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:

Pro-regenerative biomaterials recruit immunoregulatory dendritic cells after traumatic injury

Abstract

During wound healing and surgical implantation, the body establishes a delicate balance between immune activation to fight off infection and clear debris and immune tolerance to control reactivity against self-tissue. Nonetheless, how such a balance is achieved is not well understood. Here we describe that pro-regenerative biomaterials for muscle injury treatment promote the proliferation of a BATF3-dependent CD103+XCR1+CD206+CD301b+ dendritic cell population associated with cross-presentation and self-tolerance. Upregulation of E-cadherin, the ligand for CD103, and XCL-1 in injured tissue suggests a mechanism for cell recruitment to trauma. Muscle injury recruited natural killer cells that produced Xcl1 when stimulated with fragmented extracellular matrix. Without cross-presenting cells, T-cell activation increases, pro-regenerative macrophage polarization decreases and there are alterations in myogenesis, adipogenesis, fibrosis and increased muscle calcification. These results, previously observed in cancer progression, suggest a fundamental mechanism of immune regulation in trauma and material implantation with implications for both short- and long-term injury recovery.

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: Pro-regenerative and pro-fibrotic materials recruit a diverse range of innate immune cells.
Fig. 2: BATF3-dependent cross-presenting dendritic cells are enriched by pro-regenerative scaffolds and peak by 7 days post-injury.
Fig. 3: Pro-regenerative materials induce a mixed type-2/regulatory lymphocyte phenotype that regulates scavenger receptor expression on macrophages and dendritic cells.
Fig. 4: Loss of cross-presenting cells results in over-activation of T cells and attenuation of pro-regenerative macrophage polarization.
Fig. 5: Upregulation of XCL-1 in NK cells and E-cadherin in muscle infiltrate is associated with tissue damage and material implantation.
Fig. 6: BATF3 regulates post-injury tissue development including myogenesis, adipogenesis, fibrosis and calcification.

Similar content being viewed by others

Data availability

Data generated or analysed during this study are provided in the Supplementary Information. Further data are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Marwick, C. Implant recommendations. JAMA 283, 869 (2000).

    CAS  Google Scholar 

  2. Wampler, A. How differently should the FDA regulate drugs and devices? AMA J. Ethics 23, 669–672 (2021).

    Google Scholar 

  3. Anderson, J. M. Biological responses to materials. Annu. Rev. Mater. Res. 31, 81–110 (2001).

    CAS  Google Scholar 

  4. Anderson, J. M., Rodriguez, A. & Chang, D. T. Foreign body reaction to biomaterials. Semin. Immunol. 20, 86–100 (2008).

    CAS  Google Scholar 

  5. Heredia, J. E. et al. Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell 153, 376–388 (2013).

    CAS  Google Scholar 

  6. Goh, Y. P. S. et al. Eosinophils secrete IL-4 to facilitate liver regeneration. Proc. Natl Acad. Sci. USA 110, 9914–9919 (2013).

    CAS  Google Scholar 

  7. Horsley, V., Jansen, K. M., Mills, S. T. & Pavlath, G. K. IL-4 acts as a myoblast recruitment factor during mammalian muscle growth. Cell 113, 483–494 (2003).

    CAS  Google Scholar 

  8. Schiaffino, S., Pereira, M. G., Ciciliot, S. & Rovere‐Querini, P. Regulatory T cells and skeletal muscle regeneration. FEBS J. 284, 517–524 (2017).

    CAS  Google Scholar 

  9. Panduro, M., Benoist, C. & Mathis, D. Treg cells limit IFN-γ production to control macrophage accrual and phenotype during skeletal muscle regeneration. Proc. Natl Acad. Sci. USA 115, E2585–E2593 (2018).

  10. Hanna, B. S., Yaghi, O. K., Langston, P. K. & Mathis, D. The potential for Treg-enhancing therapies in tissue, in particular skeletal muscle, regeneration. Clin. Exp. Immunol. 211, 138–148 (2022).

