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.

  • Review Article
  • Published:

Host responses to implants revealed by intravital microscopy

Abstract

Biomaterials, biomedical devices and engineered cell grafts can be implanted to restitute tissue anatomy and function. Such implants can either integrate physiologically, with no or minimal scarring, or induce chronic inflammation and the foreign body response, which leads to graft failure. Intravital microscopy in small animal models can be applied to visualize the structure and integration of implanted natural and synthetic fibres, metals, cells within hydrogel carriers and engineered tissues, as well as the stepwise cellular and molecular tissue response. In this Review, we discuss how intravital microscopy can reveal regenerative and pathological responses to implants, including wound healing and graft integration, which depend on the time-controlled activation of macrophages and early neovascularization, and adverse reactions, such as the foreign body response and fibrosis. The combination of improved materials designs, detection of cell signalling using molecular reporters and targeted intervention will allow the development of strategies to improve graft integration and functionality.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Fluorescence and nonlinear microscopy to detect implant materials and molecular signalling in cells.
Fig. 2: Animal models for intravital microscopy of implant biology.
Fig. 3: Tissue-specific implant integration.
Fig. 4: Foreign body response.

Similar content being viewed by others

References

  1. O’Brien, F. J. Biomaterials & scaffolds for tissue engineering. Mater. Today 14, 88–95 (2011).

    Google Scholar 

  2. Bose, S., Roy, M. & Bandyopadhyay, A. Recent advances in bone tissue engineering scaffolds. Trends Biotechnol. 30, 546–554 (2012).

    CAS  Google Scholar 

  3. Eming, S. A., Martin, P. & Tomic-Canic, M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci. Transl Med. 6, 265sr266 (2014).

    Google Scholar 

  4. Veiseh, O. & Vegas, A. J. Domesticating the foreign body response: Recent advances and applications. Adv. Drug Deliv. Rev. 144, 148–161 (2019).

    CAS  Google Scholar 

  5. Prakasam, M. et al. Biodegradable materials and metallic implants — a review. J. Funct. Biomater. 8, 44 (2017).

    Google Scholar 

  6. Veiseh, O. et al. Size- and shape-dependent foreign body immune response to materials implanted in rodents and non-human primates. Nat. Mater. 14, 643–651 (2015).

    CAS  Google Scholar 

  7. Zhang, L. et al. Zwitterionic hydrogels implanted in mice resist the foreign-body reaction. Nat. Biotechnol. 31, 553–556 (2013).

    CAS  Google Scholar 

  8. Vegas, A. J. et al. Combinatorial hydrogel library enables identification of materials that mitigate the foreign body response in primates. Nat. Biotechnol. 34, 345–352 (2016).

    CAS  Google Scholar 

  9. Zhang, D. et al. Silk-Inspired β-peptide materials resist fouling and the foreign-body response. Angew. Chem. Int. Ed. 59, 9586–9593 (2020).

    CAS  Google Scholar 

  10. Zhang, D. et al. Dealing with the foreign-body response to implanted biomaterials: strategies and applications of new materials. Adv. Funct. Mater. 31, 2007226 (2020).

    Google Scholar 

  11. Ouanounou, A., Hassanpour, S. & Glogauer, M. The influence of systemic medications on osseointegration of dental implants. J. Can. Dent. Assoc. 82, g7 (2016).

    Google Scholar 

  12. Dondossola, E. et al. Examination of the foreign body response to biomaterials by nonlinear intravital microscopy. Nat. Biomed. Eng. 1, 0007 (2017).

    CAS  Google Scholar 

  13. Gurevich, D. B., French, K. E., Collin, J. D., Cross, S. J. & Martin, P. Live imaging the foreign body response in zebrafish reveals how dampening inflammation reduces fibrosis. J. Cell Sci. 133, jcs236075 (2020).

    CAS  Google Scholar 

  14. Appel, A. A., Anastasio, M. A., Larson, J. C. & Brey, E. M. Imaging challenges in biomaterials and tissue engineering. Biomaterials 34, 6615–6630 (2013).

    CAS  Google Scholar 

  15. Andresen, V. et al. Infrared multiphoton microscopy: subcellular-resolved deep tissue imaging. Curr. Opin. Biotechnol. 20, 54–62 (2009).

    CAS  Google Scholar 

  16. Ebrahim, S. & Weigert, R. Intravital microscopy in mammalian multicellular organisms. Curr. Opin. Cell Biol. 59, 97–103 (2019).

    CAS  Google Scholar 

  17. Li, R., Ng, T. S. C., Garlin, M. A., Weissleder, R. & Miller, M. A. Understanding the in vivo fate of advanced materials by imaging. Adv. Funct. Mater. 30, 1910369 (2020).

    CAS  Google Scholar 

  18. Pittet, M. J. & Weissleder, R. Intravital imaging. Cell 147, 983–991 (2011).

    CAS  Google Scholar 

  19. Choi, M., Kwok, S. J. J. & Yun, S. H. In vivo fluorescence microscopy: lessons from observing cell behavior in their native environment. Physiology 30, 40–49 (2015).

    CAS  Google Scholar 

  20. Zipfel, W. R. et al. Live tissue intrinsic emission microscopy using multiphoton-excited native fluorescence and second harmonic generation. Proc. Natl Acad. Sci. USA 100, 7075–7080 (2003).

    CAS  Google Scholar 

  21. Monici, M. Cell and tissue autofluorescence research and diagnostic applications. Biotechnol. Annu. Rev. 11, 227–256 (2005).

