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 transcriptomics of the infarcted heart define the dynamic onset of the border zone in response to mechanical destabilization

Abstract

The border zone (BZ) of the infarcted heart is a geographically complex and biologically enigmatic interface separating poorly perfused infarct zones (IZs) from remote zones (RZs). The cellular and molecular mechanisms of myocardial BZs are not well understood because microdissection inevitably combines them with uncontrolled amounts of RZs and IZs. Here, we use single-cell/nucleus RNA sequencing, spatial transcriptomics and multiplexed RNA fluorescence in situ hybridization to redefine the BZ based on cardiomyocyte transcriptomes. BZ1 (Nppa+Xirp2) forms a hundreds-of-micrometer-thick layer of morphologically intact cells adjacent to RZs that are detectable within an hour of injury. Meanwhile, BZ2 (Nppa+Xirp2+) forms a near-single-cell-thick layer of morphologically distorted cardiomyocytes at the IZ edge that colocalize with matricellular protein-expressing myofibroblasts and express predominantly mechanotransduction genes. Surprisingly, mechanical injury alone is sufficient to induce BZ genes. We propose a ‘loss of neighbor’ hypothesis to explain how ischemic cell death mechanically destabilizes the BZ to induce its transcriptional response.

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: CM transcripts redefine the ischemic BZ.
Fig. 2: BZ2 CMs form a thin boundary between surviving and ischemic myocardium.
Fig. 3: Cardiac immune niches after ischemic injury.
Fig. 4: Activated fibroblasts localize with BZ2 CMs.
Fig. 5: The transcriptional BZ emerges rapidly after ischemia.
Fig. 6: Morphological features of the emerging BZ.
Fig. 7: Ischemic injury is not necessary and mechanical trauma is sufficient to elicit BZ biology.

Similar content being viewed by others

Data availability

sc/snRNA-seq data and spatial transcriptomic sequencing data have been deposited to the Gene Expression Omnibus under accession no. GSE214611. All other data supporting the findings in this study are included in the main article and associated files.

Code availability

The code used to process this data is publicly available at Zenodo (https://zenodo.org/record/7055957#.Y00deezMIqu)

References

  1. Factor, S. M., Sonnenblick, E. H. & Kirk, E. S. The histologic border zone of acute myocardial infarction—islands or peninsulas? Am. J. Pathol. 92, 111–124 (1978).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Janse, M. J. et al. The “border zone” in myocardial ischemia. An electrophysiological, metabolic, and histochemical correlation in the pig heart. Circ. Res. 44, 576–588 (1979).

    Article  CAS  PubMed  Google Scholar 

  3. van Duijvenboden, K. et al. Conserved NPPB+ border zone switches from MEF2- to AP-1-driven gene program. Circulation 140, 864–879 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Vivien, C. J., Hudson, J. E. & Porrello, E. R. Evolution, comparative biology and ontogeny of vertebrate heart regeneration. NPJ Regen. Med. 1, 16012 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Smith, R. M., Black, A. J., Velamakanni, S. S., Akkin, T. & Tolkacheva, E. G. Visualizing the complex 3D geometry of the perfusion border zone in isolated rabbit heart. Appl. Opt. 51, 2713–2721 (2012).

    Article  PubMed  Google Scholar 

  6. Pirolo, J. S., Hutchins, G. M. & Moore, G. W. Infarct expansion: pathologic analysis of 204 patients with a single myocardial infarct. J. Am. Coll. Cardiol. 7, 349–354 (1986).

    Article  CAS  PubMed  Google Scholar 

  7. Erlebacher, J. A., Richter, R. C., Alonso, D. R., Devereux, R. B. & Gay, W. A. Jr. Early infarct expansion: structural or functional? J. Am. Coll. Cardiol. 6, 839–844 (1985).

    Article  CAS  PubMed  Google Scholar 

  8. Gao, X. M., Xu, Q., Kiriazis, H., Dart, A. M. & Du, X. J. Mouse model of post-infarct ventricular rupture: time course, strain- and gender-dependency, tensile strength, and histopathology. Cardiovasc Res. 65, 469–477 (2005).

