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.

  • Letter
  • Published:

Virally mediated optogenetic excitation and inhibition of pain in freely moving nontransgenic mice

Abstract

Primary nociceptors are the first neurons involved in the complex processing system that regulates normal and pathological pain1. Because of constraints on pharmacological and electrical stimulation, noninvasive excitation and inhibition of these neurons in freely moving nontransgenic animals has not been possible. Here we use an optogenetic2 strategy to bidirectionally control nociceptors of nontransgenic mice. Intrasciatic nerve injection of adeno-associated viruses encoding an excitatory opsin enabled light-inducible stimulation of acute pain, place aversion and optogenetically mediated reductions in withdrawal thresholds to mechanical and thermal stimuli. In contrast, viral delivery of an inhibitory opsin enabled light-inducible inhibition of acute pain perception, and reversed mechanical allodynia and thermal hyperalgesia in a model of neuropathic pain. Light was delivered transdermally, allowing these behaviors to be induced in freely moving animals. This approach may have utility in basic and translational pain research, and enable rapid drug screening and testing of newly engineered opsins.

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: Intrasciatic injection of AAV6-hSyn-ChR2-eYFP results in transduction of unmyelinated nociceptors projecting to spinal cord lamina I/IIo.
Figure 2: Transdermal illumination of AAV6-hSyn-ChR2-eYFP–injected mice results in tunable pain-like behavior and sensitizes mice to mechanical and thermal stimuli.
Figure 3: Transdermal illumination of AAV6-hSyn-NpHR-eYFP–injected mice desensitizes mice to mechanical and thermal stimuli and reverses mechanical allodynia and thermal hyperalgesia caused by a chronic constriction injury (CCI).

Similar content being viewed by others

References

  1. Dubin, A.E. & Patapoutian, A. Review series: nociceptors: the sensors of the pain pathway. J. Clin. Invest. 120, 3760–3772 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Fenno, L., Yizhar, O. & Deisseroth, K. The development and application of optogenetics. Annu. Rev. Neurosci. 34, 389–412 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Liske, H. et al. Optical inhibition of motor nerve and muscle activity in vivo. Muscle Nerve 47, 916–921 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Llewellyn, M.E., Thompson, K.R., Deisseroth, K. & Delp, S.L. Orderly recruitment of motor units under optical control in vivo. Nat. Med. 16, 1161–1165 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ji, Z.-G. et al. Light-evoked somatosensory perception of transgenic rats that express channelrhodopsin-2 in dorsal root ganglion cells. PLoS ONE 7, e32699 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wang, H. & Zylka, M.J. Mrgprd-expressing polymodal nociceptive neurons innervate most known classes of substantia gelatinosa neurons. J. Neurosci. 29, 13202–13209 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mourot, A. et al. Rapid optical control of nociception with an ion-channel photoswitch. Nat. Methods 9, 396–402 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kokel, D. et al. Photochemical activation of TRPA1 channels in neurons and animals. Nat. Chem. Biol. 9, 257–263 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Anonymous. Enlightened engineering. Nat. Biotechnol. 29, 849 (2011).

  10. Towne, C., Montgomery, K.L., Iyer, S.M., Deisseroth, K. & Delp, S.L. Optogenetic control of targeted peripheral axons in freely moving animals. PLoS ONE 8, e72691 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Daou, I. et al. Remote optogenetic activation and sensitization of pain pathways in freely moving mice. J. Neurosci. 33, 18631–18640 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mattis, J. et al. Principles for applying optogenetic tools derived from direct comparative analysis of microbial opsins. Nat. Methods 9, 159–172 (2012).

    Article  CAS  Google Scholar 

  13. Williams, J.C. & Denison, T. From optogenetic technologies to neuromodulation therapies. Sci. Transl. Med. 5, 177ps6 (2013).

    Article  PubMed  Google Scholar 

  14. Chow, B.Y. & Boyden, E.S. Optogenetics and translational medicine. Sci. Transl. Med. 5, 177ps5 (2013).

    PubMed  Google Scholar 

  15. Towne, C., Schneider, B.L., Kieran, D., Redmond, D.E. & Aebischer, P. Efficient transduction of non-human primate motor neurons after intramuscular delivery of recombinant AAV serotype 6. Gene Ther. 17, 141–146 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. San Sebastian, W. et al. Adeno-associated virus type 6 is retrogradely transported in the non-human primate brain. Gene Ther. 20, 1178–1183 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Asokan, A., Schaffer, D.V. & Samulski, R.J. The AAV vector toolkit: poised at the clinical crossroads. Mol. Ther. 20, 699–708 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Towne, C., Pertin, M., Beggah, A.T., Aebischer, P. & Decosterd, I. Recombinant adeno-associated virus serotype 6 (rAAV2/6)-mediated gene transfer to nociceptive neurons through different routes of delivery. Mol. Pain 5, 52 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Mogil, J.S. Animal models of pain: progress and challenges. Nat. Rev. Neurosci. 10, 283–294 (2009).

    Article  CAS  PubMed  Google Scholar 

  20. Mason, M.R.J. et al. Comparison of AAV serotypes for gene delivery to dorsal root ganglion neurons. Mol. Ther. 18, 715–724 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bennett, G.J. & Xie, Y.K. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 33, 87–107 (1988).