    CAS  Google Scholar 

  22. Asfour, H., Otridge, J., Thomasian, R., Larson, C. & Sarvazyan, N. Autofluorescence properties of balloon polymers used in medical applications. J. Biomed. Opt. 25, 106004 (2020).

    Google Scholar 

  23. Okabe, M., Ikawa, M., Kominami, K., Nakanishi, T. & Nishimune, Y. ‘Green mice’ as a source of ubiquitous green cells. FEBS Lett. 407, 313–319 (1997).

    CAS  Google Scholar 

  24. Rizzo, M. A., Davidson, M. W. & Piston, D. W. Fluorescent protein tracking and detection: applications using fluorescent proteins in living cells. Cold Spring Harb. Protoc. 2009, pdb top64 (2009).

    Google Scholar 

  25. Zhang, G., Fiore, G. L., St. Clair, T. L. & Fraser, C. L. Difluoroboron dibenzoylmethane PCL-PLA block copolymers: Matrix effects on room temperature phosphorescence. Macromolecules 42, 3162–3169 (2009).

    CAS  Google Scholar 

  26. Karman, M., Verde-Sesto, E., Weder, C. & Simon, Y. C. Mechanochemical fluorescence switching in polymers containing dithiomaleimide moieties. ACS Macro Lett. 7, 1099–1104 (2018).

    CAS  Google Scholar 

  27. Li, W. et al. α-Lipoic acid stabilized DTX/IR780 micelles for photoacoustic/fluorescence imaging guided photothermal therapy/chemotherapy of breast cancer. Biomater. Sci. 6, 1201–1216 (2018).

    CAS  Google Scholar 

  28. Adam, V. Phototransformable fluorescent proteins: which one for which application? Histochem. Cell Biol. 142, 19–41 (2014).

    CAS  Google Scholar 

  29. Miller, M. A. & Weissleder, R. Imaging the pharmacology of nanomaterials by intravital microscopy: Toward understanding their biological behavior. Adv. Drug Deliv. Rev. 113, 61–86 (2017).

    CAS  Google Scholar 

  30. Zielinski, M., Oron, D., Chauvat, D. & Zyss, J. Second-harmonic generation from a single core/shell quantum dot. Small 5, 2835–2840 (2009).

    CAS  Google Scholar 

  31. Karvounis, A., Timpu, F., Vogler-Neuling, V. V., Savo, R. & Grange, R. Barium titanate nanostructures and thin films for photonics. Adv. Opt. Mater. 8, 2001249 (2020).

    CAS  Google Scholar 

  32. Aptel, F. et al. Multimodal nonlinear imaging of the human cornea. Invest. Ophthalmol. Vis. Sci. 51, 2459–2465 (2010).

    Google Scholar 

  33. Debarre, D. et al. Imaging lipid bodies in cells and tissues using third-harmonic generation microscopy. Nat. Methods 3, 47–53 (2006).

    CAS  Google Scholar 

  34. Weigelin, B., Bakker, G. J. & Friedl, P. Third harmonic generation microscopy of cells and tissue organization. J. Cell Sci. 129, 245–255 (2016).

    CAS  Google Scholar 

  35. Bakker, G. J., Andresen, V., Hoffman, R. M. & Friedl, P. Fluorescence lifetime microscopy of tumor cell invasion, drug delivery, and cytotoxicity. Methods Enzymol. 504, 109–125 (2012).

    CAS  Google Scholar 

  36. Nobis, M. et al. A RhoA-FRET biosensor mouse for intravital imaging in normal tissue homeostasis and disease contexts. Cell Rep. 21, 274–288 (2017).

    CAS  Google Scholar 

  37. Warren, S. C. et al. Removing physiological motion from intravital and clinical functional imaging data. eLife 7, e35800 (2018).

    Google Scholar 

  38. Conway, J. R. W. et al. Intravital imaging to monitor therapeutic response in moving hypoxic regions resistant to PI3K pathway targeting in pancreatic cancer. Cell Rep. 23, 3312–3326 (2018).

    CAS  Google Scholar 

  39. Lee, M. et al. In vivo imaging of the tumor and its associated microenvironment using combined CARS/2-photon microscopy. Intravital 4, e1055430 (2015).

    Google Scholar 

  40. Cao, X., Masatani, P., Torraca, G. & Wen, Z. Q. Identification of a mixed microparticle by combined microspectroscopic techniques: a real forensic case study in the biopharmaceutical industry. Appl. Spectrosc. 64, 895–900 (2010).

    CAS  Google Scholar 

  41. Zhou, H., Simmons, C. S., Sarntinoranont, M. & Subhash, G. Raman spectroscopy methods to characterize the mechanical response of soft biomaterials. Biomacromolecules 21, 3485–3497 (2020).

    Google Scholar 

  42. Morris, M. D. & Mandair, G. S. Raman assessment of bone quality. Clin. Orthop. Relat. Res. 469, 2160–2169 (2011).

    Google Scholar 

  43. Lach, S. et al. Spectroscopic methods used in implant material studies. Molecules 25, 579 (2020).

    CAS  Google Scholar 

  44. Filho, I. P. T. et al. Hemoglobin oxygen saturation measurements using resonance Raman intravital microscopy. Am. J. Physiol. Heart Circ. Physiol. 289, H488–H495 (2005).

    Google Scholar 

  45. Taruttis, A. & Ntziachristos, V. Advances in real-time multispectral optoacoustic imaging and its applications. Nat. Photonics 9, 219–227 (2015).