    Article  CAS  PubMed  Google Scholar 

  9. Mounier-Vehier, F. et al. Borderzone infarct subtypes: preliminary study of the presumed mechanism. Eur. Neurol. 34, 11–15 (1994).

    Article  CAS  PubMed  Google Scholar 

  10. Wetstein, L., Michelson, E. L., Simson, M. B., Moore, E. N. & Harken, A. H. Increased normoxic-to-ischemic tissue borderzone as the cause for reentrant ventricular tachyarrhythmias. J. Surg. Res. 32, 526–534 (1982).

    Article  CAS  PubMed  Google Scholar 

  11. Hearse, D. J. & Yellon, D. M. The “Border zone” and myocardial protection: a time for reassessment? Acta Med. Scand. Suppl. 651, 37–46 (1981).

    CAS  PubMed  Google Scholar 

  12. Duncanson, E. R. & Mackey-Bojack, S. M. Histologic examination of the heart in the forensic autopsy. Acad. Forensic Pathol. 8, 565–615 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Jackson, B. M. et al. Extension of borderzone myocardium in postinfarction dilated cardiomyopathy. J. Am. Coll. Cardiol. 40, 1160–1167 (2002).

    Article  PubMed  Google Scholar 

  14. Yang, Y. et al. Age-related differences in postinfarct left ventricular rupture and remodeling. Am. J. Physiol. Heart Circ. Physiol. 294, H1815–H1822 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Cavasin, M. A., Tao, Z., Menon, S. & Yang, X.-P. Gender differences in cardiac function during early remodeling after acute myocardial infarction in mice. Life Sci. 75, 2181–2192 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Tsao, C. W. et al. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 145, e153–e639 (2022).

    Article  PubMed  Google Scholar 

  17. von Lueder, T. G., Kotecha, D., Atar, D. & Hopper, I. Neurohormonal blockade in heart failure. Card. Fail. Rev. 3, 19–24 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Aizarani, N. et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572, 199–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Halpern, K. B. et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature 542, 352–356 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Calcagno, D. M. et al. The myeloid type I interferon response to myocardial infarction begins in bone marrow and is regulated by Nrf2-activated macrophages. Sci. Immunol. 5, eaaz1974 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Calcagno, D. M. et al. SiglecF(HI) marks late-stage neutrophils of the infarcted heart: a single-cell transcriptomic analysis of neutrophil diversification. J. Am. Heart Assoc. 10, e019019 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dick, S. A. et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat. Immunol. 20, 29–39 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. King, K. R. et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat. Med. 23, 1481–1487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Forte, E. et al. Dynamic interstitial cell response during myocardial infarction predicts resilience to rupture in genetically diverse mice. Cell Rep. 30, 3149–3163.e6 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hu, P. et al. Single-nucleus transcriptomic survey of cell diversity and functional maturation in postnatal mammalian hearts. Genes Dev. 32, 1344–1357 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Martini, E. et al. Single-cell sequencing of mouse heart immune infiltrate in pressure overload-driven heart failure reveals extent of immune activation. Circulation 140, 2089–2107 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Ren, Z. et al. Single-cell reconstruction of progression trajectory reveals intervention principles in pathological cardiac hypertrophy. Circulation 141, 1704–1719 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Tucker, N. R. et al. Transcriptional and cellular diversity of the human heart. Circulation 142, 466–482 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wang, Z. et al. Cell-type-specific gene regulatory networks underlying murine neonatal heart regeneration at single-cell resolution. Cell Rep. 33, 108472 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Cui, M. et al. Dynamic transcriptional responses to injury of regenerative and non-regenerative cardiomyocytes revealed by single-nucleus RNA sequencing. Dev. Cell 53, 102–116.e8 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sinn, H. W., Balsamo, J., Lilien, J. & Lin, J. J. Localization of the novel Xin protein to the adherens junction complex in cardiac and skeletal muscle during development. Dev. Dyn. 225, 1–13 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Gustafson-Wagner, E. A. et al. Loss of mXinalpha, an intercalated disk protein, results in cardiac hypertrophy and cardiomyopathy with conduction defects. Am. J. Physiol. Heart Circ. Physiol. 293, H2680–H2692 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Long, P. A., Larsen, B. T., Evans, J. M. & Olson, T. M. Exome sequencing identifies pathogenic and modifier mutations in a child with sporadic dilated cardiomyopathy. J. Am. Heart Assoc. 4, e002443 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Huang, Y.-S., Xing, Y.-L. & Li, H.-W. Heterozygous desmin gene (DES) mutation contributes to familial dilated cardiomyopathy. J. Int. Med. Res. 49, 3000605211006598 (2021).

