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.

  • Technical Report
  • Published:

Panoptic imaging of transparent mice reveals whole-body neuronal projections and skull–meninges connections

Abstract

Analysis of entire transparent rodent bodies after clearing could provide holistic biological information in health and disease, but reliable imaging and quantification of fluorescent protein signals deep inside the tissues has remained a challenge. Here, we developed vDISCO, a pressure-driven, nanobody-based whole-body immunolabeling technology to enhance the signal of fluorescent proteins by up to two orders of magnitude. This allowed us to image and quantify subcellular details through bones, skin and highly autofluorescent tissues of intact transparent mice. For the first time, we visualized whole-body neuronal projections in adult mice. We assessed CNS trauma effects in the whole body and found degeneration of peripheral nerve terminals in the torso. Furthermore, vDISCO revealed short vascular connections between skull marrow and brain meninges, which were filled with immune cells upon stroke. Thus, our new approach enables unbiased comprehensive studies of the interactions between the nervous system and the rest of the body.

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: Enhancement and permanent preservation of the fluorescence signal with vDISCO.
Fig. 2: vDISCO panoptic imaging uncovers neuronal projections in intact mice.
Fig. 3: vDISCO panoptic imaging on mice with the intact skin.
Fig. 4: TBI-induced peripheral nerve degeneration revealed by vDISCO panoptic imaging.
Fig. 5: Visualizing meningeal vessels through intact skull by vDISCO panoptic imaging.
Fig. 6: Uncovering SMCs through intact skull by vDISCO panoptic imaging.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Tuchin, V. V. & Tuchin, V. Tissue Optics: Light Scattering Methods and Instruments for Medical Diagnosis (SPIE Press, Bellingham, 2007).

    Book  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. Yang, B. et al. Single-cell phenotyping within transparent intact tissue through whole-body clearing. Cell 158, 945–958 (2014).

    Article  CAS  Google Scholar 

  4. Renier, N. et al. iDISCO: a simple, rapid method to immunolabel large tissue samples for volume imaging. Cell 159, 896–910 (2014).

    Article  CAS  Google Scholar 

  5. Susaki, E. A. et al. Whole-brain imaging with single-cell resolution using chemical cocktails and computational analysis. Cell 157, 726–739 (2014).

    Article  CAS  Google Scholar 

  6. Erturk, A. et al. Three-dimensional imaging of solvent-cleared organs using 3DISCO. Nat. Protoc. 7, 1983–1995 (2012).

    Article  CAS  Google Scholar 

  7. Erturk, A. et al. Three-dimensional imaging of the unsectioned adult spinal cord to assess axon regeneration and glial responses after injury. Nat. Med. 18, 166–171 (2011).

    Article  Google Scholar 

  8. Belle, M. et al. A simple method for 3D analysis of immunolabeled axonal tracts in a transparent nervous system. Cell Rep. 9, 1191–1201 (2014).

    Article  CAS  Google Scholar 

  9. Costantini, I. et al. A versatile clearing agent for multi-modal brain imaging. Sci. Rep. 5, 9808 (2015).

    Article  CAS  Google Scholar 

  10. Ke, M. T., Fujimoto, S. & Imai, T. SeeDB: a simple and morphology-preserving optical clearing agent for neuronal circuit reconstruction. Nat. Neurosci. 16, 1154–1161 (2013).

    Article  CAS  Google Scholar 

  11. Dodt, H. U. et al. Ultramicroscopy: three-dimensional visualization of neuronal networks in the whole mouse brain. Nat. Methods 4, 331–336 (2007).

    Article  CAS  Google Scholar 

  12. Hama, H. et al. ScaleS: an optical clearing palette for biological imaging. Nat. Neurosci. 18, 1518–1529 (2015).

    Article  CAS  Google Scholar 

  13. Belle, M. et al. Tridimensional visualization and analysis of early human development. Cell 169, 161–173 (2017).

    Article  CAS  Google Scholar 

  14. Murray, E. et al. Simple, scalable proteomic imaging for high-dimensional profiling of intact systems. Cell 163, 1500–1514 (2015).