    CAS  Google Scholar 

  46. Liu, W. & Yao, J. Photoacoustic microscopy: principles and biomedical applications. Biomed. Eng. Lett. 8, 203–213 (2018).

    Google Scholar 

  47. Omar, M., Schwarz, M., Soliman, D., Symvoulidis, P. & Ntziachristos, V. Pushing the optical imaging limits of cancer with multi-frequency-band raster-scan optoacoustic mesoscopy (RSOM). Neoplasia 17, 208–214 (2015).

    Google Scholar 

  48. Lee, D., Park, S., Noh, W.-C., Im, J.-S. & Kim, C. Photoacoustic imaging of dental implants in a porcine jawbone ex vivo. Opt. Lett. 42, 1760–1763 (2017).

    CAS  Google Scholar 

  49. SoRelle, E. D. et al. Spatiotemporal tracking of brain-tumor-associated myeloid cells in vivo through optical coherence tomography with plasmonic labeling and speckle modulation. ACS Nano 13, 7985–7995 (2019).

    CAS  Google Scholar 

  50. Si, P., Honkala, A., de la Zerda, A. & Smith, B. R. Optical microscopy and coherence tomography of cancer in living subjects. Trends Cancer 6, 205–222 (2020).

    CAS  Google Scholar 

  51. Wang, M., Kim, M., Xia, F. & Xu, C. Impact of the emission wavelengths on in vivo multiphoton imaging of mouse brains. Biomed. Opt. Express 10, 1905–1918 (2019).

    CAS  Google Scholar 

  52. Bakker, G.-J. et al. Intravital deep-tumor single-beam 2-, 3- and 4-photon microscopy. Preprint at bioRxiv https://doi.org/10.1101/2020.09.29.312827 (2020).

    Article  Google Scholar 

  53. Dondossola, E. et al. Intravital microscopy of osteolytic progression and therapy response of cancer lesions in the bone. Sci. Transl Med. 10, eaao5726 (2018).

    Google Scholar 

  54. Greenbaum, A. et al. Bone CLARITY: Clearing, imaging, and computational analysis of osteoprogenitors within intact bone marrow. Sci. Transl Med. 9, eaah6518 (2017).

    Google Scholar 

  55. Graf, B. W. et al. Long-term time-lapse multimodal intravital imaging of regeneration and bone-marrow-derived cell dynamics in skin. Technology 01, 8–19 (2013).

    Google Scholar 

  56. Haeger, A. et al. Collective cancer invasion forms an integrin-dependent radioresistant niche. J. Exp. Med. 217, e20181184 (2020).

    Google Scholar 

  57. Jiang, D. et al. Injury triggers fascia fibroblast collective cell migration to drive scar formation through N-cadherin. Nat. Commun. 11, 5653 (2020).

    CAS  Google Scholar 

  58. Giampieri, S. et al. Localized and reversible TGFβ signalling switches breast cancer cells from cohesive to single cell motility. Nat. Cell Biol. 11, 1287–1296 (2009).

    CAS  Google Scholar 

  59. Vennin, C. et al. CAF hierarchy driven by pancreatic cancer cell p53-status creates a pro-metastatic and chemoresistant environment via perlecan. Nat. Commun. 10, 3637 (2019).

    Google Scholar 

  60. Denais, C. M. et al. Nuclear envelope rupture and repair during cancer cell migration. Science 352, 353–358 (2016).

    CAS  Google Scholar 

  61. Erard, M., Dupré-Crochet, S. & Nüße, O. Biosensors for spatiotemporal detection of reactive oxygen species in cells and tissues. Am. J. Physiol. Regul. Integr. Comp. Physiol. 314, R667–R683 (2018).

    CAS  Google Scholar 

  62. You, S. et al. Intravital imaging by simultaneous label-free autofluorescence-multiharmonic microscopy. Nat. Commun. 9, 2125 (2018).

    Google Scholar 

  63. Heymann, F. et al. Polypropylene mesh implantation for hernia repair causes myeloid cell–driven persistent inflammation. JCI Insight 4, e123862 (2019).

    Google Scholar 

  64. Xie, X. et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat. Immunol. 21, 1119–1133 (2020).

    CAS  Google Scholar 

  65. Karreman, M. A. et al. Fast and precise targeting of single tumor cells in vivo by multimodal correlative microscopy. J. Cell Sci. 129, 444–456 (2016).

    CAS  Google Scholar 

  66. Gurevich, D. B. et al. Live imaging of wound angiogenesis reveals macrophage orchestrated vessel sprouting and regression. EMBO J. 37, e97786 (2018).

    Google Scholar 

  67. Marsh, E., Gonzalez, D. G., Lathrop, E. A., Boucher, J. & Greco, V. Positional stability and membrane occupancy define skin fibroblast homeostasis. Cell 175, 1620–1633.e13 (2018).

    CAS  Google Scholar 

  68. Lämmermann, T. et al. Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature 498, 371–375 (2013).

    Google Scholar 

  69. Kozai, T. D. Y., Jaquins-Gerstl, A. S., Vazquez, A. L., Michael, A. C. & Cui, X. T. Brain tissue responses to neural implants impact signal sensitivity and intervention strategies. ACS Chem. Neurosci. 6, 48–67 (2015).

    CAS  Google Scholar 

  70. Holstein, J. H. et al. Intravital microscopic studies of angiogenesis during bone defect healing in mice calvaria. Injury 42, 765–771 (2011).