    Article  PubMed  Google Scholar 

  37. McCalmon, S. A. et al. Modulation of angiotensin II-mediated cardiac remodeling by the MEF2A target gene Xirp2. Circ. Res. 106, 952–960 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Verdonschot, J. A. J. et al. A mutation update for the FLNC gene in myopathies and cardiomyopathies. Hum. Mutat. 41, 1091–1111 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ortiz-Genga, M. F. et al. Truncating FLNC mutations are associated with high-risk dilated and arrhythmogenic cardiomyopathies. J. Am. Coll. Cardiol. 68, 2440–2451 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Hall, C. L. et al. Filamin C variants are associated with a distinctive clinical and immunohistochemical arrhythmogenic cardiomyopathy phenotype. Int. J. Cardiol. 307, 101–108 (2020).

    Article  PubMed  Google Scholar 

  41. Oz, S. et al. Reduction in filamin C transcript is associated with arrhythmogenic cardiomyopathy in Ashkenazi Jews. Int. J. Cardiol. 317, 133–138 (2020).

    Article  PubMed  Google Scholar 

  42. Begay, R. L. et al. Filamin C truncation mutations are associated with arrhythmogenic dilated cardiomyopathy and changes in the cell–cell adhesion structures. JACC Clin. Electrophysiol. 4, 504–514 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Valdés-Mas, R. et al. Mutations in filamin C cause a new form of familial hypertrophic cardiomyopathy. Nat. Commun. 5, 5326 (2014).

    Article  PubMed  Google Scholar 

  44. Han, P. et al. A long noncoding RNA protects the heart from pathological hypertrophy. Nature 514, 102–106 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Günthel, M. Epigenetic state changes underlie metabolic switch in mouse post-infarction border zone cardiomyocytes. J. Cardiovasc. Dev. Dis. 8, 134 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Guo, H. et al. Intercalated disc protein Xinβ is required for Hippo-YAP signaling in the heart. Nat. Commun. 11, 4666 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tao, G. et al. Pitx2 promotes heart repair by activating the antioxidant response after cardiac injury. Nature 534, 119–123 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Nadadur, R. D. et al. Pitx2 modulates a Tbx5-dependent gene regulatory network to maintain atrial rhythm. Sci. Transl. Med. 8, 354ra115 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Man, J. C. K. et al. Genetic dissection of a super enhancer controlling the Nppa-Nppb cluster in the heart. Circ. Res. 128, 115–129 (2021).

    Article  CAS  PubMed  Google Scholar 

  50. Cao, D. J. et al. Cytosolic DNA sensing promotes macrophage transformation and governs myocardial ischemic injury. Circulation 137, 2613–2634 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Farbehi, N. et al. Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury. eLife 8, e43882 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Muhl, L. et al. Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination. Nat. Commun. 11, 3953 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dela Paz, N. G. & D’Amore, P. A. Arterial versus venous endothelial cells. Cell Tissue Res. 335, 5–16 (2009).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  55. Kalucka, J. et al. Single-cell transcriptome atlas of murine endothelial cells. Cell 180, 764–779.e20 (2020).

    Article  CAS  PubMed  Google Scholar 

  56. Fujimoto, N. et al. Single-cell mapping reveals new markers and functions of lymphatic endothelial cells in lymph nodes. PLoS Biol. 18, e3000704 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Gerarduzzi, C., Hartmann, U., Leask, A. & Drobetsky, E. The matrix revolution: matricellular proteins and restructuring of the cancer microenvironment. Cancer Res. 80, 2705–2717 (2020).

