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.

  • Brief Communication
  • Published:

In vivo photopharmacology with a caged mu opioid receptor agonist drives rapid changes in behavior

Abstract

Photoactivatable drugs and peptides can drive quantitative studies into receptor signaling with high spatiotemporal precision, yet few are compatible with behavioral studies in mammals. We developed CNV-Y-DAMGO—a caged derivative of the mu opioid receptor-selective peptide agonist DAMGO. Photoactivation in the mouse ventral tegmental area produced an opioid-dependent increase in locomotion within seconds of illumination. These results demonstrate the power of in vivo photopharmacology for dynamic studies into animal behavior.

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: Design and validation of CNV-Y-DAMGO in vitro and ex vivo.
Fig. 2: In vivo uncaging with CNV-Y-DAMGO rapidly modulates pain-related behavior and locomotion.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available within the paper and its Extended Data files. Source data are provided with this paper.

References

  1. Ellis-Davies, G. C. R. Caged compounds: photorelease technology for control of cellular chemistry and physiology. Nat. Methods 4, 619–628 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Paoletti, P., Ellis-Davies, G. C. R. & Mourot, A. Optical control of neuronal ion channels and receptors. Nat. Rev. Neurosci. 20, 514–532 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ankenbruck, N., Courtney, T., Naro, Y. & Deiters, A. Optochemical control of biological processes in cells and animals. Angew. Chem. Int. Ed. Engl. 57, 2768–2798 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Font, J. et al. Optical control of pain in vivo with a photoactive mGlu5 receptor negative allosteric modulator. eLife 6, e23545 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  5. López-Cano, M. et al. Remote local photoactivation of morphine produces analgesia without opioid-related adverse effects. Br. J. Pharmacol. https://doi.org/10.1111/bph.15645 (2021).

  6. Cuttoli, R. D. De et al. Optofluidic control of rodent learning using cloaked caged glutamate. Proc. Natl Acad. Sci. USA 117, 6831–6835 (2020).

    Article  Google Scholar 

  7. Taura, J. et al. Remote control of movement disorders using a photoactive adenosine A 2A receptor antagonist. J. Control. Release 283, 135–142 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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. 589, 2447–2457 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gomes, I. et al. Biased signaling by endogenous opioid peptides. Proc. Natl Acad. Sci. USA 117, 11820–11828 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. 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 

  11. Banghart, M. R., He, X. J. & Sabatini, B. L. A caged enkephalin optimized for simultaneously probing mu and delta opioid receptors. ACS Chem. Neurosci. 9, 684–690 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Craves, F. B., Law, P. Y., Hunt, C. A. & Loh, H. H. The metabolic disposition of radiolabeled enkephalins in vitro and in situ. J. Pharmacol. Exp. Ther. 206, 492–506 (1978).

    CAS  PubMed  Google Scholar 

  13. Handa, B. K. et al. Analogues of β-LPH61-64 possessing selective agonist activity at μ-opiate receptors. Eur. J. Pharmacol. 70, 531–540 (1981).

    Article  CAS  PubMed  Google Scholar 

  14. He, X. J. et al. Convergent, functionally independent signaling by mu and delta opioid receptors in hippocampal parvalbumin interneurons. eLife 10, e69746 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Castro, D. C. et al. An endogenous opioid circuit determines state-dependent reward consumption. Nature 598, 646–651 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Manning, B. H., Morgan, M. J. & Franklin, K. B. J. Morphine analgesia in the formalin test: evidence for forebrain and midbrain sites of action. Neuroscience 63, 289–294 (1994).

    Article  CAS  PubMed  Google Scholar 

  17. Johnson, S. W. & North, R. A. Opioids excite dopamine neurons by hyperpolarization of local interneurons. J. Neurosci. 12, 483–488 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Joyce, E. M. & Iversen, S. D. The effect of morphine applied locally to mesencephalic dopamine cell bodies on spontaneous motor activity in the rat. Neurosci. Lett. 14, 207–212 (1979).

    Article  CAS  PubMed  Google Scholar 

  19. Hüll, K., Morstein, J. & Trauner, D. In vivo photopharmacology. Chem. Rev. 118, 10710–10747 (2018).

    Article  PubMed  Google Scholar 

  20. Tochitsky, I., Kienzler, M. A., Isacoff, E. & Kramer, R. H. Restoring vision to the blind with chemical photoswitches. Chem. Rev. 118, 10748–10773 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gomila, A. M. J. et al. Photocontrol of endogenous glycine receptors in vivo. Cell Chem. Biol. 27, 1425–1433.e7 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Donthamsetti, P. et al. Cell specific photoswitchable agonist for reversible control of endogenous dopamine receptors. Nat. Commun. 12, 4775 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Acosta-Ruiz, A. et al. Branched photoswitchable tethered ligands enable ultra-efficient optical control and detection of G protein-coupled receptors in vivo. Neuron 105, 446–463.e13 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Davenport, C. M. et al. Relocation of an extrasynaptic GABAA receptor to inhibitory synapses freezes excitatory synaptic strength and preserves memory. Neuron 109, 123–134.e4 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. Zhang, Y. et al. Battery-free, lightweight, injectable microsystem for in vivo wireless pharmacology and optogenetics. Proc. Natl Acad. Sci. USA 116, 21427–21437 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Frank, J. A. et al. In vivo photopharmacology enabled by multifunctional fibers. ACS Chem. Neurosci. 11, 3802–3813 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Gilbert, D. et al. Caged capsaicins: new tools for the examination of TRPV1 channels in somatosensory neurons. ChemBioChem 8, 89–97 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Tanowitz, M. & von Zastrow, M. A novel endocytic recycling signal that distinguishes the membrane trafficking of naturally occurring opioid receptors. J. Biol. Chem. 278, 45978–45986 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Pologruto, T. A., Sabatini, B. L. & Svoboda, K. ScanImage: flexible software for operating laser scanning microscopes. Biomed. Eng. Online 2, 13 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. Berg for technical support, J. Isaacson for helpful discussions and J. Chang-Weinberg for manuscript editing. This work was supported by the National Institute of Neurological Disorders and Stroke and the National Institute of Mental Health (U01NS113295 to M.R.B., including Diversity Supplements to D.A.J. and A.E.L.), the National Institute of General Medical Sciences (R35GM133802 to M.R.B., T32GM007240 to X.J.H.), the Brain and Behavior Research Foundation (M.R.B.), the Esther A. and Joseph Klingenstein Fund and Simons Foundation (M.R.B.), the Rita Allen Foundation (M.R.B.) and by Plan Nacional Sobre Drogas (Spanish Ministerio de Sandidad, 2021I070 to J.B.).