    CAS  Google Scholar 

  71. Alieva, M., Ritsma, L., Giedt, R. J., Weissleder, R. & van Rheenen, J. Imaging windows for long-term intravital imaging. Intravital 3, e29917 (2014).

    Google Scholar 

  72. Doloff, J. C. et al. Colony stimulating factor-1 receptor is a central component of the foreign body response to biomaterial implants in rodents and non-human primates. Nat. Mater. 16, 671–680 (2017).

    CAS  Google Scholar 

  73. MacRae, C. A. & Peterson, R. T. Zebrafish as tools for drug discovery. Nat. Rev. Drug Discov. 14, 721–731 (2015).

    CAS  Google Scholar 

  74. Witherel, C. E., Gurevich, D., Collin, J. D., Martin, P. & Spiller, K. L. Host–biomaterial interactions in zebrafish. ACS Biomater. Sci. Eng. 4, 1233–1240 (2018).

    CAS  Google Scholar 

  75. Zhang, X. et al. The zebrafish embryo as a model to quantify early inflammatory cell responses to biomaterials. J. Biomed. Mater. Res. A 105, 2522–2532 (2017).

    CAS  Google Scholar 

  76. White, R. M. et al. Transparent adult zebrafish as a tool for in vivo transplantation analysis. Cell Stem Cell 2, 183–189 (2008).

    CAS  Google Scholar 

  77. Wancket, L. M. Animal models for evaluation of bone implants and devices: comparative bone structure and common model uses. Vet. Pathol. 52, 842–850 (2015).

    CAS  Google Scholar 

  78. Sandison, J. C. A method for the microscopic study of the growth of transplanted bone in the transparent chamber of the rabbit’s ear. Anat. Rec. 40, 41–49 (1928).

    Google Scholar 

  79. Albrektsson, T. & Albrektsson, B. Microcirculation in grafted bone: a chamber technique for vital microscopy of rabbit bone transplants. Acta Orthop. Scand. 49, 1–7 (1978).

    CAS  Google Scholar 

  80. Hsieh, A. S., Winet, H., Bao, J. Y., Glas, H. & Plenk, H. Evidence for reperfusion injury in cortical bone as a function of crush injury ischemia duration: a rabbit bone chamber study. Bone 28, 94–103 (2001).

    CAS  Google Scholar 

  81. Penel, G., Delfosse, C., Descamps, M. & Leroy, G. Composition of bone and apatitic biomaterials as revealed by intravital Raman microspectroscopy. Bone 36, 893–901 (2005).

    CAS  Google Scholar 

  82. Veronesi, F. et al. Osseointegration is improved by coating titanium implants with a nanostructured thin film with titanium carbide and titanium oxides clustered around graphitic carbon. Mater. Sci. Eng. C Mater. Biol. Appl. 70, 264–271 (2017).

    CAS  Google Scholar 

  83. Diekmann, J. et al. Examination of a biodegradable magnesium screw for the reconstruction of the anterior cruciate ligament: A pilot in vivo study in rabbits. Mater. Sci. Eng. C Mater. Biol. Appl. 59, 1100–1109 (2016).

    CAS  Google Scholar 

  84. Ribatti, D., Annese, T. & Tamma, R. The use of the chick embryo CAM assay in the study of angiogenic activiy of biomaterials. Microvasc. Res. 131, 104026 (2020).

    CAS  Google Scholar 

  85. Ling, T.-Y. et al. Differentiation of lung stem/progenitor cells into alveolar pneumocytes and induction of angiogenesis within a 3D gelatin – Microbubble scaffold. Biomaterials 35, 5660–5669 (2014).

    CAS  Google Scholar 

  86. Moreno-Jiménez, I. et al. The chorioallantoic membrane (CAM) assay for the study of human bone regeneration: a refinement animal model for tissue engineering. Sci. Rep. 6, 32168 (2016).

    Google Scholar 

  87. Valdes, T. I., Kreutzer, D. & Moussy, F. The chick chorioallantoic membrane as a novel in vivo model for the testing of biomaterials. J. Biomed. Mater. Res. 62, 273–282 (2002).

    CAS  Google Scholar 

  88. Woloszyk, A., Liccardo, D. & Mitsiadis, T. A. Three-dimensional imaging of the developing vasculature within stem cell-seeded scaffolds cultured in ovo. Front. Physiol. 7, 146 (2016).

    Google Scholar 

  89. Rodrigues, M., Kosaric, N., Bonham, C. A. & Gurtner, G. C. Wound healing: a cellular perspective. Physiol. Rev. 99, 665–706 (2019).

    CAS  Google Scholar 

  90. Schmidt, A., von Woedtke, T., Vollmar, B., Hasse, S. & Bekeschus, S. Nrf2 signaling and inflammation are key events in physical plasma-spurred wound healing. Theranostics 9, 1066–1084 (2019).

    CAS  Google Scholar 

  91. Krzyszczyk, P., Schloss, R., Palmer, A. & Berthiaume, F. The role of macrophages in acute and chronic wound healing and interventions to promote pro-wound healing phenotypes. Front. Physiol. 9, 419 (2018).

    Google Scholar 

  92. Lu, H. et al. Fibroblast transdifferentiation promotes conversion of M1 macrophages and replenishment of cardiac resident macrophages following cardiac injury in mice. Eur. J. Immunol. 50, 795–808 (2020).

    CAS  Google Scholar 

  93. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 16, 582–598 (2016).

