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:

Ptchd1 mediates opioid tolerance via cholesterol-dependent effects on μ-opioid receptor trafficking

Abstract

Repeated exposure to opioids causes tolerance, which limits their analgesic utility and contributes to overdose and abuse liability. However, the molecular mechanisms underpinning tolerance are not well understood. Here, we used a forward genetic screen in Caenorhabditis elegans for unbiased identification of genes regulating opioid tolerance which revealed a role for PTR-25/Ptchd1. We found that PTR-25/Ptchd1 controls μ-opioid receptor trafficking and that these effects were mediated by the ability of PTR-25/Ptchd1 to control membrane cholesterol content. Electrophysiological studies showed that loss of Ptchd1 in mice reduced opioid-induced desensitization of neurons in several brain regions and the peripheral nervous system. Mice and C. elegans lacking Ptchd1/PTR-25 display similarly augmented responses to opioids. Ptchd1 knockout mice fail to develop analgesic tolerance and have greatly diminished somatic withdrawal. Thus, we propose that Ptchd1 plays an evolutionarily conserved role in protecting the μ-opioid receptor against overstimulation.

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: Forward genetic screen in C. elegans identifies PTR-25/Ptchd1 as a regulator of opioid tolerance.
Fig. 2: Knockout of Ptchd1 in mice enhances opioid efficacy and eliminates tolerance.
Fig. 3: Ptchd1 regulates opioid efficacy and response desensitization.
Fig. 4: Molecular mechanism of Ptchd1 action in MOR regulation.
Fig. 5: Role of cholesterol in mediating the effects of Ptchd1 on MOR desensitization in an endogenous neuronal setting.
Fig. 6: Proposed mechanism for Ptchd1 regulation of MOR signaling and opioid tolerance.

Similar content being viewed by others

Data availability

The data that support the findings of this study are published online as Source data files for all display items and are provided with this paper. Additional information is available from the corresponding authors upon reasonable request.

Code availability

Multi-Worm Tracker is an automated behavioral tracking instrument that uses custom software. Additional information about custom software is available from the corresponding authors (B.G.) upon reasonable request.

References

  1. Gainetdinov, R. R., Premont, R. T., Bohn, L. M., Lefkowitz, R. J. & Caron, M. G. Desensitization of G protein-coupled receptors and neuronal functions. Annu. Rev. Neurosci. 27, 107–144 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Abramow-Newerly, M., Roy, A. A., Nunn, C. & Chidiac, P. RGS proteins have a signalling complex: interactions between RGS proteins and GPCRs, effectors, and auxiliary proteins. Cell Signal. 18, 579–591 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Hilger, D., Masureel, M. & Kobilka, B. K. Structure and dynamics of GPCR signaling complexes. Nat. Struct. Mol. Biol. 25, 4–12 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Collett, B. J. Opioid tolerance: the clinical perspective. Br. J. Anaesth. 81, 58–68 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Martyn, J. A. J., Mao, J. & Bittner, E. A. Opioid tolerance in critical illness. N. Engl. J. Med. 380, 365–378 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Colvin, L. A., Bull, F. & Hales, T. G. Perioperative opioid analgesia—when is enough too much? A review of opioid-induced tolerance and hyperalgesia. Lancet 393, 1558–1568 (2019).

    Article  PubMed  Google Scholar 

  7. Darcq, E. & Kieffer, B. L. Opioid receptors: drivers to addiction? Nat. Rev. Neurosci. 19, 499–514 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Matthes, H. W. et al. Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the μ-opioid-receptor gene. Nature 383, 819–823 (1996).

    Article  CAS  PubMed  Google Scholar 

  9. Williams, J. T. et al. Regulation of μ-opioid receptors: desensitization, phosphorylation, internalization, and tolerance. Pharm. Rev. 65, 223–254 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Corder, G. et al. Loss of μ opioid receptor signaling in nociceptors, but not microglia, abrogates morphine tolerance without disrupting analgesia. Nat. Med. 23, 164–173 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Shenoy, S. K. & Lefkowitz, R. J. β-Arrestin-mediated receptor trafficking and signal transduction. Trends Pharmacol. Sci. 32, 521–533 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bohn, L. M., Gainetdinov, R. R., Lin, F. T., Lefkowitz, R. J. & Caron, M. G. μ-Opioid receptor desensitization by β-arrestin-2 determines morphine tolerance but not dependence. Nature 408, 720–723 (2000).