    Google Scholar 

  11. Kratofil, R. M. A monocyte–leptin–angiogenesis pathway critical for repair post-infection. Nature 609, 166–173 (2022).

  12. Witherel, C. E., Abebayehu, D., Barker, T. H. & Spiller, K. L. Macrophage and fibroblast interactions in biomaterial‐mediated fibrosis. Adv. Healthc. Mater. 8, 1801451 (2019).

    Google Scholar 

  13. Chung, L. et al. Interleukin 17 and senescent cells regulate the foreign body response to synthetic material implants in mice and humans. Sci. Transl. Med. 12, eaax3799 (2020).

    CAS  Google Scholar 

  14. Maddur, M. S., Miossec, P., Kaveri, S. V. & Bayry, J. Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am. J. Pathol. 181, 8–18 (2012).

    CAS  Google Scholar 

  15. Wolfram, D. et al. T regulatory cells and TH17 cells in peri–silicone implant capsular fibrosis. Plast. Reconstr. Surg. 129, 327e–337e (2012).

    CAS  Google Scholar 

  16. Babensee, J. E. Interaction of dendritic cells with biomaterials. Semin. Immunol. 20, 101–108 (2008).

    CAS  Google Scholar 

  17. Kou, P. M. & Babensee, J. E. Macrophage and dendritic cell phenotypic diversity in the context of biomaterials. J. Biomed. Mater. Res. A 96, 239–260 (2011).

    Google Scholar 

  18. Kumamoto, Y. et al. CD301b+ dermal dendritic cells drive T helper 2 cell-mediated immunity. Immunity 39, 733–743 (2013).

    CAS  Google Scholar 

  19. Gao, Y. et al. Control of T helper 2 responses by transcription factor IRF4-dependent dendritic cells. Immunity 39, 722–732 (2013).

    CAS  Google Scholar 

  20. Ohta, T. et al. Crucial roles of XCR1-expressing dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune homeostasis. Sci. Rep. 6, 23505 (2016).

    CAS  Google Scholar 

  21. Lei, Y. et al. Aire-dependent production of XCL1 mediates medullary accumulation of thymic dendritic cells and contributes to regulatory T cell development. J. Exp. Med. 208, 383–394 (2011).

    CAS  Google Scholar 

  22. Saligrama, N. et al. Opposing T cell responses in experimental autoimmune encephalomyelitis. Nature 572, 481–487 (2019).

    CAS  Google Scholar 

  23. Li, J. et al. KIR+CD8+ T cells suppress pathogenic T cells and are active in autoimmune diseases and COVID-19. Science 376, eabi9591 (2022).

    CAS  Google Scholar 

  24. Brown, B. N. et al. Macrophage phenotype as a predictor of constructive remodeling following the implantation of biologically derived surgical mesh materials. Acta Biomater. 8, 978–987 (2012).

    CAS  Google Scholar 

  25. Lin, H.-H. et al. The macrophage F4/80 receptor is required for the induction of antigen-specific efferent regulatory T cells in peripheral tolerance. J. Exp. Med. 201, 1615–1625 (2005).

    CAS  Google Scholar 

  26. Madsen, D. H. et al. M2-like macrophages are responsible for collagen degradation through a mannose receptor-mediated pathway. J. Cell Biol. 202, 951–966 (2013).

    CAS  Google Scholar 

  27. Dziki, J. et al. An acellular biologic scaffold treatment for volumetric muscle loss: results of a 13-patient cohort study. NPJ Regen. Med. 1, 16008 (2016).

    Google Scholar 

  28. Sicari, B. M. et al. An acellular biologic scaffold promotes skeletal muscle formation in mice and humans with volumetric muscle loss. Sci. Transl. Med. 6, 234ra258 (2014).

    Google Scholar 

  29. Han, N. et al. Electrodiagnostic evaluation of individuals implanted with extracellular matrix for the treatment of volumetric muscle injury: case series. Phys. Ther. 96, 540–549 (2016).