    Article  CAS  Google Scholar 

  15. Tainaka, K. et al. Whole-body imaging with single-cell resolution by tissue decolorization. Cell 159, 911–924 (2014).

    Article  CAS  Google Scholar 

  16. Jing, D. et al. Tissue clearing of both hard and soft tissue organs with the PEGASOS method. Cell Res. 28, 803–818 (2018).

    Article  CAS  Google Scholar 

  17. Pan, C. et al. Shrinkage-mediated imaging of entire organs and organisms using uDISCO. Nat. Methods 13, 859–867 (2016).

    Article  CAS  Google Scholar 

  18. Kubota, S. I. et al. Whole-body profiling of cancer metastasis with single-cell resolution. Cell Rep. 20, 236–250 (2017).

    Article  CAS  Google Scholar 

  19. Tuchin, V. V. Optical Methods for Biomedical Diagnosis (SPIE Press, Bellingham, WA, USA, 2016).

  20. Muyldermans, S. Nanobodies: natural single-domain antibodies. Annu. Rev. Biochem. 82, 775–797 (2013).

    Article  CAS  Google Scholar 

  21. Feng, G. et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron 28, 41–51 (2000).

    Article  CAS  Google Scholar 

  22. Quan, T. et al. NeuroGPS-Tree: automatic reconstruction of large-scale neuronal populations with dense neurites. Nat. Methods 13, 51–54 (2016).

    Article  CAS  Google Scholar 

  23. Renier, N. et al. Mapping of brain activity by automated volume analysis of immediate early genes. Cell 165, 1789–1802 (2016).

    Article  CAS  Google Scholar 

  24. GageG. J., KipkeD. R. & ShainW. Whole animal perfusion fixation for rodents. J. Vis. Exp. 65, e3564 (2012).

    Google Scholar 

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

    Article  Google Scholar 

  26. Leijnse, J. N. & D’Herde, K. Revisiting the segmental organization of the human spinal cord. J. Anat. 229, 384–393 (2016).

    Article  CAS  Google Scholar 

  27. Gimenez-Arnau, A. Standards for the protection of skin barrier function. Curr. Probl. Dermatol. 49, 123–134 (2016).

    Article  Google Scholar 

  28. Haeryfar, S. M. & Hoskin, D. W. Thy-1: more than a mouse pan-T cell marker. J. Immunol. 173, 3581–3588 (2004).

    Article  CAS  Google Scholar 

  29. Smith, D. H., Johnson, V. E. & Stewart, W. Chronic neuropathologies of single and repetitive TBI: substrates of dementia? Nature Rev. Neurol. 9, 211–221 (2013).

    Article  CAS  Google Scholar 

  30. Frei, K. Posttraumatic dystonia. J. Neurol. Sci. 379, 183–191 (2017).

    Article  Google Scholar 

  31. Williams, G., Schache, A. & Morris, M. E. Running abnormalities after traumatic brain injury. Brain Inj. 27, 434–443 (2013).

    Article  Google Scholar 

  32. Chen, Y., Constantini, S., Trembovler, V., Weinstock, M. & Shohami, E. An experimental model of closed head injury in mice: pathophysiology, histopathology, and cognitive deficits. J. Neurotrauma 13, 557–568 (1996).

    Article  CAS  Google Scholar 

  33. Erturk, A. et al. Interfering with the chronic immune response rescues chronic degeneration after traumatic brain injury. J. Neurosci. 36, 9962–9975 (2016).

    Article  Google Scholar 

  34. Evans, T. M. et al. The effect of mild traumatic brain injury on peripheral nervous system pathology in wild-type mice and the G93A mutant mouse model of motor neuron disease. Neuroscience 298, 410–423 (2015).