    Article  CAS  PubMed  Google Scholar 

  58. Nishioka, T. et al. Tenascin C may aggravate left ventricular remodeling and function after myocardial infarction in mice. Am. J. Physiol. Heart Circ. Physiol. 298, H1072–H1078 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. McCurdy, S. M. et al. SPARC mediates early extracellular matrix remodeling following myocardial infarction. Am. J. Physiol. Heart Circ. Physiol. 301, H497–H505 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Murry, C. E., Giachelli, C. M., Schwartz, S. M. & Vracko, R. Macrophages express osteopontin during repair of myocardial necrosis. Am. J. Pathol. 145, 1450–1462 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Schellings, M. W. M. et al. Absence of SPARC results in increased cardiac rupture and dysfunction after acute myocardial infarction. J. Exp. Med. 206, 113–123 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Oka, T. et al. Genetic manipulation of periostin expression reveals a role in cardiac hypertrophy and ventricular remodeling. Circ. Res. 101, 313–321 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Trueblood, N. A. et al. Exaggerated left ventricular dilation and reduced collagen deposition after myocardial infarction in mice lacking osteopontin. Circ. Res. 88, 1080–1087 (2001).

    Article  CAS  PubMed  Google Scholar 

  64. Richardson, W. J. & Holmes, J. W. Why is infarct expansion such an elusive therapeutic target? J. Cardiovasc. Transl. Res. 8, 421–430 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Luther, D. J., Thodeti, C. K. & Meszaros, J. G. Injury models to study cardiac remodeling in the mouse: myocardial infarction and ischemia–reperfusion. Methods Mol. Biol. 1037, 325–342 (2013).

    Article  CAS  PubMed  Google Scholar 

  66. Monroe, T. O. et al. YAP partially reprograms chromatin accessibility to directly induce adult cardiogenesis in vivo. Dev. Cell 48, 765–779.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Xiao, Y., Leach, J., Wang, J. & Martin, J. F. Hippo/Yap signaling in cardiac development and regeneration. Curr. Treat. Options Cardiovasc. Med. 18, 38 (2016).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

This publication includes data generated at the UC San Diego IGM Genomics Center utilizing an Illumina NovaSeq 6000 that was purchased with funding from a National Institutes of Health SIG grant (#S10 OD026929). We thank IGM and the Nikon Imaging Center at UCSD for technical assistance. The work was funded by National Institutes of Health (NIH) grants NIH UL1TR001442 (UCSD), AHA17IRG33410543 (K.R.K.), NIH R00HL129168 (K.R.K.), NIH DP2AR075321 (K.R.K.), NIH HL142251 (F.S.) and NIH T32HL105373 (D.M.C.), National Heart Lung and Blood Institute (NHLBI) grants NHLBI T32HL007444 (V.K.N.) and NHLBI HL162369 (F.S.) and Department of Defense grant no. W81XWH1810380 (F.S.).

Author information

Authors and Affiliations

Authors

Contributions

D.M.C., N.T., V.K.N. and K.R.K. designed and performed the experiments, analyzed the data and wrote the manuscript. J.M.M., A.T., R.S., J.L. and Y.L. performed the experiments. J.M.D. and E.A. provided the human tissue samples. K.L.C., K.Z. and F.S. provided guidance on experimental design. Z.F. performed all mouse surgeries and developed new methods including the NP models. K.R.K. conceived the project and provided funding. All authors reviewed the results and commented on the manuscript.

Corresponding author

Correspondence to K. R. King.

Ethics declarations

Competing interests

F.S. is a cofounder and has an equity interest in Papillon Therapeutics; he is a consultant and has equity interest and a research grant from LEXEO Therapeutics. The other authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Rafael Kramann 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 Integration of single cell and nuclei RNA-seq heart datasets during acute responses to MI.