    CAS  Google Scholar 

  94. Liu, Y., Rath, B., Tingart, M. & Eschweiler, J. Role of implants surface modification in osseointegration: A systematic review. J. Biomed. Mater. Res. A 108, 470–484 (2020).

    CAS  Google Scholar 

  95. Li, J. et al. In vitro and in vivo evaluations of mechanical properties, biocompatibility and osteogenic ability of sintered porous titanium alloy implant. RSC Adv. 8, 36512–36520 (2018).

    CAS  Google Scholar 

  96. Sanchez, C. J. et al. Staphylococcus aureus biofilms decrease osteoblast viability, inhibits osteogenic differentiation, and increases bone resorption in vitro. BMC Musculoskelet. Disord. 14, 187 (2013).

    Google Scholar 

  97. Lee, H. G. et al. Aggravation of inflammatory response by costimulation with titanium particles and mechanical perturbations in osteoblast- and macrophage-like cells. Am. J. Physiol. Cell Physiol. 304, C431–C439 (2013).

    CAS  Google Scholar 

  98. Katou, F., Andoh, N., Motegi, K. & Nagura, H. Immuno-inflammatory responses in the tissue adjacent to titanium miniplates used in the treatment of mandibular fractures. J. Craniomaxillofac. Surg. 24, 155–162 (1996).

    CAS  Google Scholar 

  99. Riviș, M. et al. The implications of titanium alloys applied in maxillofacial osteosynthesis. Appl. Sci. 10, 3203 (2020).

    Google Scholar 

  100. Perino, G. et al. Diagnostic guidelines for the histological particle algorithm in the periprosthetic neo-synovial tissue. BMC Clin. Pathol. 18, 7 (2018).

    CAS  Google Scholar 

  101. Lechner, J., Noumbissi, S. & von Baehr, V. Titanium implants and silent inflammation in jawbone — a critical interplay of dissolved titanium particles and cytokines TNF-α and RANTES/CCL5 on overall health? EPMA J. 9, 331–343 (2018).

    Google Scholar 

  102. Khosravi, N., Maeda, A., DaCosta, R. S. & Davies, J. E. Nanosurfaces modulate the mechanism of peri-implant endosseous healing by regulating neovascular morphogenesis. Commun. Biol. 1, 72 (2018).

    Google Scholar 

  103. Mouraret, S. et al. A pre-clinical murine model of oral implant osseointegration. Bone 58, 177–184 (2014).

    CAS  Google Scholar 

  104. Wang, B. et al. Mechanoadaptive strain and functional osseointegration of dental implants in rats. Bone 137, 115375 (2020).

    CAS  Google Scholar 

  105. Bandaru, P. et al. Mechanical cues regulating proangiogenic potential of human mesenchymal stem cells through YAP-mediated mechanosensing. Small 16, 2001837 (2020).

    CAS  Google Scholar 

  106. Loebel, C., Mauck, R. L. & Burdick, J. A. Local nascent protein deposition and remodelling guide mesenchymal stromal cell mechanosensing and fate in three-dimensional hydrogels. Nat. Mater. 18, 883–891 (2019).

    CAS  Google Scholar 

  107. Sparks, D. S. et al. A preclinical large-animal model for the assessment of critical-size load-bearing bone defect reconstruction. Nat. Protoc. 15, 877–924 (2020).

    CAS  Google Scholar 

  108. Holzapfel, B. M. et al. Tissue engineered humanized bone supports human hematopoiesis in vivo. Biomaterials 61, 103–114 (2015).

    CAS  Google Scholar 

  109. Fakhry, M., Hamade, E., Badran, B., Buchet, R. & Magne, D. Molecular mechanisms of mesenchymal stem cell differentiation towards osteoblasts. World J. Stem Cell 5, 136–148 (2013).

    Google Scholar 

  110. Deckers, M. M. L. et al. Bone morphogenetic proteins stimulate angiogenesis through osteoblast-derived vascular endothelial growth factor A. Endocrinology 143, 1545–1553 (2002).

    CAS  Google Scholar 

  111. Abarrategi, A. et al. Versatile humanized niche model enables study of normal and malignant human hematopoiesis. J. Clin. Invest. 127, 543–548 (2017).

    Google Scholar 

  112. Xie, Y. et al. Osteoimmunomodulatory effects of biomaterial modification strategies on macrophage polarization and bone regeneration. Regen. Biomater. 7, 233–245 (2020).

    CAS  Google Scholar 

  113. Benson, R. A. et al. Non-invasive multiphoton imaging of islets transplanted into the pinna of the NOD mouse ear reveals the immediate effect of anti-CD3 treatment in autoimmune diabetes. Front. Immunol. 9, 1006 (2018).

    Google Scholar 

  114. Samuel, R. et al. Generation of functionally competent and durable engineered blood vessels from human induced pluripotent stem cells. Proc. Natl Acad. Sci. USA 110, 12774–12779 (2013).

    CAS  Google Scholar 

  115. van den Berg, C. W. et al. Renal subcapsular transplantation of PSC-derived kidney organoids induces neo-vasculogenesis and significant glomerular and tubular maturation in vivo. Stem Cell Rep. 10, 751–765 (2018).

    Google Scholar 

  116. Rudnicki, M. et al. Hypoxia response and VEGF-A expression in human proximal tubular epithelial cells in stable and progressive renal disease. Lab. Invest. 89, 337–346 (2009).