    Article  CAS  Google Scholar 

  35. Louveau, A. et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341 (2015).

    Article  CAS  Google Scholar 

  36. Nedergaard, M. Neuroscience. Garbage truck of the brain. Science 340, 1529–1530 (2013).

    Article  CAS  Google Scholar 

  37. Da Mesquita, S. et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 560, 185–181 (2018).

    Article  Google Scholar 

  38. Andres, K. H., von During, M., Muszynski, K. & Schmidt, R. F. Nerve fibres and their terminals of the dura mater encephali of the rat. Anat. Embryol. 175, 289–301 (1987).

    Article  CAS  Google Scholar 

  39. Louveau, A. et al. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J. Clin. Investig. 127, 3210–3219 (2017).

    Article  Google Scholar 

  40. Choi, I. et al. Visualization of lymphatic vessels by Prox1-promoter directed GFP reporter in a bacterial artificial chromosome-based transgenic mouse. Blood 117, 362–365 (2011).

    Article  CAS  Google Scholar 

  41. Faust, N., Varas, F., Kelly, L. M., Heck, S. & Graf, T. Insertion of enhanced green fluorescent protein into the lysozyme gene creates mice with green fluorescent granulocytes and macrophages. Blood 96, 719–726 (2000).

    CAS  PubMed  Google Scholar 

  42. Llovera, G. et al. The choroid plexus is a key cerebral invasion route for T cells after stroke. Acta Neuropathol. 134, 851–868 (2017).

    Article  CAS  Google Scholar 

  43. Iqbal, A. J. et al. Human CD68 promoter GFP transgenic mice allow analysis of monocyte to macrophage differentiation in vivo. Blood 124, e33–e44 (2014).

    Article  Google Scholar 

  44. Hong, G., Antaris, A. L. & Dai, H. Near-infrared fluorophores for biomedical imaging. Nat. Biomed. Eng. 1, 0010 (2017).

    Article  Google Scholar 

  45. Noristani, H. N. et al. RNA-Seq analysis of microglia reveals time-dependent activation of specific genetic programs following spinal cord injury. Front. Mol. Neurosci. 10, 90 (2017).

    Article  Google Scholar 

  46. Villapol, S., Byrnes, K. R. & Symes, A. J. Temporal dynamics of cerebral blood flow, cortical damage, apoptosis, astrocyte-vasculature interaction and astrogliosis in the pericontusional region after traumatic brain injury. Front. Neurol. 5, 82 (2014).

    Article  Google Scholar 

  47. Leslie, M. Small but mighty. Science 360, 594–597 (2018).

    Article  CAS  Google Scholar 

  48. Deverman, B. E. et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat. Biotechnol. 34, 204–209 (2016).

    Article  CAS  Google Scholar 

  49. Hellal, F. et al. Microtubule stabilization reduces scarring and causes axon regeneration after spinal cord injury. Science 331, 928–931 (2011).

    Article  CAS  Google Scholar 

  50. Herisson, F. et al. Direct vascular channels connect skull bone marrow and the brain surface enabling myeloid cell migration. Nat. Neurosci. 21, 1209–1217 (2018).

    Article  CAS  Google Scholar 

  51. Niess, J. H. et al. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 307, 254–258 (2005).

    Article  CAS  Google Scholar 

  52. Nikic, I. et al. A reversible form of axon damage in experimental autoimmune encephalomyelitis and multiple sclerosis. Nat. Med. 17, 495–499 (2011).

    Article  CAS  Google Scholar 

  53. Treweek, J. B. et al. Whole-body tissue stabilization and selective extractions via tissue-hydrogel hybrids for high-resolution intact circuit mapping and phenotyping. Nat. Protoc. 10, 1860–1896 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Vascular Dementia Research Foundation, Synergy Excellence Cluster Munich (SyNergy; EXC 1010), ERA-Net Neuron (01EW1501A; A.E.), Fritz Thyssen Stiftung (reference 10.17.1.019MN; A.E.), DFG (reference ER 810/2-1; A.E.), National Institutes of Health (A.E. and M.N.), Helmholtz ICEMED Alliance (A.E.), the Novo Nordisk Foundation (M.N.), the Howard Hughes Medical Institute (B.T.K.) and the Lundbeck Foundation (A.L.R.X. and M.N.). We thank A. Weingart for illustrations, F. Hellal for technical advice and critical reading of the manuscript, and F. P. Quacquarelli and G. Locatelli for help during initial optimization. A.E, C.P., R.C., A.L. and M.I.T. are members of the Graduate School of Systemic Neurosciences at the Ludwig Maximilian University of Munich.