(a) Overall experimental design and integration of sc/snRNA-seq data with whole transcriptome spatial data. Hearts were harvested at several time points following experimental MI and collected for snRNA-seq. The resulting data matrices were integrated with available scRNA-seq data. (b) Experimental timepoints post-MI that were examined in our study with a summary table of total numbers of cells, nuclei and spatial pixels analyzed to support the robustness of our claims across biological replicates. (c) Gating strategy to isolate nuclei using DAPI and FACS. (d) Mitochondrial QC metrics of samples and replicates for both single nuclei, single cell and integrated sn/sc data. (e) UMAP plots annotated by major cell types (left) and subsets (right) after removing nuclei and cells that have more than 5% mitochondrial counts. (f) UMAP plots displaying composition of single nuclei (left) and single cell (right) derived samples. (g) Subcluster composition as derived from UMAPs shown in (f). (h) UMAP plots split by timepoint and across biological replicates. (i) Average subcluster composition displayed in (h). (l) QC metric of samples and replicates for both single nuclei and single cell represented in counts per sample (nCounts) and features per sample (nGenes).

Extended Data Fig. 2 Integration and quantification of spatial transcriptomic datasets.

(a) UMAP plots split by sample. (b) Quantification of cluster classification as a percentage of total pixels captured by sample (data presented as mean values ± SEM; n = 2, sham; n = 1, 1 hr and 4hrs; n = 3, 72 hrs and 168hrs post-MI). (c) Quality control metrics, split by sample. (d) Spatial plot showing results of clustering across different time points after MI and replicates.

Extended Data Fig. 3 High resolution clustering of CMs at 24 hrs and 72 hrs post-MI.

(a) UMAP plots of subset and reclustered CMs as shown in Fig. 1b with higher resolution (top) and annotated based on spatial mapping (bottom). (b) Heatmap of resulting DEGs based on resolution. (f) Heatmap of DEGs based on spatial mapping. (c) Select gene ontology terms enriched in BZ2 CMs related to Ras/Rho signaling. (d) Average scaled expression of guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs) in CM subsets. (e) Spatial feature plots of representative Ras/Rho related genes showing distribution in BZ.

Extended Data Fig. 4 Mapping of snRNA-seq derived CM subsets to space.

(a) Strategy for mapping labels (IZ, RZ, BZ1 and BZ2) from CM nuclei to spatial clusters (see methods). (b) UMAP plot of integrated dataset composed of 34,116 pixels in 16 samples (summarized in Extended Data Fig. 1b, see Extended Data fig. 8). (c) DEGs based on high-resolution clustering (top) and post-classification regions (bottom). (d) Histogram of counts from representative samples (green, sham; red, 72 hrs post-MI). (e) Histogram of BZ1 scores. (f) Dot plot of BZ1 and BZ2 scores. (g,h) Feature plots (g) and violin plots (h) of CM, BZ1 and BZ2 scores. (i) Results of ROC analysis (AUC, area under the curve) and stepwise label-mapping (reference cluster indicated above). (j) Classification results shown in UMAP space. (k) AUC as a function of gene-set length.

Extended Data Fig. 5 Spatial transcriptomics of human STEMI tissue.

(a) H&E of cross section of a single human heart sample from a patient presenting with anterior wall STEMI. (b) Spatial transcriptomic clustering results based on DEG analysis and assessment of BZ marker genes shown in space in (c) and by violin plots in (d). (e) Heatmap of cluster defining DEGs. LQ, low quality.

Extended Data Fig. 6 Segmentation, classification and quantification of mFISH data.

(a) After mFISH imaging, slides were stained for WGA to label the CM membrane. (b) Areas with well-defined perimeters were converted to regions-of-interest (shown in white). (c) Expression of BZ1 markers (Nppa, Clu) and BZ2 markers (Xirp2, Flnc) across ROIs. Scatter plot was visually inspected to establish thresholds and classify ROIs as RZ, BZ1, or BZ2 (blue, purple, red). (d) Quantification of neighbor composition in 25-pixel radius (n = 625 cells, and 1008 cells examined over 1 sample for BZ1 and BZ2, respectively). (e) Quantification of minimum distance to the IZ border defined by Tnnt2 staining. (f) To quantify direct contact of CMs, the percentage of Tnnt2 + pixels were calculated in ROI perimeters with various degrees of thickness (n = 625 cells, and 1008 cells examined over 1 sample for BZ1 and BZ2, respectively). (g) Contiguous regions of respective CM subsets were measured. Images shown are representative of 2 separate experiments. Scale bars indicate 500 μm. Boxplots presented with mean; box: 25th-75th%; whiskers: 2.5th-97.5th %. **** P-value < .0001; Mann-Whitney Test, two-sided.