    Article  CAS  PubMed  Google Scholar 

  22. Lin, J.Y., Knutsen, P.M., Muller, A., Kleinfeld, D. & Tsien, R.Y. ReaChR: a red-shifted variant of channelrhodopsin enables deep transcranial optogenetic excitation. Nat. Neurosci. 16, 1499–1508 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bashkatov, A.N., Genina, E.A., Kochubey, V.I. & Tuchin, V.V. Optical properties of human skin, subcutaneous and mucous tissues in the wavelength range from 400 to 2000 nm. J. Phys. D Appl. Phys. 38, 2543–2555 (2005).

    Article  CAS  Google Scholar 

  24. Yizhar, O. et al. Neocortical excitation/inhibition balance in information processing and social dysfunction. Nature 477, 171–178 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Konermann, S. et al. Optical control of mammalian endogenous transcription and epigenetic states. Nature 500, 472–476 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sohal, V.S., Zhang, F., Yizhar, O. & Deisseroth, K. Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature 459, 698–702 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tillman, D.B., Treede, R.D., Meyer, R.A. & Campbell, J.N. Response of C fibre nociceptors in the anaesthetized monkey to heat stimuli: estimates of receptor depth and threshold. J. Physiol. (Lond.) 485, 753–765 (1995).

    Article  CAS  Google Scholar 

  28. Gao, G.-P. et al. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc. Natl. Acad. Sci. USA 99, 11854–11859 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lee, J.H. et al. Global and local fMRI signals driven by neurons defined optogenetically by type and wiring. Nature 465, 788–792 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kügler, S., Kilic, E. & Bähr, M. Human synapsin 1 gene promoter confers highly neuron-specific long-term transgene expression from an adenoviral vector in the adult rat brain depending on the transduced area. Gene Ther. 10, 337–347 (2003).

    Article  PubMed  Google Scholar 

  31. Gold, M.S. Whole-cell recording in isolated primary sensory neurons. Methods Mol. Biol. 851, 73–97 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Hentschke, H. & Stüttgen, M.C. Computation of measures of effect size for neuroscience data sets. Eur. J. Neurosci. 34, 1887–1894 (2011).

    Article  PubMed  Google Scholar 

  33. Chaplan, S.R., Bach, F.W., Pogrel, J.W., Chung, J.M. & Yaksh, T.L. Quantitative assessment of tactile allodynia in the rat paw. J. Neurosci. Methods 53, 55–63 (1994).

    Article  CAS  PubMed  Google Scholar 

  34. Dixon, W.J. Efficient analysis of experimental observations. Annu. Rev. Pharmacol. Toxicol. 20, 441–462 (1980).

    Article  CAS  PubMed  Google Scholar 

  35. Sommer, C. & Schäfers, M. Painful mononeuropathy in C57BL/Wld mice with delayed Wallerian degeneration: differential effects of cytokine production and nerve regeneration on thermal and mechanical hypersensitivity. Brain Res. 784, 154–162 (1998).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H. Liske, X. Qian, S. Mackey, A. Weitz, D.J. Clark, D.C. Yeomans, P. Sabhaie, C. Gorini, S. Young and H. Scutt for useful discussions and assistance with experiments. This study was supported by the US National Institutes of Health (National Institute of Neurological Disorders and Stroke grant R01-NS080954), the Stanford Bio-X NeuroVentures program and the Stanford Bio-X Interdisciplinary Initiatives program. S.M.I. was supported by an Office of Technology Licensing Stanford Graduate Fellowship and by a Howard Hughes Medical Institute International Student Research Fellowship. K.L.M. was supported by a Bio-X Bioengineering Graduate Fellowship and by a Stanford Interdisciplinary Graduate Fellowship. C.T. was supported by a Swiss National Science Foundation Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

S.M.I., K.L.M., C.T. and S.L.D. designed the experiments. S.M.I. and K.L.M. performed the experiments. C.R. performed cell culture and created vectors. S.Y.L. performed electrophysiology. K.D. contributed reagents and tools. S.M.I., K.L.M. and S.L.D. wrote and edited the paper, with comments from all other authors.

Corresponding author

Correspondence to Scott L Delp.

Ethics declarations

Competing interests

C.T., K.D., and S.L.D. have a financial interest in Circuit Therapeutics, Inc., which, however, did not support this work. S.M.I., K.L.M., C.T., K.D. and S.L.D. have disclosed these findings to the Stanford Office of Technology Licensing for potential use in the identification of new treatments for pain.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 4611 kb)

Supplementary Video 1

Video demonstration of transdermal optogenetic activation of nociceptors. A mouse with bilateral ChR2 expression is placed in a clear cylinder on a transparent glass plate, and allowed to habituate to its environment. When blue light (473 nm, 1 mW/mm2) is shone on the plantar surface of the mouse's skin it immediately withdraws its paw, engages in prolonged licking behavior, and shakes its paw. However, when yellow light (593 nm, 1 mW/mm2) is shone on the skin, there is no observable change in behavior, and the mouse moves around normally. Also, when blue light (473 nm, 1 mW/mm2) is shone on the paw of a bilaterally injected YFP+ mouse, no change in behavior is seen, and the mouse moves around normally. (MOV 8136 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Iyer, S., Montgomery, K., Towne, C. et al. Virally mediated optogenetic excitation and inhibition of pain in freely moving nontransgenic mice. Nat Biotechnol 32, 274–278 (2014). https://doi.org/10.1038/nbt.2834

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.2834

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