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:

Optogenetic pharmacology for control of native neuronal signaling proteins

Abstract

The optical neuroscience revolution is transforming how we study neural circuits. By providing a precise way to manipulate endogenous neuronal signaling proteins, it also has the potential to transform our understanding of molecular neuroscience. Recent advances in chemical biology have produced light-sensitive compounds that photoregulate a wide variety of proteins underlying signaling between and within neurons. Chemical tools for optopharmacology include caged agonists and antagonists and reversibly photoswitchable ligands. These reagents act on voltage-gated ion channels and neurotransmitter receptors, enabling control of neuronal signaling with a high degree of spatial and temporal precision. By covalently attaching photoswitch molecules to genetically tagged proteins, the newly emerging methodology of optogenetic pharmacology allows biochemically precise control in targeted subsets of neurons. Now that the tools for manipulating endogenous neuronal signaling proteins are available, they can be implemented in vivo to enhance our understanding of the molecular bases of brain function and dysfunctions.

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: Photochemical tools for controlling neural function.
Figure 2: Three methods of photocontrol for mapping mechanisms of brain function.
Figure 3: Optogenetic pharmacology enables photocontrol of ion channels and neurotransmitter receptors in mouse hippocampal neurons.
Figure 4: Expression options for optogenetic pharmacology.

Similar content being viewed by others

References

  1. 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 

  2. Matsuzaki, M. et al. Dendritic spine geometry is critical for AMPA receptor expression in hippocampal CA1 pyramidal neurons. Nat. Neurosci. 4, 1086–1092 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ellis-Davies, G.C.R. Two-photon microscopy for chemical neuroscience. ACS Chem. Neurosci. 2, 185–197 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. Shepherd, G.M.G., Pologruto, T.A. & Svoboda, K. Circuit analysis of experience-dependent plasticity in the developing rat barrel cortex. Neuron 38, 277–289 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Lutz, C. et al. Holographic photolysis of caged neurotransmitters. Nat. Methods 5, 821–827 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Canepari, M., Nelson, L.L., Papageorgiou, G.G., Corrie, J.E.T. & Ogden, D.D. Photochemical and pharmacological evaluation of 7-nitroindolinyl-and 4-methoxy-7-nitroindolinyl-amino acids as novel, fast caged neurotransmitters. J. Neurosci. Methods 112, 29–42 (2001).

    Article  CAS  PubMed  Google Scholar 

  8. Callaway, E.M. & Katz, L.C. Photostimulation using caged glutamate reveals functional circuitry in living brain slices. Proc. Natl. Acad. Sci. USA 90, 7661–7665 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Carter, A.G. & Sabatini, B.L. State-dependent calcium signaling in dendritic spines of striatal medium spiny neurons. Neuron 44, 483–493 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Donato, L. et al. Water-soluble, donor-acceptor biphenyl derivatives in the 2-(o-nitrophenyl)propyl series: highly efficient two-photon uncaging of the neurotransmitter γ-aminobutyric acid at λ = 800nm. Angew. Chem. Int. Ed. Engl. 51, 1840–1843 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rial Verde, E.M., Zayat, L., Etchenique, R. & Yuste, R. Photorelease of GABA with visible light using an inorganic caging group. Front. Neural Circuits 2, 2 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Fino, E. et al. RuBi-glutamate: two-photon and visible-light photoactivation of neurons and dendritic spines. Front. Neural Circuits 3, 2 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Banghart, M.R. & Sabatini, B.L. Photoactivatable neuropeptides for spatiotemporally precise delivery of opioids in neural tissue. Neuron 73, 249–259 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Palma-Cerda, F. et al. New caged neurotransmitter analogs selective for glutamate receptor subtypes based on methoxynitroindoline and nitrophenylethoxycarbonyl caging groups. Neuropharmacology 63, 624–634 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Polosukhina, A. et al. Photochemical restoration of visual responses in blind mice. Neuron 75, 271–282 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. Yue, L. et al. Robust photoregulation of GABAA receptors by allosteric modulation with a propofol analogue. Nature Commun. 3, 1095 (2012).

    Article  Google Scholar 

  21. Volgraf, M. et al. Reversibly caged glutamate: a photochromic agonist of ionotropic glutamate receptors. J. Am. Chem. Soc. 129, 260–261 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Adesnik, H., Nicoll, R.A. & England, P.M. Photoinactivation of native AMPA receptors reveals their real-time trafficking. Neuron 48, 977–985 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. 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 