    Google Scholar 

  30. Mase, V. J. Jr et al. Clinical application of an acellular biologic scaffold for surgical repair of a large, traumatic quadriceps femoris muscle defect. Orthopedics 33, 511 (2010).

    Google Scholar 

  31. Sadtler, K. et al. Divergent immune responses to synthetic and biological scaffolds. Biomaterials 192, 405–415 (2019).

    CAS  Google Scholar 

  32. Wolf, M. T. et al. A biologic scaffold-associated type 2 immune microenvironment inhibits tumor formation and synergizes with checkpoint immunotherapy. Sci. Transl. Med. 11, eaat7973 (2019).

    CAS  Google Scholar 

  33. Sadtler, K. et al. Developing a pro-regenerative biomaterial scaffold microenvironment requires T helper 2 cells. Science 352, 366–370 (2016).

    CAS  Google Scholar 

  34. Bachem, A. et al. Expression of XCR1 characterizes the Batf3-dependent lineage of dendritic cells capable of antigen cross-presentation. Front. Immunol. 3, 214 (2012).

    Google Scholar 

  35. Sattler, S. et al. IL-10-producing regulatory B cells induced by IL-33 (BregIL-33) effectively attenuate mucosal inflammatory responses in the gut. J. Autoimmun. 50, 107–122 (2014).

    CAS  Google Scholar 

  36. Chong, W. P., Zhou, J., Law, H. K. W., Tu, W. & Lau, Y. L. Natural killer cells become tolerogenic after interaction with apoptotic cells. Eur. J. Immunol. 40, 1718–1727 (2010).

    CAS  Google Scholar 

  37. Lee, W., Kim, H. S., Hwang, S. S. & Lee, G. R. The transcription factor Batf3 inhibits the differentiation of regulatory T cells in the periphery. Exp. Mol. Med. 49, e393 (2017).

    CAS  Google Scholar 

  38. Van den Bossche, J. et al. Alternatively activated macrophages engage in homotypic and heterotypic interactions through IL-4 and polyamine-induced E-cadherin/catenin complexes. Blood 114, 4664–4674 (2009).

    Google Scholar 

  39. Van den Bossche, J. et al. E-cadherin expression in macrophages dampens their inflammatory responsiveness in vitro, but does not modulate M2-regulated pathologies in vivo. Sci. Rep. 5, 12599 (2015).

    Google Scholar 

  40. Shore, E. M. et al. A recurrent mutation in the BMP type I receptor ACVR1 causes inherited and sporadic fibrodysplasia ossificans progressiva. Nat. Genet. 38, 525–527 (2006).

    CAS  Google Scholar 

  41. Kajabadi, N. et al. Activation of β-catenin in mesenchymal progenitors leads to muscle mass loss. Dev. Cell 58, 489–505.e7 (2023).

    CAS  Google Scholar 

  42. Böttcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037.e14 (2018).

    Google Scholar 

  43. Hernandez, C., Huebener, P. & Schwabe, R. F. Damage-associated molecular patterns in cancer: a double-edged sword. Oncogene 35, 5931–5941 (2016).

    CAS  Google Scholar 

  44. Varela-Chinchilla, C. D., Salinas-McQuary, G., Segura-Azuara, N. D. L. Á. & Trinidad-Calderón, P. A. Breast implant illness: surgical, autoimmune, and breast reconstruction associations. Surgeries 3, 111–125 (2022).

    Google Scholar 

  45. Huang, H., Li, F., Gordon, J. R. & Xiang, J. Synergistic enhancement of antitumor immunity with adoptively transferred tumor-specific CD4+ and CD8+ T cells and intratumoral lymphotactin transgene expression. Cancer Res. 62, 2043–2051 (2002).

    CAS  Google Scholar 

  46. Stojanovic, B. et al. IL-33/ST2 axis mediates resistance to EAE by promoting regulatory B and tolerogenic dendritic cells. J. Neuroimmunol. 275, 11–12 (2014).