Author information

Authors and Affiliations

Authors

Contributions

X.M., D.A.J., X.J.H. and M.R.B. designed research. X.M., D.A.J., A.E.L., X.J.H., S.P.M., J.C.Y., A.R. and J.B. performed research. X.M. contributed new reagents or analytical tools. X.M., D.A.J., X.J.H. and M.R.B. analyzed data. M.R.B., X.M. and D.A.J. wrote the paper.

Corresponding author

Correspondence to Matthew R. Banghart.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks James Frank and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Nina Vogt, in collaboration with the Nature Methods team.

Additional information

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

Extended data

Extended Data Fig. 1 Synthesis of CNV-Y-DAMGO (4).

The N-terminal amine in DAMGO (3) was Boc-protected prior to phenol alkylation with 6. Global deprotection with TFA produced CNV-Y-DAMGO (4) in good overall yield.

Extended Data Fig. 2 In vitro characterization of CNV-Y-DAMGO activity at opioid receptors.

(A) Dose-response curves at the delta opioid receptor (DOR) using a GloSensor assay of cAMP signaling in HEK293T cells (n = 5 wells per data point, 1 representative independent experiment shown). Data were normalized to the maximal response to leucine-enkephalin (LE, 1 µM) and are expressed as the mean ± SEM. (B) Same as A, but using the kappa opioid receptor (KOR) and dynorphin A(1-8) (Dyn8, 1 µM) for normalization. (C) Same as A, but using the nociceptin/orphanin FQ receptor (NOP) and nociception/orphanin FQ (N/OFQ, 1 µM) for normalization. (D) Dose-response curves at the mu opioid receptor (MOR) using a NanoBiT-based luminescence complementation assay of β-arrestin signaling in HEK293T cells (n = 4 wells per data point, 3 independent experiments averaged). Data were normalized to the maximal response to DAMGO (10 µM) and are expressed as the mean ± SEM.

Source data

Extended Data Fig. 3 In vitro characterization of CNV-Y-DAMGO (4) photolysis and dark stability.

(A) Waterfall plot of HPLC chromatograms of CNV-Y-DAMGO (0.4 mM) during illumination with 375 nm light (10 mW) in PBS, pH 7.2. (B) Enlarged HPLC chromatogram after 2 min of illumination (top) and mass spectrograms corresponding to the indicated peaks (bottom). (C) HPLC chromatograms of a sample of CNV-Y-DAMGO (1 mM) in PBS left for 24 hours in the dark. (D) Summary of MNI-Glutamate and CNV-Y-DAMGO photouncaging reactions over time, as measured by HPLC (n = 3 samples per condition). Samples were optical density-matched at 375 nm in PBS and illuminated with 375 nm laser irradiation. Data are expressed as the mean ± SEM. (E) UV-VIS spectrum of samples of CNV-Y-DAMGO (0.4 mM) in PBS before and after irradiation with 375 nm light (10 mW) for the indicated time periods.

Source data

Extended Data Fig. 4 DAMGO infusion into the VTA and location of optofluidic cannula placements.

(A) Schematic of the experimental configuration for DAMGO infusion through an optofluidic cannula. (B) Experimental timeline. (C) Movement velocity vs. time in the open field for a cohort of mice after DAMGO (200 µM, 0.5 uL) infusion. (D) Average data from C (n = 6 mice). Data are expressed as the mean ± SEM. (E) Example maps of open field locomotor activity from a single mouse after infusion of either saline or DAMGO. (F) Change in distance traveled (after-before uncaging) mapped to the implant location for the mice shown in Fig. 2g,h.

Source data

Supplementary information

Reporting Summary

Peer Review File

Representative formalin-treated mouse infused with CNV-Y-DAMGO in the NAc and exposed to light: minutes 29–31 postinjection.

Representative formalin-treated control mouse exposed to light: minutes 29–31 postinjection.

Representative mouse infused with CNV-Y-DAMGO in the VTA and exposed to light.

Representative mouse infused with CNV-Y-DAMGO in the VTA and exposed to light after treatment with naloxone.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, X., Johnson, D.A., He, X.J. et al. In vivo photopharmacology with a caged mu opioid receptor agonist drives rapid changes in behavior. Nat Methods 20, 682–685 (2023). https://doi.org/10.1038/s41592-023-01819-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-023-01819-w

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