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:

Chronic in vivo imaging in the mouse spinal cord using an implanted chamber

Abstract

Understanding and treatment of spinal cord pathology is limited in part by a lack of time-lapse in vivo imaging strategies at the cellular level. We developed a chronically implanted spinal chamber and surgical procedure suitable for time-lapse in vivo multiphoton microscopy of mouse spinal cord without the need for repeat surgical procedures. We routinely imaged mice repeatedly for more than 5 weeks postoperatively with up to ten separate imaging sessions and observed neither motor-function deficit nor neuropathology in the spinal cord as a result of chamber implantation. Using this chamber we quantified microglia and afferent axon dynamics after a laser-induced spinal cord lesion and observed massive microglia infiltration within 1 d along with a heterogeneous dieback of axon stumps. By enabling chronic imaging studies over timescales ranging from minutes to months, our method offers an ideal platform for understanding cellular dynamics in response to injury and therapeutic interventions.

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

Figure 1: An imaging chamber for longitudinal optical access to mouse spinal cord without the need for repeated surgeries.
Figure 2: Longitudinal 2PEF imaging of axons and blood vessels over many weeks after surgery.
Figure 3: Histological analysis of reactive microglia and astrocytes, and tissue morphology after chamber implantation.
Figure 4: Imaging and quantification of microglial scar formation at the site of a laser-induced SCI.
Figure 5: 2PEF imaging and quantification of axon dieback after a laser-induced SCI.

Similar content being viewed by others

References

  1. Kobat, D. et al. Deep tissue multiphoton microscopy using longer wavelength excitation. Opt. Express 17, 13354–13364 (2009).

    Article  Google Scholar 

  2. Christie, R.H. et al. Growth arrest of individual senile plaques in a model of Alzheimer′s disease observed by in vivo multiphoton microscopy. J. Neurosci. 21, 858–864 (2001).

    Article  CAS  Google Scholar 

  3. Yan, P. et al. Characterizing the appearance and growth of amyloid plaques in APP/PS1 mice. J. Neurosci. 29, 10706–10714 (2009).

    Article  CAS  Google Scholar 

  4. Meyer-Luehmann, M. et al. Rapid appearance and local toxicity of amyloid-beta plaques in a mouse model of Alzheimer′s disease. Nature 451, 720–724 (2008).

    Article  CAS  Google Scholar 

  5. Prada, C.M. et al. Antibody-mediated clearance of amyloid-beta peptide from cerebral amyloid angiopathy revealed by quantitative in vivo imaging. J. Neurosci. 27, 1973–1980 (2007).

    Article  CAS  Google Scholar 

  6. Kienast, Y. et al. Real-time imaging reveals the single steps of brain metastasis formation. Nat. Med. 16, 116–122 (2010).

    Article  CAS  Google Scholar 

  7. Lam, C.K., Yoo, T., Hiner, B., Liu, Z. & Grutzendler, J. Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization. Nature 465, 478–482 (2010).

    Article  CAS  Google Scholar 

  8. Holtmaat, A. et al. Long-term, high-resolution imaging in the mouse neocortex through a chronic cranial window. Nat. Protoc. 4, 1128–1144 (2009).

    Article  CAS  Google Scholar 

  9. Yang, G., Pan, F., Parkhurst, C.N., Grutzendler, J. & Gan, W.B. Thinned-skull cranial window technique for long-term imaging of the cortex in live mice. Nat. Protoc. 5, 201–208 (2010).

    Article  CAS  Google Scholar 

  10. Drew, P.J. et al. Chronic optical access through a polished and reinforced thinned skull. Nat. Methods 7, 981–984 (2010).

    Article  CAS  Google Scholar 

  11. Bhatt, D.H., Otto, S.J., Depoister, B. & Fetcho, J.R. Cyclic AMP-induced repair of zebrafish spinal circuits. Science 305, 254–258 (2004).

    Article  CAS  Google Scholar 

  12. Kerschensteiner, M., Schwab, M.E., Lichtman, J.W. & Misgeld, T. In vivo imaging of axonal degeneration and regeneration in the injured spinal cord. Nat. Med. 11, 572–577 (2005).

    Article  CAS  Google Scholar 

  13. Davalos, D. et al. Stable in vivo imaging of densely populated glia, axons and blood vessels in the mouse spinal cord using two-photon microscopy. J. Neurosci. Methods 169, 1–7 (2008).

    Article  Google Scholar 

  14. Dibaj, P. et al. NO mediates microglial response to acute spinal cord injury under ATP control in vivo. Glia 58, 1133–1144 (2010).

    Article  Google Scholar 

  15. Johannssen, H.C. & Helmchen, F. In vivo Ca2+ imaging of dorsal horn neuronal populations in mouse spinal cord. J. Physiol. (Lond.) 588, 3397–3402 (2010).

    Article  CAS  Google Scholar 

  16. Ylera, B. et al. Chronically CNS-injured adult sensory neurons gain regenerative competence upon a lesion of their peripheral axon. Curr. Biol. 19, 930–936 (2009).