    Google Scholar 

  47. Hubert, F.-X. et al. Aire regulates the transfer of antigen from mTECs to dendritic cells for induction of thymic tolerance. Blood 118, 2462–2472 (2011).

    CAS  Google Scholar 

  48. Yamazaki, C. et al. Conservation of a chemokine system, XCR1 and its ligand, XCL1, between human and mice. Biochem. Biophys. Res. Commun. 397, 756–761 (2010).

    CAS  Google Scholar 

  49. Mombaerts, P. et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell 68, 869–877 (1992).

    CAS  Google Scholar 

  50. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    CAS  Google Scholar 

  51. Ngo, T. B. et al. Label-free cleared tissue microscopy and machine learning for 3D histopathology of biomaterial implants. J. Biomed. Mater. Res. A 111, 840–850 (2023).

    CAS  Google Scholar 

  52. Guo, M. et al. Rapid image deconvolution and multiview fusion for optical microscopy. Nat. Biotechnol. 38, 1337–1346 (2020).

    CAS  Google Scholar 

  53. Belkina, A. C. et al. Automated optimized parameters for T-distributed stochastic neighbor embedding improve visualization and analysis of large datasets. Nat. Commun. 10, 5415 (2019).

    Google Scholar 

  54. McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at arXiv https://doi.org/10.48550/arXiv.1802.03426 (2018).

  55. Van Gassen, S. et al. FlowSOM: using self‐organizing maps for visualization and interpretation of cytometry data. Cytometry A 87, 636–645 (2015).

    Google Scholar 

Download references

Acknowledgements

We would like to thank R. Germain, Y. Belkaid, M. Wolf, R. Tussiwand and B. Warner for helpful conversations, and V. Sundaresan for organizational assistance and review of the manuscript. We also acknowledge M. Bur and L. Portnoy for oversight of the animal study protocols. This work was funded by the intramural research program of the National Institute of Biomedical Imaging and Bioengineering, NIH. The contents of this publication are the sole responsibility of the authors and do not necessarily reflect the views, opinions or policies of the NIH and the Department of Health and Human Services (HHS). Mention of trade names, commercial products or organizations does not imply endorsement by the US Government.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: K.S. Methodology and investigation: R.L., T.B.N., S.D., D.F., K.M.A., M.B., A.J., A.L., M.F., M.K., E.M., P.F., H.D.V. and K.S. Data analysis: R.L., T.B.N., D.F., A.J., P.F., Y.S., J.L., H.D.V. and K.S. Writing (original draft): R.L. and K.S. Writing (review and editing): R.L., T.B.N., A.J., M.K. and K.S. Funding and supervision: K.S.

Corresponding author

Correspondence to Kaitlyn Sadtler.

Ethics declarations

Competing interests

R.L., T.B.N. and K.S. have filed a provisional US patent application (US63/367,994) related to the work described in this paper. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Materials thanks the anonymous reviewers 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 Material preparation and injury model.