Extended Data Fig. 7 Colocalization of immune and fibroblast subsets across time.

(a) Spatial plots showing subclustering results (shown in 3c) across 72 hrs post-MI and 168 hrs post-MI replicates. (b) Colocalized immune subsets in 72 hrs post-MI replicate. (c) Spatially correlated and clustered gene-set scores. (d) Spatially correlated and clustered genes based on spearman rank correlation analysis for all regions (left) and infarct zone (right) of 72 hrs post-MI sample. (e) All gene-set scores projected to clusters. (f) Volcano plot comparing gene-set scores in BZ1 and BZ2 (Wilcoxon Ranked Sum Test, two-sided).

Extended Data Fig. 8 Spatial transcriptomics on mechanical injury models.

(a) H&E sections at low magnification (lower panel) and high magnification (top panel). Representative of at least 2 separate experiments. (b) Clustering results of spatial transcriptomic data based on integrated dataset as shown in Extended Data Fig. 3. (c) Hif1 scores applied to representative 72 hrs post-MI sample (left) and NP (right) with quantifications plotted per pixel in (d). In comparison to post-MI hearts, needle pass injuries have a significantly lower Hif1 score compared to the positive control (72 hr MI) samples and is unchanged from negative controls in sham samples. (e) Experimental design of fluorescent dye assay to assess regional perfusion. Wheat germ agglutinin (WGA) conjugated to AlexaFluor 488 was injected directly into LV 10 min or 72 hrs post NP injury and hearts were harvested 20 minutes after injection for imaging and quantification (data presented as mean values ± SEM n = 3 biologically independent samples). The representative image in the left panel designates the visualization of the needle pass area designated as the negative control and the reference site designated as neighbors, and the right panel represents a positive control area remote from the site of injury. Quantification of samples harvested 30 minutes and 72 hrs post-NP injury reveal that in both time courses, Neighbors and Positive controls are significantly increased as compared to the Negative Control (Needle pass) sites and are not different from each other (f) Representative H&E-stained sections of NP injuries highlighting area of injury in dash lines and directions of adjacent myocyte bundles in arrows used to quantify gene expression. Representative of 2 separate experiments, each of which had tissue sections aligned with the long axis of the needle insertion. (g) Anisotropy ratios in each sample with non-injured area (right) showing no significant anisotropy in the borderzones of needle pass injuries (n = 2 biologically independent samples). Data presented as mean values ± SEM. *P-value < 0.05, **P-value < 0.01, ***P-value < 0.001, ****P-value < 0.0001; One-way ANOVA with Tukey’s post-hoc analysis.

Supplementary information

Reporting Summary

Supplementary Table 1

Differentially expressed genes of major cell types: (1.1) cardiomyocytes; (1.2) immune cells; (1.3) and stromal cells. P values were derived from Seurat and FDR-adjusted for multiple hypothesis testing

Supplementary Table 2

Differentially expressed genes of cell type subsets: (2.1–2.7) monocyte and macrophage subsets; (2.8) dendritic cell subsets; (2.9 and 2.10) neutrophil subsets; (2.11–2.18) endothelial cell subsets; (2.19–2.25) fibroblast subsets; (2.26–2.30) cardiomyocyte subsets. P values were derived from Seurat and FDR-adjusted for multiple hypothesis testing

Supplementary Table 3

Differentially expressed genes derived from spatial transcriptomics after label mapping. P values were derived from Seurat and FDR-adjusted for multiple hypothesis testing

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

Calcagno, D.M., Taghdiri, N., Ninh, V.K. et al. Single-cell and spatial transcriptomics of the infarcted heart define the dynamic onset of the border zone in response to mechanical destabilization. Nat Cardiovasc Res 1, 1039–1055 (2022). https://doi.org/10.1038/s44161-022-00160-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-022-00160-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing