneva and E. Bradbury for providing ChABC vectors. This work was supported in part using the resources and services of the Gene Expression Core Facility and the Bertarelli Platform for Gene Therapy at the School of Life Sciences of the EPFL. We acknowledge the foundational work by the many investigators whose work we could not cite due to space limits.

Author information

Authors and Affiliations

Authors

Contributions

J.W.S., J.B., M.V.S., M.A.A., M.A.S. and G.C. conceived and designed experiments. J.W.S., M.A.A., C.K., M.G., T.H.H., A.L., A.d.C., N.R., V.A. and N.D.J. conducted experiments. J.W.S., M.A.S., M.G., Q.B. and A.Y.Y.T. analysed the data. B.S. contributed essential resources. J.W.S., M.A.S., M.A.A. and G.C. wrote the manuscript. All authors contributed to the editing of the manuscript.

Corresponding authors

Correspondence to Mark A. Anderson, Jordan W. Squair or Grégoire Courtine.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Binhai Zheng 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 figures and tables

Extended Data Fig. 1 Clustering tree of 180 cell types and subtypes in the snRNA-seq atlas.

a, Clustering tree of the mouse spinal cord, revealing the hierarchical relationships between spinal cord cell types across levels 1 to 3 in the taxonomy, with cell types at level 3 highlighted. Text at the top of the tree shows the clades of the clustering tree corresponding to the major cell types of the mouse spinal cord (i.e., level 1 in the taxonomy). b, As in a, but showing level 4 in the taxonomy. c, As in a, but showing level 5 in the taxonomy.

Extended Data Fig. 2 Neuronal susceptibility and resilience to SCI.

a, Boxplot highlighting the proportion of neurons within individual libraries from the severity experiment, as compared to other major cell types. b, Boxplot showing the log2-odds ratio comparing the proportions of neurons from each level 4 subtype between the uninjured spinal cord, for all comparisons involving the injured spinal cord at 7 days post-injury. Cerebrospinal fluid-contacting neurons are the lone subpopulation to exhibit statistically significant resilience following SCI. **, p < 0.01; ***, p < 0.001. c, Scatterplot highlighting an individual comparison from b, showing the proportions of neurons from each level 4 subtype in the uninjured spinal cord, x-axis, and 7 days after a complete injury, y-axis. Color shows the log2-odds ratio. Cerebrospinal fluid-contacting neurons are highlighted. d, Proportion, y-axis, and absolute number, point size, of cerebrospinal fluid-contacting neurons recovered from each experimental condition. Dotted line shows the proportion of cerebrospinal fluid-contacting neurons in the uninjured spinal cord. e, Representative histological photomicrographs show injured spinal cords across injury severities after staining for NeuN and PKD1L2, a marker of cerebrospinal fluid-contacting neurons. f, Quantification of histological data demonstrating increasing proportions of cerebrospinal fluid-contacting neurons across injury severities. g, Volcano plot showing differentially expressed genes in cerebrospinal fluid-contacting neurons following spinal cord injury, as compared to other neuron subtypes.

Extended Data Fig. 3 Conserved and divergent neuronal responses to SCI.

a, Heatmap showing fold changes for all genes differentially expressed after SCI in at least one level 4 neuron subtype at 1 day, top, and 1 month, bottom, after injury. Patterns of differential expression are broadly conserved at 1 day, but more subtype-specific at 1 month. b, Heatmap showing fold changes for selected genes with broadly conserved patterns of differential expression across level 4 neuron subtypes at 1 day post-injury. c, Heatmap showing coefficients estimated by linear mixed models within each neuron subtype for up- or downregulation of the early-conserved neuronal module over the injury timecourse. d, Heatmap showing coefficients estimated by linear mixed models within each neuron subtype for selected GO term modules with broadly conserved patterns of up- or downregulation across level 4 neuron subtypes at 1 day post-injury. e, Dot plot showing median expression of the circuit reorganization module in each level 4 neuron subtype across timepoints. Point color and size both show median expression of the circuit reorganization module. f, Boxplots showing expression of the axon development, left, and dendrite development, right modules in Vsx2-expressing neurons across timepoints. g, Boxplot showing expression of the circuit reorganization module in each level 4 neuron subtype within the uninjured spinal cord. Vsx2-expressing neurons display the highest expression of the circuit reorganization module in the uninjured spinal cord. h, Dendrogram showing expression of the growth factor Gdnf across levels 1 to 4 of the neuron taxonomy. Point color shows mean expression in each neuron subtype, while point size reflects the proportion of neurons of that subtype with detectable expression. i, Scatterplots comparing basal expression of the circuit reorganization module in the uninjured spinal cord, x-axis, with the SCI-induced upregulation of this module at each timepoint after injury, y-axis, for each level 4 neuron subtype. Inset text shows the Pearson correlation. Basal and induced expression of the circuit reorganization module is maximally correlated at 1 month post-injury, coinciding with the temporal window of opportunity for natural recovery after SCI. j, Timeline of Vsx2ON neuron diphtheria toxin ablation experiments. Two weeks before complete crush SCI, animals received an injection of AAVs expressing DTR. At eight weeks, animals received daily injections of diphtheria toxin for 7 days. Kinematics were then recorded and tissue was collected for evaluation. k, Histological verification of Vsx2ON neuron ablation in the lower thoracic region. Images show loss of Vsx2ON neurons in the thoracic spinal cord, above and below the level of the crush SCI. Bar graph shows the number of Vsx2ON neurons found in each animal (n = 4 mice per group, independent samples two-tailed t-test, t = 11.7, p = 2.4 × 10–5). l, Locomotor performance in the Vsx2ON ablation experiment, as quantified in Supplementary Fig. 3 (n > 10 gait cycles per mouse, n = 4 mice per group).