(a) Protein profile of native versus decellularized ECM via SDS-PAGE and Coomassie blue stain. (b) Double stranded DNA (dsDNA) quantification in native, mechanically scraped, and decellularized tissue as a function of starting tissue weight (n = 2, native; n = 3, scraped and decell), ANOVA with Tukey’s posthoc correction for multiple comparisons. (ce) Hematoxylin and eosin (H&E) staining of native (c), mechanically scraped (d), and decellularized (e) tissue with inset of picrosirius red brightfield and polarized light of final product. (f) ECM powder pre-hydration (g) polyethylene powder. (h) Image of injury generated in quadriceps muscle group with (i) application of ECM scaffold after injury. (j) Sheet form ECM at 7 days post-injury (dpi) showing retention in injury space. (k) Cleared tissue light sheet microscopy of control injury space at 63 dpi showing permanent loss of volume at injury site. (l) Light sheet image of ECMtx muscle injury at 7 dpi. Representative of n = 3 mice. (m) H&E of injury-material interface at 7-, 21-, and 42-days post-injury. Scale bar = 500 μm. (n) Muscle weight (n = 3, control; n = 4 ECMtx) MannWhitney test (two-tailed). (o) grip strength (n = 3) at 63 dpi. Mann-Whitney test (two-tailed). (p) Gene expression (n = 4, ECMtx; n = 5 PEtx) of Gli1 (p = 0.012) and Col1a1 (p = 0.013). ANOVA with Šídák’s multiple comparisons test. (q) Picrosirius red (PSR) staining of polyethylene particles at 7-, 21-, and 42-dpi at the capsule (skin facing), center of implant, and muscle interface. (r) Transverse histopathologic comparison (H&E) of an uninjured quadriceps (top) versus an injured muscle with ECM at 7 days post injury (bottom). (s) Example of PSR image processing for quantification. (t) Brightfield quantification of PSR staining. ANOVA with Tukey’s posthoc correction for multiple comparisons (u) Red (dense collagen) channel staining index in polarized light. ANOVA with Tukey’s posthoc correction for multiple comparisons (v) Green (less dense collagen) channel staining index in polarized light (h – j, n = 3 mice averaged from n = 5 fields of view per mouse). Data are means ± SEM. * = P < 0.05, ** = P < 0.01, *** = P < 0.001.

Source data

Extended Data Fig. 2 Immune cell populations over time.

Immune cell infiltration as a proportion of live CD45+ immune cells. (a) 3 (b) 7 (c) 21 (d) 42 days post-injury. N = 5 mice. (e) Major myeloid populations at 7 days in ECM particulate/powder (white) versus intact sheets (blue), n = 5 mice. (f) Hematoxylin and Eosin (H&E) of muscle-implant interface and ECM-capsule interface. (g) Major myeloid populations at 7 days in PE particulate (white, n = 5 mice) versus large (>2 mm, blue, n = 2 mice) beads (h) Picrosirius red (left) and H&E (right) of large 2 mm beads. Box plots are median with interquartile range, whiskers are minimum to maximum. ANOVA with Tukey post-hoc correction for multiple comparisons (a – d) or Šídák’s multiple comparisons test (e, g). * = P < 0.05, ** = P < 0.01, *** = P < 0.001, **** = P < 0.0001.

Source data

Extended Data Fig. 3 Phenotyping markers of macrophages and dendritic cells.

(a) MHCII+ antigen presenting cells in the wound microenvironment at 7 days post injury as a proportion of live CD45 + MHCII+ cells, n = 5 (b) MHCII+ macrophages (F4/80 + CD68 + MHCII+) (c) MHCII- Macrophages (F4/80 + CD68 + MHCII-) (d) DCs (CD11c+MHCII+). Black/grey = control injury; teal = ECMtx injury; pink = PEtx injury. Bars are mean for b – d. (e) CD103 mean fluorescence intensity, and (f) XCR1 mean fluorescence intensity on dendritic cells (black), MHCII+ macrophages (blue), and MHCII- macrophages (yellow). (af) n = 4 mice, control; n = 5 uninjured, ECMtx, PEtx. (g) Hierarchical clustering of FlowSOM-derived cell populations, Normalized min = 0, max = 100. Box plots are median with interquartile range, whiskers are minimum to maximum. ANOVA with Tukey post-hoc correction for multiple comparisons. * = P < 0.05, ** = P < 0.01, *** = P < 0.001, **** = P < 0.0001.

Source data

Extended Data Fig. 4 Dendritic cell profiling in injury space.

