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:

Rapid optical control of nociception with an ion-channel photoswitch

Abstract

Local anesthetics effectively suppress pain sensation, but most of these compounds act nonselectively, inhibiting activity of all neurons. Moreover, their actions abate slowly, preventing precise spatial and temporal control of nociception. We developed a photoisomerizable molecule, quaternary ammonium–azobenzene–quaternary ammonium (QAQ), that enables rapid and selective optical control of nociception. QAQ is membrane-impermeant and has no effect on most cells, but it infiltrates pain-sensing neurons through endogenous ion channels that are activated by noxious stimuli, primarily TRPV1. After QAQ accumulates intracellularly, it blocks voltage-gated ion channels in the trans form but not the cis form. QAQ enables reversible optical silencing of mouse nociceptive neuron firing without exogenous gene expression and can serve as a light-sensitive analgesic in rats in vivo. Because intracellular QAQ accumulation is a consequence of nociceptive ion-channel activity, QAQ-mediated photosensitization is a platform for understanding signaling mechanisms in acute and chronic pain.

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: Intracellular QAQ photosensitized voltage-gated ion channels.
Figure 2: Intracellular QAQ as a photoswitchable inhibitor of neuronal activity.
Figure 3: TRPV1 channels and P2X7 receptors as a conduit for QAQ entry into cells.
Figure 4: Photosensitization of intact DRGs recorded with a three-dimensional multielectrode array (MEA).
Figure 5: Photosensitization of spinal cord slices.
Figure 6: QAQ optically regulated nociception in vivo, in live rats.

Similar content being viewed by others

References

  1. Yizhar, O., Fenno, L.E., Davidson, T.J., Mogri, M. & Deisseroth, K. Optogenetics in neural systems. Neuron 71, 9–34 (2011).

    Article  CAS  Google Scholar 

  2. Kramer, R.H., Fortin, D. & Trauner, D. New photochemical tools for controlling neuronal activity. Curr. Opin. Neurobiol. 19, 544–552 (2009).

    Article  CAS  Google Scholar 

  3. Fehrentz, T., Schönberger, M. & Trauner, D. Optochemical genetics. Angew. Chem. Int. Edn. 50, 12156–12182 (2011).

    Article  CAS  Google Scholar 

  4. Fortin, D. et al. Photochemical control of endogenous ion channels and cellular excitability. Nat. Methods 5, 331–338 (2008).

    Article  CAS  Google Scholar 

  5. Banghart, M.R. et al. Photochromic blockers of voltage-gated potassium channels. Angew. Chem. Int. Ed. 48, 9097–9101 (2009).

    Article  CAS  Google Scholar 

  6. Mourot, A. et al. Tuning photochromic ion channel blockers. ACS Chem. Neurosci. 2, 536–543 (2011).

    Article  CAS  Google Scholar 

  7. Basbaum, A.I., Bautista, D.M., Scherrer, G. & Julius, D. Cellular and molecular mechanisms of pain. Cell 139, 267–284 (2009).

    Article  CAS  Google Scholar 

  8. Cavanaugh, D.J. et al. Trpv1 reporter mice reveal highly restricted brain distribution and functional expression in arteriolar smooth muscle cells. J. Neurosci. 31, 5067–5077 (2011).

    Article  CAS  Google Scholar 

  9. Chung, M.-K., Güler, A.D. & Caterina, M.J. TRPV1 shows dynamic ionic selectivity during agonist stimulation. Nat. Neurosci. 11, 555–564 (2008).

    Article  CAS  Google Scholar 

  10. Binshtok, A.M., Bean, B.P. & Woolf, C.J. Inhibition of nociceptors by TRPV1-mediated entry of impermeant sodium channel blockers. Nature 449, 607–610 (2007).

    Article  CAS  Google Scholar 

  11. Binshtok, A.M. et al. Coapplication of lidocaine and the permanently charged sodium channel blocker QX-314 produces a long-lasting nociceptive blockade in rodents. Anesthesiology 111, 127–137 (2009).

    Article  CAS  Google Scholar 

  12. Scholz, A. Mechanisms of (local) anaesthetics on voltage-gated sodium and other ion channels. Br. J. Anaesth. 89, 52–61 (2002).

    Article  CAS  Google Scholar 

  13. Hille, B. Ion Channels of Excitable Membranes 3rd edn. (Sinauer Associates, 2001).

  14. Strichartz, G.R. The inhibition of sodium currents in myelinated nerve by quaternary derivatives of lidocaine. J. Gen. Physiol. 62, 37–57 (1973).

    Article  CAS  Google Scholar 

  15. Kawaguchi, A. et al. Enhancement of sodium current in NG108–15 cells during neural differentiation is mainly due to an increase in NaV1.7 expression. Neurochem. Res. 32, 1469–1475 (2007).