Source data

Extended Data Fig. 4 Neurons remain differentiated after SCI.

a, Volcano plots showing differential expression for all level 4 neuron subtypes simultaneously across timepoints. Enlarged points represent the key marker genes for each neuron subtype shown in Supplementary Fig. 11a. Marker genes shown in grey show no evidence of differential expression after SCI in their respective neuron subtype. b, Differential expression of key marker genes for level 4 neuron subtypes across timepoints and injury severities (log2-fold change, y-axis, and false discovery rate, point color). Timepoints or severities for which key marker genes show evidence of statistically significant differential expression, compared to uninjured neurons, are plotted with light grey backgrounds. Genes without statistical evidence of differential expression (i.e., FDR greater than 5%) are shown as white points. n.d., genes that were not detectably expressed and could not be subjected to DE analysis. c, Volcano plot showing differential expression for averaged gene expression modules comprising the top 5, top 10, or top 50 marker genes identified for each level 4 neuron subtype by unbiased comparisons with all other neurons, simultaneously across all timepoints. The vast majority of marker gene modules show no evidence of downregulation after SCI in their respective neuron subtype. d, As in c, but showing differential expression for averaged gene expression modules comprising the top 50 marker genes for each level 4 neuron subtype, shown separately for each condition in the timecourse experiment.

Extended Data Fig. 5 Failure of tripartite barrier formation in old mice.

a, Sankey diagram showing the proportions of immune cell subtypes in young and old mice at seven days post-injury. b, Volcano plot showing the statistical significance of changes in immune cell subtype proportions between young and old mice. c, Composite tiled scans and confocal insets of albumin and GFAP in horizontal sections from representative old and young mice at two weeks after SCI. d, Line graph demonstrates albumin intensity at specific distances rostral and caudal to lesion centers. Bottom right, bar graph indicates the area under the curve (independent samples two-tailed t-test, n = 5 per group, t = –3.47, p = 0.022). e, Dendrogram showing cell type prioritizations assigned by Augur across the cellular taxonomy of the spinal cord in comparisons of young and old mice at seven days post-injury. The eight level 5 cell types with the highest AUCs are annotated. f, Sankey diagram showing the proportions of vascular cell subtypes in young and old mice at seven days post-injury. g, Sankey diagram showing the proportions of astroependymal cells expressing Id3 in young and old mice at seven days post-injury (p = 1.9 × 10–3, χ2 test). h, Average expression of the BBB identity module, left, and the peripheral vascular identity module, right, in capillary endothelial cells from young and old mice at seven days post-injury. i, Heatmap showing log-fold changes for all genes differentially expressed in at least one level 4 cell type in comparisons of injured versus uninjured mice, top, and old versus young mice, bottom. j, Heterogeneity of differential expression in comparisons of injured versus uninjured mice, top, and old versus young mice, bottom. Each point shows a gene differentially expressed in at least one level 4 cell type. The x-axis shows the average log2-fold change across all cell types; the y-axis shows the standard deviation of the log2-fold change across cell types (“response heterogeneity”); point size reflects the total number of cell types in which the gene is differentially expressed; and point color reflects the proportion of cell types in which the sign of the log2-fold change was the same as the modal sign (“direction consistency”).

Extended Data Fig. 6 Shared and distinct programs of gene expression across lesion compartments.

a, Left, total number of differentially expressed genes detected within each lesion compartment at 7 days and 2 months after SCI. Right, legend showing the position of each lesion compartment, as in Fig. 6c. b-c, Volcano plots showing differentially expressed genes for all lesion compartments at 7 days, b, and 2 months, c. The top three genes per lesion compartment, as ranked by the product of the log2-fold change and the –log10 p-value, are annotated. d-e, Heatmap showing log2-fold changes for all genes differentially expressed in at least one lesion compartment at 7 days, d, and 2 months, e. f-g, Heatmap showing log2-fold changes for the top five genes differentially expressed in each lesion compartment at 7 days, f, and 2 months, g. h-i, Visualization of selected differentially expressed genes specific to individual lesion compartments at 7 days, h, and 2 months, i, on the two- dimensional coordinate system of the injured spinal cord.