Author information

Authors and Affiliations

Authors

Contributions

A.E. initiated and led all aspects of the project. R.C. and C.P. developed the method and conducted most of the experiments. R.C. A.G., C.P., H.S.B., M.R., and B.M. analyzed data. M.I.T. stitched and analyzed the whole mouse body scans. A.P.D., B.F., S.Z. and L.M. helped to optimize the protocols. I.B., H.S.B., and S.L. helped to investigate skull–meninges connections. D.T. and M.K. contributed spinal cord injury experiments; C.B. and A.L., MCAO experiments; and A.X., B.K. and M.N., cisterna magna injection experiments. A.E., R.C. and C.P. wrote the paper. All the authors edited the manuscript.

Corresponding author

Correspondence to Ali Ertürk.

Ethics declarations

Competing interests

A.E. filed a patent on some of the technologies presented in this work.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–22 and Supplementary Tables 1 and 2

Reporting Summary

Supplementary Video 1

vDISCO reveals individual microglia in CX3CR1GFP/+ mouse brain. 3D brain reconstruction of a vDISCO boosted CX3CR1GFP/+ mouse brain, in which microglia express GFP, imaged with 4x objective by light-sheet microscopy. After vDISCO, all labelled microglia became evident, allowing quantification of their numbers in each brain region and assessment of the details of the microglia ramifications. Similar results were observed in 2 independent animals

Supplementary Video 2

vDISCO reveals whole body neuronal projections in Thy1-GFPM mouse. 3D reconstruction of neuronal projections in a Thy1-GFPM mouse scanned by light-sheet microscopy. The muscles (red) are visualized in autofluorescent channel (blue-green spectra). The bones and internal organs (white) are prominent with PI labeling. The GFP expressing neurons are boosted with nanobody conjugated with Atto 647N and imaged in far-red channel. The overall view of the entire labeled nervous system in the Thy1-GFPM mouse and fine details of neuronal connections are evident throughout the whole body. Comparable labeling and imaging results were achieved in 5 independent animals, whole body reconstruction was done in 2 mice

Supplementary Video 3

Neuronal projections from spinal cord to right forelimb in Thy1-GFPM mouse. 3D visualization obtained by light-sheet microscopy of neuronal projections from spinal cord to right forelimb of a Thy1-GFPM mouse. The muscles are shown in red, bones in white and the neurons in green. The fine details of axonal extensions and their endings at neuromuscular junctions are visible. Similar results were observed in 2 independent animals

Supplementary Video 4

vDISCO imaging of neuronal projections in the spinal cord and muscles. The first part of the video is the 2D orthoslicing of the spinal cord of an intact Thy1-GFPM mouse in dorso-ventral orientation. The muscles are shown in red, bones in white and the neurons in green. The details of neuronal cell bodies in ganglia embedded in the spinal cord vertebra and their axonal extensions into the CNS and PNS are visible. In the second part, neuronal connections (green) from spinal cord to muscles are shown in 3D and 2D. Similar results were observed in 2 independent animals

Supplementary Video 5

vDISCO imaging of CX3CR1GFP/+ mouse with intact skin. The first part of the video is the 3D reconstruction of the inguinal area from a CX3CR1GFP/+ mouse cleared with intact skin, showing inguinal lymph nodes and surrounding tissues (skin and muscles). CX3CR1 GFP+ immune cells are shown in cyan and cell nuclei labeled by PI are shown in magenta. The second part is the 2D orthoslicing visualization of a confocal scan of the same area, showing the subcellular details of CX3CR1 GFP+ cells in the lymph node and around the hair follicles. Similar results were observed in 2 independent animals