    Article  CAS  Google Scholar 

  17. Kim, J.V. et al. Two-photon laser scanning microscopy imaging of intact spinal cord and cerebral cortex reveals requirement for CXCR6 and neuroinflammation in immune cell infiltration of cortical injury sites. J. Immunol. Methods 352, 89–100 (2010).

    Article  CAS  Google Scholar 

  18. Dray, C., Rougon, G. & Debarbieux, F. Quantitative analysis by in vivo imaging of the dynamics of vascular and axonal networks in injured mouse spinal cord. Proc. Natl. Acad. Sci. USA 106, 9459–9464 (2009).

    Article  CAS  Google Scholar 

  19. Farrar, M.J., Wise, F.W., Fetcho, J.R. & Schaffer, C.B. In vivo imaging of myelin in the vertebrate central nervous system using third harmonic generation microscopy. Biophys. J. 100, 1362–1371 (2011).

    Article  CAS  Google Scholar 

  20. Crawley, J.N. What's Wrong with My Mouse? Behavioral Phenotyping of Transgenic and Knockout Mice (Wiley-Liss, New York, 2000).

  21. Streit, W.J., Walter, S.A. & Pennell, N.A. Reactive microgliosis. Prog. Neurobiol. 57, 563–581 (1999).

    Article  CAS  Google Scholar 

  22. Tanaka, T., Ueno, M. & Yamashita, T. Engulfment of axon debris by microglia requires p38 MAPK activity. J. Biol. Chem. 284, 21626–21636 (2009).

    Article  CAS  Google Scholar 

  23. Aldskogius, H. & Kozlova, E.N. Central neuron-glial and glial-glial interactions following axon injury. Prog. Neurobiol. 55, 1–26 (1998).

    Article  CAS  Google Scholar 

  24. Garcia-Alias, G. et al. Therapeutic time window for the application of chondroitinase ABC after spinal cord injury. Exp. Neurol. 210, 331–338 (2008).

    Article  CAS  Google Scholar 

  25. Rolls, A., Shechter, R. & Schwartz, M. The bright side of the glial scar in CNS repair. Nat. Rev. Neurosci. 10, 235–241 (2009).

    Article  CAS  Google Scholar 

  26. Tator, C.H. Review of treatment trials in human spinal cord injury: issues, difficulties, and recommendations. Neurosurgery 59, 957–987 (2006).

    Article  Google Scholar 

  27. Thuret, S., Moon, L.D. & Gage, F.H. Therapeutic interventions after spinal cord injury. Nat. Rev. Neurosci. 7, 628–643 (2006).

    Article  CAS  Google Scholar 

  28. Seif, G.I., Nomura, H. & Tator, C.H. Retrograde axonal degeneration (“dieback”) in the corticospinal tract after transection injury of the rat spinal cord: a confocal microscopy study. J. Neurotrauma 24, 1513–1528 (2007).

    Article  Google Scholar 

  29. Silver, J., Horn, K.P., Busch, S.A., Hawthorne, A.L. & van Rooijen, N. Another barrier to regeneration in the CNS: activated macrophages induce extensive retraction of dystrophic axons through direct physical interactions. J. Neurosci. 28, 9330–9341 (2008).

    Article  Google Scholar 

  30. Xu, H.T., Pan, F., Yang, G. & Gan, W.B. Choice of cranial window type for in vivo imaging affects dendritic spine turnover in the cortex. Nat. Neurosci. 10, 549–551 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the US National Institutes of Health (DP OD006411 to J.R.F. and R01 EB002019 to C.B.S.) and the National Science and Research Council of Canada (to M.J.F.) for financial support, IMRA America, Inc. for the loan of their FCPA μJewel D-400 laser, J. Siebert for critically reading this manuscript, N. Ellis for his assistance in the machine shop and M. Riccio for his assistance with the MicroCT imaging.

Author information

Authors and Affiliations

Authors

Contributions

M.J.F., T.A.C., J.R.F. and C.B.S. conceived and designed the experiments. M.J.F. performed surgeries and imaging experiments, I.M.B. performed behavioral assays, and D.H.S. performed histopathology. M.J.F., I.M.B., J.R.F. and C.B.S. analyzed data. J.R.F., T.A.C. and C.B.S. contributed reagents and materials. M.J.F., J.R.F. and C.B.S. wrote the paper.

Corresponding author

Correspondence to Chris B Schaffer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5, Supplementary Notes 1–7, Supplementary Protocol (PDF 1956 kb)

Supplementary Video 1

Rendering of MicroCT image data from a mouse taken 6 d after implantation of the imaging chamber, showing normal spine alignment and no vertebral damage. (MOV 7283 kb)

Supplementary Video 2

Video of a mouse taken 2 weeks after implanting the imaging chamber, showing locomotion, grooming and exploratory behavior. (MOV 7639 kb)

Supplementary Video 3

A series of 2PEF image stacks taken at different times after a laser-induced spinal cord injury, showing GFP-expressing axons (green) and Texas Red-dextran labeled vasculature (red). (MOV 29674 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Farrar, M., Bernstein, I., Schlafer, D. et al. Chronic in vivo imaging in the mouse spinal cord using an implanted chamber. Nat Methods 9, 297–302 (2012). https://doi.org/10.1038/nmeth.1856

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.1856

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