Extended Data Fig. 7 Cell type deconvolution of the 2D spatial atlas.

a, Major cell types assigned to each spatial barcode, visualized for each experimental condition on the two-dimensional coordinate system of the injured spinal cord. b, Sankey diagram showing the cellular composition of each lesion compartment at 7 days, for major (level 1) cell types. c, Sankey diagram showing the cellular composition of each lesion compartment at 2 months, for level 1 cell types. d, Sankey diagram showing the cellular composition of each lesion compartment at 2 months, for level 2 cell types. e, Sankey diagram showing the evolution of the cellular composition of the entire injured spinal cord between 7 days and 2 months, for level 1 cell types. f, Sankey diagram showing the evolution of the cellular composition of the entire injured spinal cord between 7 days and 2 months, for level 2 cell types. g, Visualization of the deconvolution weights assigned by RCTD for selected level 2 cell types at each timepoint, on the two-dimensional coordinate system of the injured spinal cord.

Extended Data Fig. 8 Molecular basis of spatial prioritization at the gene level.

a-c, Volcano plots showing correlations between the AUCs assigned by Magellan at each spatial barcode and transcriptome-wide gene expression across the same spatial barcodes (a, 7 days versus uninjured; b, 2 months versus uninjured; c, 7 days versus 2 months). Inset pie charts show the proportions of all tested genes that are significantly correlated with the spatial prioritizations. d, Heatmap showing Pearson correlations between spatial prioritizations and gene expression for each pairwise comparison of experimental conditions, for the top 40 most recurrently correlated genes across all comparisons. e, Heatmap showing Pearson correlations between spatial prioritizations and gene expression for each pairwise comparison of experimental conditions, for the top 40 most variably correlated genes across all comparisons. f, Visualization of selected genes prioritized by their correlation to spatial prioritizations on the two-dimensional coordinate system of the injured spinal cord.

Extended Data Fig. 9 Inhibitory and facilitating molecules in the 3D spatial atlas.

Expression of selected inhibitory and facilitating molecules across the 3D spatial transcriptomic atlas at 7 days. a, Cspg4; b, Cspg5; c, Ncan; d, Acan; e, Lama1.

Extended Data Fig. 10 A rejuvenative gene therapy reestablishes the tripartite barrier to restore walking.

a, Left, experimental design of a gene therapy intervention to promote the formation of the tripartite barrier, reproduced from Fig. 8a. Right, a second chronophotography series showing walking in old mice without (top) and with (bottom) a gene therapy intervention to promote the formation of the tripartite barrier. b, Composite tiled scans of GFAP and CD45 in horizontal sections from representative old and treated mice. c, Horizontal sections from representative old and treated mice identifying a restoration of Sox9ONId3ON cells in the astrocyte border region in treated mice. d, Composite tiled scans and confocal insets of albumin and GFAP in horizontal sections from representative old and treated mice after SCI. e, Line graph demonstrates albumin intensity at specific distances rostral and caudal to lesion centers. Bottom right, bar graph indicates the area under the curve (independent samples two-tailed t-test, n = 5 per group, t = 4.07, p = 0.0099). f, Locomotor performance in the gene therapy experiment, as quantified in Supplementary Fig. 3 (n > 10 gait cycles per mouse, (n = 5 mice per group; Tukey’s honestly significant difference test). *, p < 0.05; **, p < 0.01; ***, p < 0.001. g, Left, schematic overview of the classification pipeline using high-resolution kinematics data from young and old mice. Right, experimental conditions assigned to individual steps in old mice that received gene therapy by a machine-learning model trained on kinematics data from untreated animals.

Source data

Supplementary information

Supplementary information

These files contain Supplementary Notes 1–6, Figs. 1–42 and References.

Reporting Summary

Supplementary Table 1

Raw kinematics data for all experiments presented in the paper.

Supplementary Table 2

List of genes used to calculate gene module scores reported in the paper.

Supplementary Video 1

The Tabulae Paralytica: multimodal single-cell and spatial atlases of SCI.

Supplementary Video 2

Ablating Vsx2ON neurons prevents the natural recovery of walking after SCI.

Supplementary Video 3

A rejuvenative gene therapy re-establishes the tripartite barrier to restore walking in old mice.

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

Skinnider, M.A., Gautier, M., Teo, A.Y.Y. et al. Single-cell and spatial atlases of spinal cord injury in the Tabulae Paralytica. Nature 631, 150–163 (2024). https://doi.org/10.1038/s41586-024-07504-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07504-y

This article is cited by

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