Supplementary Video 6

vDISCO reveals lymphatic vessels of different organs in Prox1-EGFP mouse. After applying vDISCO whole-body labeling of a Prox1-EGFP line mouse, the lungs and intestine were further imaged using high-magnification light-sheet microscopy. Prox1-EGFP lymphatic vessels (green) are evident throughout the tissues. Single experiment

Supplementary Video 7

Microglia and peripheral immune cells in CX3CR1GFP/+ x CCR2RFP/+ mouse. Multiplexed visualization of CX3CR1GFP/+ x CCR2RFP/+ transgenic mouse head after panoptic imaging with two different nanoboosters (anti-GFP conjugated to Atto647N and anti-RFP conjugated to Atto594N). The CX3CR1 GFP+ microglia cells in the brain parenchyma vs. CCR2 RFP+ peripheral immune cells in the meningeal vessels were clearly visible in 3D reconstruction and 2D orthoslicing. Similar results were observed from 3 independent double transgenic mice

Supplementary Video 8

Revealing short skull meninges connections (SMCs) in intact CX3CR1GFP/+ mouse heads. 2D orthoslicing of transparent head from the sagittal view of a CX3CR1GFP/+ line mouse imaged by light-sheet microscope. The vasculature was labelled by Lectin (red) and CX3CR1 GFP+ immune cells and microglia cells (green) were boosted by vDISCO. Short skull-meninges connections (SMCs) containing CX3CR1 GFP+ immune cells are evident. Similar results were observed in 3 independent animals

Supplementary Video 9

Cellular details of SMCs in intact mouse heads. 3D visualization of skull and brain interface in a CX3CR1GFP/+ line mouse imaged by confocal microscope. Vasculature was labelled with lectin (magenta) and CX3CR1 GFP+ cells (green) were boosted by vDISCO. Short vascular connections between skull marrow and meninges at the sagittal sinus and brain interfaces are clearly visualized. We also observed occasional connections between neighbouring skull marrow regions. Note that lectin dye is taken up by phagocytic cells in the skull marrow similar to dextran54. Similar results were observed in 3 independent animals

Supplementary Video 10

LysM GFP+ immune cells observed in SMCs upon ischemic stroke lesion. 2D orthoslicing of LysM-EGFP mouse head with MCAO. LysM GFP+ neutrophils and monocytes are shown in red and cell nuclei labelled by PI are shown in green. LysM GFP+ cells increased in the lesion site indicating the infiltration of immune cells after MCAO. In addition, many LysM GFP+ cells are observed in the vascular connections to the meninges suggesting that SMCs can be a potential cell trafficking route and play a role in the neuroinflammatory process. Similar results were observed from 4 independent mice per group

Supplementary Video 11

Widespread inflammation upon SCI assessed by panoptic vDISCO imaging. Panoptic imaging of CD68-EGFP transgenic mouse upon SCI. 2D orthoslicing in horizontal and sagittal views clearly shows the activated immune cells (green) in the muscles, spinal cord roots and meningeal compartments. Vertebra, which become prominent with PI labelling, are shown in white. Similar results were observed from 3 independent mice per group

Supplementary Video 12

Extensive details of neuronal connections in the mouse brain revealed by vDISCO. A Thy1-GFPM mouse brain was imaged by light-sheet microscopy more than a year after vDISCO. The details of the neuronal structures down to the subcellular level were imaged by Zeiss 20x objective on the light-sheet microscope. Similar results were observed in 2 independent animals

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cai, R., Pan, C., Ghasemigharagoz, A. et al. Panoptic imaging of transparent mice reveals whole-body neuronal projections and skull–meninges connections. Nat Neurosci 22, 317–327 (2019). https://doi.org/10.1038/s41593-018-0301-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-018-0301-3

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