    Article  CAS  Google Scholar 

  16. Rogawski, M.A., Inoue, K., Suzuki, S. & Barker, J.L. A slow calcium-dependent chloride conductance in clonal anterior pituitary cells. J. Neurophysiol. 59, 1854–1870 (1988).

    Article  CAS  Google Scholar 

  17. Hoshi, T., Zagotta, W.N. & Aldrich, R.W. Biophysical and molecular mechanisms of Shaker potassium channel inactivation. Science 250, 533–538 (1990).

    Article  CAS  Google Scholar 

  18. Khakh, B.S., Bao, X.R., Labarca, C. & Lester, H.A. Neuronal P2X transmitter-gated cation channels change their ion selectivity in seconds. Nat. Neurosci. 2, 322–330 (1999).

    Article  CAS  Google Scholar 

  19. Virginio, C., MacKenzie, A., Rassendren, F.A., North, R.A. & Surprenant, A. Pore dilation of neuronal P2X receptor channels. Nat. Neurosci. 2, 315–321 (1999).

    Article  CAS  Google Scholar 

  20. Wang, H. et al. Bradykinin produces pain hypersensitivity by potentiating spinal cord glutamatergic synaptic transmission. J. Neurosci. 25, 7986–7992 (2005).

    Article  CAS  Google Scholar 

  21. Bautista, D.M. et al. TRPA1 mediates the inflammatory actions of environmental irritants and proalgesic agents. Cell 124, 1269–1282 (2006).

    Article  CAS  Google Scholar 

  22. Voets, T. et al. The principle of temperature-dependent gating in cold- and heat-sensitive TRP channels. Nature 430, 748–754 (2004).

    Article  CAS  Google Scholar 

  23. de Castro, F., Silos-Santiago, I., López de Armentia, M., Barbacid, M. & Belmonte, C. Corneal innervation and sensitivity to noxious stimuli in trkA knockout mice. Eur. J. Neurosci. 10, 146–152 (1998).

    Article  CAS  Google Scholar 

  24. Rózsa, A.J. & Beuerman, R.W. Density and organization of free nerve endings in the corneal epithelium of the rabbit. Pain 14, 105–120 (1982).

    Article  Google Scholar 

  25. Wenk, H.N. & Honda, C.N. Silver nitrate cauterization: characterization of a new model of corneal inflammation and hyperalgesia in rat. Pain 105, 393–401 (2003).

    Article  CAS  Google Scholar 

  26. Zhang, F. et al. Multimodal fast optical interrogation of neural circuitry. Nature 446, 633–639 (2007).

    Article  CAS  Google Scholar 

  27. Chow, B.Y. et al. High-performance genetically targetable optical neural silencing by light-driven proton pumps. Nature 463, 98–102 (2010).

    Article  CAS  Google Scholar 

  28. Zhang, F., Aravanis, A.M., Adamantidis, A.R., de Lecea, L. & Deisseroth, K. Circuit-breakers: optical technologies for probing neural signals and systems. Nat. Rev. Neurosci. 8, 577–581 (2007).

    Article  CAS  Google Scholar 

  29. Malin, S.A., Davis, B.M. & Molliver, D.C. Production of dissociated sensory neuron cultures and considerations for their use in studying neuronal function and plasticity. Nat. Protoc. 2, 152–160 (2007).

    Article  CAS  Google Scholar 

  30. McKemy, D.D., Neuhausser, W.M. & Julius, D. Identification of a cold receptor reveals a general role for TRP channels in thermosensation. Nature 416, 52–58 (2002).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M.R. Banghart and M. Kienzler for helping synthesize QAQ, S. Scott for his help with computer programming, D. Bautista for helpful comments, A. Nicke (Max Planck Institute of Brain Research) for the P2X7 receptor clone, A. Blatz (Photoswitch Biosciences, Inc.) for HEK-293 cells stably expressing Cav2.2 and F. Ory for artistic input. This work was supported by US National Institutes of Health grants MH088484 to R.H.K. and PN2 EY018241 to R.H.K. and D.T. (the University of California Berkeley Nanomedicine Development Center) and by the Center for Integrated Protein Science, Munich to D.T.

Author information

Authors and Affiliations

Authors

Contributions

A.M. and R.H.K. wrote the paper. A.M., T.F., Y.L.F., C.M.S., F.N., D.T. and. R.H.K. designed experiments. AM., T.F., Y.L.F., C.M.S. and C.H. performed electrophysiological experiments and analyzed data. A.M. and D.D. performed in vivo experiments.

Corresponding authors

Correspondence to Dirk Trauner or Richard H Kramer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 (PDF 1391 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Mourot, A., Fehrentz, T., Le Feuvre, Y. et al. Rapid optical control of nociception with an ion-channel photoswitch. Nat Methods 9, 396–402 (2012). https://doi.org/10.1038/nmeth.1897

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.1897

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