  24. Banghart, M., Borges, K., Isacoff, E.Y., Trauner, D. & Kramer, R.H. Light-activated ion channels for remote control of neuronal firing. Nat. Neurosci. 7, 1381–1386 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Fortin, D.L. et al. Optogenetic photochemical control of designer K+ channels in mammalian neurons. J. Neurophysiol. 106, 488–496 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sandoz, G., Levitz, J., Kramer, R.H. & Isacoff, E.Y. Optical control of endogenous proteins with a photoswitchable conditional subunit reveals a role for TREK1 in GABAB signaling. Neuron 74, 1005–1014 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Baranauskas, G., Tkatch, T., Nagata, K., Yeh, J.Z. & Surmeier, D.J. Kv3.4 subunits enhance the repolarizing efficiency of Kv3.1 channels in fast-spiking neurons. Nat. Neurosci. 6, 258–266 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Volgraf, M. et al. Allosteric control of an ionotropic glutamate receptor with an optical switch. Nat. Chem. Biol. 2, 47–52 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Szobota, S. et al. Remote control of neuronal activity with a light-gated glutamate receptor. Neuron 54, 535–545 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Wyart, C. et al. Optogenetic dissection of a behavioral module in the vertebrate spinal cord. Nature 461, 407–410 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Caporale, N. et al. LiGluR restores visual responses in rodent models of inherited blindness. Mol. Ther. 19, 1212–1219 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Howe, J.R. Homomeric and heteromeric ion channels formed from the kainate-type subunits GluR6 and KA2 have very small, but different, unitary conductances. J. Neurophysiol. 76, 510–519 (1996).

    Article  CAS  PubMed  Google Scholar 

  33. Feldbauer, K. et al. Channelrhodopsin-2 is a leaky proton pump. Proc. Natl. Acad. Sci. USA 106, 12317–12322 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lester, H.A., Krouse, M.E.M., Nass, M.M.M., Wassermann, N.H. & Erlanger, B.F. A covalently bound photoisomerizable agonist: comparison with reversibly bound agonists at Electrophorus electroplaques. J. Gen. Physiol. 75, 207–232 (1980).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tochitsky, I. et al. Optochemical control of genetically engineered neuronal nicotinic acetylcholine receptors. Nat. Chem. 4, 105–111 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Airan, R.D., Thompson, K.R., Fenno, L.E., Bernstein, H. & Deisseroth, K. Temporally precise in vivo control of intracellular signaling. Nature 458, 1025–1029 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Levitz, J. et al. Optical control of metabotropic glutamate receptors. Nat. Neurosci. (2013).

  38. Schröder-Lang, S. et al. Fast manipulation of cellular cAMP level by light in vivo. Nat. Methods 4, 39–42 (2007).

    Article  PubMed  Google Scholar 

  39. Wu, Y.I. et al. A genetically encoded photoactivatable Rac controls the motility of living cells. Nature 461, 104–108 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Strickland, D. et al. TULIPs: tunable, light-controlled interacting protein tags for cell biology. Nat. Methods 9, 379–384 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, X.X., Chung, H.K., Lam, A.J. & Lin, M.Z. Optical control of protein activity by fluorescent protein domains. Science 338, 810–814 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Müller, K. et al. A red/far-red light-responsive bi-stable toggle switch to control gene expression in mammalian cells. Nucleic Acids Res. 41, e77 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Stuber, G.D., Hnasko, T.S., Britt, J.P., Edwards, R.H. & Bonci, A. Dopaminergic terminals in the nucleus accumbens but not the dorsal striatum co-release glutamate. J. Neurosci. 30, 8229–8233 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Carter, M.E. et al. Tuning arousal with optogenetic modulation of locus coeruleus neurons. Nat. Neurosci. 13, 1526–1533 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Donaldson, Z.R., Nautiyal, K.M., Ahmari, S.E. & Hen, R. Genetic approaches for understanding the role of serotonin receptors in mood and behavior. Curr. Opin. Neurobiol. 23, 399–406 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bartos, M., Vida, I. & Jonas, P. Synaptic mechanisms of synchronized gamma oscillations in inhibitory interneuron networks. Nat. Rev. Neurosci. 8, 45–56 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Adesnik, H., Bruns, W., Taniguchi, H., Huang, Z.J. & Scanziani, M. A neural circuit for spatial summation in visual cortex. Nature 490, 226–231 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Royer, S. et al. Control of timing, rate and bursts of hippocampal place cells by dendritic and somatic inhibition. Nat. Neurosci. 15, 769–775 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Noguchi, J. et al. In vivo two-photon uncaging of glutamate revealing the structure-function relationships of dendritic spines in the neocortex of adult mice. J. Physiol. (Lond.) 589, 2447–2457 (2011).

    Article  CAS  Google Scholar 

  50. Stein, M. et al. Azo-propofols: photochromic potentiators of GABAA receptors. Angew. Chem. Int. Ed. Engl. 51, 10500–10504 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants to R.H.K. from the National Eye Institute and the US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Richard H Kramer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kramer, R., Mourot, A. & Adesnik, H. Optogenetic pharmacology for control of native neuronal signaling proteins. Nat Neurosci 16, 816–823 (2013). https://doi.org/10.1038/nn.3424

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3424

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