(a) Dendritic cell phenotyping by CD103 and XCR1 expression in CD11c+MHCIIhi DCs at 3, 7-, 21, and 42-days post-injury. (n = 5 mice, ANOVA with Tukey’s post-hoc correction for multiple comparisons) (b) Xcr1 gene expression by RT-PCR in muscle homogenate and single cells isolated from injured muscle. (n = 5 mice, Kruskall-Wallis with FDR correction for multiple comparisons). (c) Representative FACS plots of CD11c+MHCII+ dendritic cells in three treatment groups. Green = XCR1 + CD103+ tDCs; purple = XCR1-CD103 dendritic cells. (d) tDC quantification with particulate and larger geometries (n = 2 mice PE large geometry; n = 5 mice ECMtx and PE particulate, unpaired t-test, two-tailed, not significant). (e) Xcr1 gene expression in WT (black) and Batf3−/− mice (red), (n = 5 mice). Data are means ± SEM, ANOVA with Šídák’s multiple comparisons test. Box plots are median with interquartile range, whiskers are minimum to maximum. * = P < 0.05, ** = P < 0.01, *** = P < 0.001, **** = P < 0.0001. Panel b created with BioRender.com.

Source data

Extended Data Fig. 5 Dendritic cell phenotyping in local tissue, draining lymph node, and peripheral blood.

(ab) Repeatability of findings across litters and species. XCR1 + CD103+ cells are present in multiple runs with mice from (a) different litters (n = 5 mice per litter, ANOVA with Šídák’s multiple comparisons test) and (b) species (rat data extracted from raw data used generated in PMID 35462366). (c) CD103 + XCR1+ Dendritic cells in the skin overlying a muscle injury at 7 days post-injury in an ECM-treated mouse, as a proportion of CD11c+MHCII+ dendritic cells. (n = 3 skin; and n = 5 muscle, mean ± SEM, unpaired t-test, two-tailed, p = 0.0003) (d) DC expression of CD8α, CD103, and XCR1 in injured muscle tissue (purple), peripheral blood (green), and draining (inguinal) lymph node (blue), n = 5 mice, representative of at least 2 independent experiments. Box plots are median with interquartile range, whiskers are minimum to maximum. * = P < 0.05; ** = P < 0.01, *** = P < 0.001.

Source data

Extended Data Fig. 6 Adaptive immune responses in wild type and Batf3−/− mice.

(a) UMAP of lymphoid populations in muscle at 7 days post-injury. (b) tSNE of lymphoid populations in muscle at 7 days post-injury. (c) Treg populations in peripheral blood at 21 days post-injury, n = 5 mice, ANOVA with Tukey post-hoc correction for multiple comparisons. (d) Treg populations in draining (inguinal) lymph node at 21 days post-injury, n = 5 mice, ANOVA with Tukey post-hoc correction for multiple comparisons. (e) ST2+ regulatory B cells as a proportion of total B cells in the draining lymph node at 7 days post-injury, n = 5 mice (Control, ECM, PE), n = 3 mice (uninjured). WT (black), Batf3-/- (red), Uninjured WT (grey), ANOVA with Šídák’s multiple comparisons test (f) Activation phenotype of T cells in uninjured muscle versus 7 days post-injury. (g) Cytokine/Chemokine profile, log2 fold change over uninjured control, n = 5 mice. (h) Representative blots quantified in g. (i) RT-PCR of immune genes in muscle injury at 7 days post-injury. (n = 10 mice Il10, Il4, IlTgfb1, Xcl1; n = 5 mice all other genes, ANOVA with Šídák’s multiple comparisons test) (j) Count of Tregs and iTregs in the muscle of WT v Batf3−/− mice at 7 days post-injury, WT (black) Batf3-/- (red) n = 4 mice (PEtx WT) n = 5 mice all others, ANOVA with Šídák’s multiple comparisons test. Data are means ± SEM (panels e, j). Box plots are median with interquartile range, whiskers = minimum to maximum. * = P < 0.05, ** = P < 0.01. *** = P < 0.001. **** = P < 0.0001.

Source data

Extended Data Fig. 7 Systemic immune responses in wild type and Batf3−/− mice.