    CAS  Google Scholar 

  117. Schepers, A. G. et al. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science 337, 730–735 (2012).

    CAS  Google Scholar 

  118. Richardson, A., Park, M., Watson, S. L., Wakefield, D. & Di Girolamo, N. Visualizing the fate of transplanted K14-confetti corneal epithelia in a mouse model of limbal stem cell deficiency. Investig. Ophthalmol. Vis. Sci. 59, 1630–1640 (2018).

    CAS  Google Scholar 

  119. Nalbach, L. et al. Improvement of islet transplantation by the fusion of islet cells with functional blood vessels. EMBO Mol. Med. 13, e12616 (2021).

    CAS  Google Scholar 

  120. Koffler, J. et al. Improved vascular organization enhances functional integration of engineered skeletal muscle grafts. Proc. Natl Acad. Sci. USA 108, 14789–14794 (2011).

    CAS  Google Scholar 

  121. Perry, L., Merdler, U., Elishaev, M. & Levenberg, S. Enhanced host neovascularization of prevascularized engineered muscle following transplantation into immunocompetent versus immunocompromised mice. Cells 8, 1472 (2019).

    CAS  Google Scholar 

  122. Juhas, M., Engelmayr, G. C., Fontanella, A. N., Palmer, G. M. & Bursac, N. Biomimetic engineered muscle with capacity for vascular integration and functional maturation in vivo. Proc. Natl Acad. Sci. USA 111, 5508–5513 (2014).

    CAS  Google Scholar 

  123. Juhas, M. et al. Incorporation of macrophages into engineered skeletal muscle enables enhanced muscle regeneration. Nat. Biomed. Eng. 2, 942–954 (2018).

    CAS  Google Scholar 

  124. Perry, L., Landau, S., Flugelman, M. Y. & Levenberg, S. Genetically engineered human muscle transplant enhances murine host neovascularization and myogenesis. Commun. Biol. 1, 161 (2018).

    Google Scholar 

  125. Calcagni, M. et al. In vivo visualization of the origination of skin graft vasculature in a wild-type/GFP crossover model. Microvasc. Res. 82, 237–245 (2011).

    Google Scholar 

  126. Celli, S., Albert, M. L. & Bousso, P. Visualizing the innate and adaptive immune responses underlying allograft rejection by two-photon microscopy. Nat. Med. 17, 744–749 (2011).

    CAS  Google Scholar 

  127. Chen, Q. et al. In situ sprayed bioresponsive immunotherapeutic gel for post-surgical cancer treatment. Nat. Nanotechnol. 14, 89–97 (2019).

    CAS  Google Scholar 

  128. Kastrup, C. J. et al. Painting blood vessels and atherosclerotic plaques with an adhesive drug depot. Proc. Natl Acad. Sci. USA 109, 21444–21449 (2012).

    CAS  Google Scholar 

  129. Eles, J. R., Vazquez, A. L., Kozai, T. D. Y. & Cui, X. T. Meningeal inflammatory response and fibrous tissue remodeling around intracortical implants: An in vivo two-photon imaging study. Biomaterials 195, 111–123 (2019).

    CAS  Google Scholar 

  130. Kozai, T. D. Y., Jaquins-Gerstl, A. S., Vazquez, A. L., Michael, A. C. & Cui, X. T. Dexamethasone retrodialysis attenuates microglial response to implanted probes in vivo. Biomaterials 87, 157–169 (2016).

    CAS  Google Scholar 

  131. Wei, Q. et al. Protein interactions with polymer coatings and biomaterials. Angew. Chem. Int. Ed. 53, 8004–8031 (2014).

    CAS  Google Scholar 

  132. Faust, J. J. et al. An actin-based protrusion originating from a podosome-enriched region initiates macrophage fusion. Mol. Biol. Cell 30, 2254–2267 (2019).

    CAS  Google Scholar 

  133. Selders, G. S., Fetz, A. E., Radic, M. Z. & Bowlin, G. L. An overview of the role of neutrophils in innate immunity, inflammation and host-biomaterial integration. Regen. Biomater. 4, 55–68 (2017).

    CAS  Google Scholar 

  134. Koschwanez, H. E., Reichert, W. M. & Klitzman, B. Intravital microscopy evaluation of angiogenesis and its effects on glucose sensor performance. J. Biomed. Mater. Res. A 93A, 1348–1357 (2010).

    CAS  Google Scholar 

  135. Kwee, B. J. & Mooney, D. J. Manipulating the intersection of angiogenesis and inflammation. Ann. Biomed. Eng. 43, 628–640 (2015).

    Google Scholar 

  136. Witherel, C. E., Abebayehu, D., Barker, T. H. & Spiller, K. L. Macrophage and fibroblast interactions in biomaterial-mediated fibrosis. Adv. Healthc. Mater. 8, e1801451 (2019).

    Google Scholar 

  137. Filová, E. et al. Analysis and three-dimensional visualization of collagen in artificial scaffolds using nonlinear microscopy techniques. J. Biomed. Opt. 15, 066011 (2010).

    Google Scholar 

  138. Druecke, D. et al. Neovascularization of poly(ether ester) block-copolymer scaffolds in vivo: Long-term investigations using intravital fluorescent microscopy. J. Biomed. Mater. Res. A 68A, 10–18 (2004).

    CAS  Google Scholar 

  139. Klenke, F. M. et al. Impact of pore size on the vascularization and osseointegration of ceramic bone substitutes in vivo. J. Biomed. Mater. Res. A 85A, 777–786 (2008).