(a) UMAP (top row) of lymphoid populations in draining lymph node (ILN) at 7 days post-injury. tSNE (bottom row) of lymphoid populations in ILN at 7 days post-injury. (bd) Proportion of (b) B cells (c) αβ T cells and (d) γδ T cells in ILN in WT v Batf3−/− mice, ANOVA with Šídák’s multiple comparisons test. (e) Proportion of B cells positive for XCR1 and CD103. ANOVA with Šídák’s multiple comparisons test (f) Proportion of γδ T cells positive for XCR1 and CD103, ANOVA with Šídák’s multiple comparisons test. (g) Activation of CD103 + XCR1+ non-myeloid cells over time. (h) Proportion of XCR1 and CD103 positive non-myeloid cells over time. (i) Xcr1 gene expression in CD103 + XCR1 + T and B cells compared to CD103-XCR1 T and B cells; n = 5 mice, Paired t-test, two-tailed. (j) Proportion of T cells that are CD103 + XCR1+ in lymph node (ILN), peripheral blood, and muscle. Control = black, ECMtx = teal, PEtx = pink. Data are means ± SEM, n = 5 mice. Tukey with ANOVA post-hoc (k) Example flow cytometry plot showing CD103 + XCR1 + T cells (blue) versus other T cells (grey). (ln) Expression of CD103 and XCR1 on (l) CD4+ FoxP3+ (m) CD4 + HELIOS+, and (n) CD8 + HELIOS+ Tregs. Control = black, ECMtx = teal, PEtx = pink. Data are means ± SEM. n = 5 mice (t = 7, 21 days), n = 3 mice (t = 0). (o) Activation of gamma delta T cells in the inguinal lymph node of WT and Batf3/- mice. Dark blue = active (CD62L-CD44+), Medium blue = central memory (CD62L-CD44-), Light blue = Naïve (CD62L + CD44-). Blood 21 days = active, n = 5. Grey band = range in uninjured mice, n = 3. Data are means ± SEM. ANOVA with Šídák’s multiple comparisons test, * = P < 0.05, ** = P < 0.01. *** = P < 0.001. **** = P < 0.0001. WT (black) Batf3−/− (red). Box plots are median with interquartile range, whiskers = minimum to maximum.

Source data

Extended Data Fig. 8 Gene expression in WT and Batf3-/- mice.

(a) Cdh1 (encoding E-Cadherin) expression in Control, ECM, and PE. Data are mean ± SEM, n = 5 mice ANOVA with Šídák’s multiple comparisons test (b) Col1a1 (encoding Collagen I) expression with all samples from all treatment groups pooled (n = 15 mice), unpaired t-test, two-tailed.

Source data

Extended Data Fig. 9 Gating strategy for myeloid panel.

(a) Representative plots and gates from sample stained with 22 color myeloid phenotyping panel. Example data are from 7 days post-injury. (b) Fluorescence Minus One (FMO) Controls for select myeloid markers. FMO’s from select myeloid panel markers at 7 days post-injury in macrophage (top row) and dendritic cell (bottom row) populations. Grey = unstained control, black dashed line = FMO, red = full stained control.

Source data

Extended Data Fig. 10 Lymphoid panel gating strategy.

Displayed is a representative sample from the inguinal lymph node at 21 days post-injury.

Source data

Supplementary information

Supplementary Information

Legend for Supplementary Video 1 and Discussion.

Reporting Summary

Supplementary Video 1

Three-dimensional reconstruction of the light-sheet microscopy data.

Supplementary Table 1

Reagent list for antibodies and PCR primers.

Source data

Source Data Figs. 1–6

Source data for all the graphs and statistics for Figs. 1–6. Separate tabs are listed for each sheet with the statistics immediately following the raw data.

Source Data Extended Data Figs. 1–10

Source data for all the graphs and statistics for Extended Data Figs. 1–10. Separate tabs are listed for each sheet with the statistics immediately following the raw 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

Lokwani, R., Josyula, A., Ngo, T.B. et al. Pro-regenerative biomaterials recruit immunoregulatory dendritic cells after traumatic injury. Nat. Mater. 23, 147–157 (2024). https://doi.org/10.1038/s41563-023-01689-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-023-01689-9

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