    CAS  Google Scholar 

  140. Jang, G. H., Hwang, M. P., Kim, S. Y., Jang, H. S. & Lee, K. H. A systematic in-vivo toxicity evaluation of nanophosphor particles via zebrafish models. Biomaterials 35, 440–449 (2014).

    CAS  Google Scholar 

  141. Reismann, D. et al. Longitudinal intravital imaging of the femoral bone marrow reveals plasticity within marrow vasculature. Nat. Commun. 8, 2153 (2017).

    Google Scholar 

  142. Stiers, P.-J., van Gastel, N., Moermans, K., Stockmans, I. & Carmeliet, G. An ectopic imaging window for intravital imaging of engineered bone tissue. JBMR Plus 2, 92–102 (2018).

    Google Scholar 

  143. Wang, H. et al. Two-photon dual imaging platform for in vivo monitoring cellular oxidative stress in liver injury. Sci. Rep. 7, 45374 (2017).

    CAS  Google Scholar 

  144. Reissaus, C. A. et al. A versatile, portable intravital microscopy platform for studying beta-cell biology in vivo. Sci. Rep. 9, 8449 (2019).

    Google Scholar 

  145. Sanman, L. E., van der Linden, W. A., Verdoes, M. & Bogyo, M. Bifunctional probes of cathepsin protease activity and pH reveal alterations in endolysosomal pH during bacterial infection. Cell Chem. Biol. 23, 793–804 (2016).

    CAS  Google Scholar 

  146. Jaffer, F. A. et al. Optical visualization of cathepsin K activity in atherosclerosis with a novel, protease-activatable fluorescence sensor. Circulation 115, 2292–2298 (2007).

    CAS  Google Scholar 

  147. Sun, W. W. et al. Nanoarchitecture and dynamics of the mouse enteric glycocalyx examined by freeze-etching electron tomography and intravital microscopy. Commun. Biol. 3, 5 (2020).

    CAS  Google Scholar 

  148. Astone, M. et al. Zebrafish mutants and TEAD reporters reveal essential functions for Yap and Taz in posterior cardinal vein development. Sci. Rep. 8, 10189 (2018).

    Google Scholar 

  149. Subramanian, B. C. et al. The LTB4–BLT1 axis regulates actomyosin and β2-integrin dynamics during neutrophil extravasation. J. Cell Biol. 219, e201910215 (2020).

    CAS  Google Scholar 

  150. LeBleu, V. S. et al. Identification of human epididymis protein-4 as a fibroblast-derived mediator of fibrosis. Nat. Med. 19, 227–231 (2013).

    CAS  Google Scholar 

  151. Jain, N. & Vogel, V. Spatial confinement downsizes the inflammatory response of macrophages. Nat. Mater. 17, 1134–1144 (2018).

    CAS  Google Scholar 

  152. de Buhr, N. & von Köckritz-Blickwede, M. How neutrophil extracellular traps become visible. J. Immunol. Res. 2016, 4604713 (2016).

    Google Scholar 

  153. Honda, M. et al. Intravital imaging of neutrophil recruitment in hepatic ischemia-reperfusion injury in mice. Transplantation 95, 551–558 (2013).

    Google Scholar 

  154. Sedin, J. et al. High resolution intravital imaging of the renal immune response to injury and infection in mice. Front. Immunol. 10, 2744 (2019).

    CAS  Google Scholar 

  155. Yam, A. O. & Chtanova, T. Imaging the neutrophil: Intravital microscopy provides a dynamic view of neutrophil functions in host immunity. Cell. Immunol. 350, 103898 (2020).

    CAS  Google Scholar 

  156. Turk, M., Naumenko, V., Mahoney, D. J. & Jenne, C. N. Tracking cell recruitment and behavior within the tumor microenvironment using advanced intravital imaging approaches. Cells 7, 69 (2018).

    Google Scholar 

  157. Park, J. et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl Med. 8, 361ra138 (2016).

    Google Scholar 

  158. Handschuh, J., Amore, J. & Müller, A. J. From the cradle to the grave of an infection: host-pathogen interaction visualized by intravital microscopy. Cytom. A 97, 458–470 (2020).

    Google Scholar 

  159. Kreisel, D. et al. In vivo two-photon imaging reveals monocyte-dependent neutrophil extravasation during pulmonary inflammation. Proc. Natl Acad. Sci. USA 107, 18073–18078 (2010).

    CAS  Google Scholar 

  160. Yamamoto, N. et al. Cellular dynamics visualized in live cells in vitro and in vivo by differential dual-color nuclear-cytoplasmic fluorescent-protein expression. Cancer Res. 64, 4251–4256 (2004).

    CAS  Google Scholar 

  161. Burnett, S. H. et al. Conditional macrophage ablation in transgenic mice expressing a Fas-based suicide gene. J. Leukoc. Biol. 75, 612–623 (2004).

    CAS  Google Scholar 

  162. Nguyen-Chi, M. et al. Identification of polarized macrophage subsets in zebrafish. eLife 4, e07288 (2015).

    Google Scholar 

  163. Snippert, H. J. et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143, 134–144 (2010).

    CAS  Google Scholar 

  164. Yoshikawa, S. et al. Intravital imaging of Ca2+ signals in lymphocytes of Ca2+ biosensor transgenic mice: indication of autoimmune diseases before the pathological onset. Sci. Rep. 6, 18738 (2016).

    CAS  Google Scholar 

  165. Everhart, M. B. et al. Duration and intensity of NF-κB activity determine the severity of endotoxin-induced acute lung injury. J. Immunol. 176, 4995–5005 (2006).

    CAS  Google Scholar 

  166. Nicholls, P. J. et al. Measuring nonapoptotic caspase activity with a transgenic reporter in mice. Preprint at bioRxiv https://doi.org/10.1101/196105 (2021).

    Article  Google Scholar 

  167. Park, S.-J. et al. Imaging inflammation using an activated macrophage probe with Slc18b1 as the activation-selective gating target. Nat. Commun. 10, 1111 (2019).

    Google Scholar 

  168. Uddin, M. I. et al. Applications of azo-based probes for imaging retinal hypoxia. ACS Med. Chem. Lett. 6, 445–449 (2015).

    CAS  Google Scholar 

  169. Xiang, J. et al. Biocompatible green and red fluorescent organic dots with remarkably large two-photon action cross sections for targeted cellular imaging and real-time intravital blood vascular visualization. ACS Appl. Mater. Interfaces 7, 14965–14974 (2015).

    CAS  Google Scholar 

  170. Kim, H. Y. et al. Quantitative imaging of tumor-associated macrophages and their response to therapy using (64)Cu-labeled Macrin. ACS Nano 12, 12015–12029 (2018).

    CAS  Google Scholar 

  171. Smith, B. R. et al. Selective uptake of single-walled carbon nanotubes by circulating monocytes for enhanced tumour delivery. Nat. Nanotechnol. 9, 481–487 (2014).

    CAS  Google Scholar 

  172. Maeda, H. et al. Real-time intravital imaging of pH variation associated with osteoclast activity. Nat. Chem. Biol. 12, 579–585 (2016).

    CAS  Google Scholar 

  173. Pezzotti, G. Raman piezo-spectroscopic analysis of natural and synthetic biomaterials. Anal. Bioanal. Chem. 381, 577–590 (2005).

    CAS  Google Scholar 

  174. Wagner, R. L. Erlauterungstaflen zur Physiologie und Entwicklungsgeschichte (Leopold Voss, 1839).

  175. Sandison, J. C. Observations on the growth of blood vessels as seen in the transparent chamber introduced into the rabbit’s ear. Am. J. Anat. 41, 475–496 (1928).

    Google Scholar 

  176. Algire, G. H. An adaptation of the transparent-chamber technique to the mouse. J. Natl Cancer Inst. 4, 1–11 (1943).

    Google Scholar 

  177. Kiehn, C. L., Cebul, F., Berg, M., Gutentag, J. & Glover, D. M. A study of the vascularization of experimental bone grafts by means of radioactive phosphorus and the transparent chamber. Ann. Surg. 136, 404–411 (1952).

    CAS  Google Scholar 

  178. Funk, W., Endrich, B. & Messmer, K. A novel method for follow-up studies of the microcirculation in non-malignant tissue implants. Res. Exp. Med. 186, 259–270 (1986).

    CAS  Google Scholar 

  179. Brånemark, P. I. Intravital microscopy. Its present status and its potentialities. Med. Biol. Illus. 16, 100–108 (1966).

    Google Scholar 

  180. Mazo, I. B. et al. Hematopoietic progenitor cell rolling in bone marrow microvessels: parallel contributions by endothelial selectins and vascular cell adhesion molecule 1. J. Exp. Med. 188, 465–474 (1998).

    CAS  Google Scholar 

  181. Spanel-Borowski, K. The chick chorioallantoic membrane as test system for biocompatible materials. Res. Exp. Med. 189, 69–75 (1989).

    CAS  Google Scholar 

  182. Valdes, T. I., Klueh, U., Kreutzer, D. & Moussy, F. Ex ova chick chorioallantoic membrane as a novel in vivo model for testing biosensors. J. Biomed. Mater. Res. A 67A, 215–223 (2003).

    CAS  Google Scholar 

  183. Weigelin, B. et al. Cytotoxic T cells are able to efficiently eliminate cancer cells by additive cytotoxicity. Nat. Commun. https://doi.org/10.1038/s41467-021-25282-3 (2021).

    Article  Google Scholar 

  184. Schiessl, I. M., Fremter, K., Burford, J. L., Castrop, H. & Peti-Peterdi, J. Long-term cell fate tracking of individual renal cells using Serial intravital microscopy. Methods Mol. Biol. 2150, 25–44 (2020).

    CAS  Google Scholar 

  185. Urciuolo, A. et al. Intravital three-dimensional bioprinting. Nat. Biomed. Eng. 4, 901–915 (2020).

    CAS  Google Scholar 

Download references

Acknowledgements

The authors thank J. Conway and P. Timpson for helpful discussions. This work was supported by the MD Anderson Cancer Center Prostate Cancer SPORE (P50 CA140388-09) and Bayer HealthCare Pharmaceuticals (57440) to E.D. and the NWO-VICI (918.11.626), the European Research Council (617430-DEEPINSIGHT), the NWO Gravity Program Institute for Chemical Immunology and the Cancer Genomics Center, the Netherlands to P.F. The Genitourinary Cancers Program of the Cancer Center Support Grant (CCSG) shared resources at MD Anderson Cancer Center is supported by NIH/NCI award number P30 CA016672.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Eleonora Dondossola or Peter Friedl.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dondossola, E., Friedl, P. Host responses to implants revealed by intravital microscopy. Nat Rev Mater 7, 6–22 (2022). https://doi.org/10.1038/s41578-021-00369-x

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41578-021-00369-x

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research