    Article  CAS  PubMed  Google Scholar 

  13. Gillis, A. et al. Critical assessment of G protein-biased agonism at the μ-opioid receptor. Trends Pharmacol. Sci. 41, 947–959 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Terzi, D., Cao, Y., Agrimaki, I., Martemyanov, K. A. & Zachariou, V. R7BP modulates opiate analgesia and tolerance but not withdrawal. Neuropsychopharmacology 37, 1005–1012 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Wang, Y. et al. Blockade of PDGFR-β activation eliminates morphine analgesic tolerance. Nat. Med. 18, 385–387 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Inoue, M. & Ueda, H. Protein kinase C-mediated acute tolerance to peripheral μ-opioid analgesia in the bradykinin-nociception test in mice. J. Pharmacol. Exp. Ther. 293, 662–669 (2000).

    CAS  PubMed  Google Scholar 

  17. Zhou, J. et al. Molecular mechanisms of opioid tolerance: from opioid receptors to inflammatory mediators (review). Exp. Ther. Med. 22, 1004 (2021).

  18. Wang, D. et al. Genetic behavioral screen identifies an orphan anti-opioid system. Science https://doi.org/10.1126/science.aau2078 (2019).

  19. Minevich, G., Park, D. S., Blankenberg, D., Poole, R. J. & Hobert, O. CloudMap: a cloud-based pipeline for analysis of mutant genome sequences. Genetics 192, 1249–1269 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhong, Y., Gu, L. J., Sun, X. G., Yang, S. H. & Zhang, X. H. Comprehensive analysis of patched domain-containing genes reveals a unique evolutionary pattern. Genet. Mol. Res. 13, 7318–7331 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Tora, D. et al. Cellular functions of the autism risk factor PTCHD1 in mice. J. Neurosci. 37, 11993–12005 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wells, M. F., Wimmer, R. D., Schmitt, L. I., Feng, G. & Halassa, M. M. Thalamic reticular impairment underlies attention deficit in Ptchd1Y/− mice. Nature 532, 58–63 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Noor, A. et al. Disruption at the PTCHD1 locus on Xp22.11 in autism spectrum disorder and intellectual disability. Sci. Transl. Med. 2, 49ra68 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Hawrylycz, M. J. et al. An anatomically comprehensive atlas of the adult human brain transcriptome. Nature 489, 391–399 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ung, D. C. et al. Ptchd1 deficiency induces excitatory synaptic and cognitive dysfunctions in mouse. Mol. Psychiatry 23, 1356–1367 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Matsui, A., Jarvie, B. C., Robinson, B. G., Hentges, S. T. & Williams, J. T. Separate GABA afferents to dopamine neurons mediate acute action of opioids, development of tolerance, and expression of withdrawal. Neuron 82, 1346–1356 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nestler, E. J. Is there a common molecular pathway for addiction? Nat. Neurosci. 8, 1445–1449 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Khomula, E. V., Araldi, D., Bonet, I. J. M. & Levine, J. D. Opioid-induced hyperalgesic priming in single nociceptors. J. Neurosci. 41, 31–46 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Claing, A., Laporte, S. A., Caron, M. G. & Lefkowitz, R. J. Endocytosis of G protein-coupled receptors: roles of G protein-coupled receptor kinases and β-arrestin proteins. Prog. Neurobiol. 66, 61–79 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Li, X., Saha, P., Li, J., Blobel, G. & Pfeffer, S. R. Clues to the mechanism of cholesterol transfer from the structure of NPC1 middle lumenal domain bound to NPC2. Proc. Natl Acad. Sci. USA 113, 10079–10084 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, Y. et al. Structural basis for cholesterol transport-like activity of the hedgehog receptor patched. Cell 175, 1352–1364 e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Qiu, Y., Wang, Y., Law, P. Y., Chen, H. Z. & Loh, H. H. Cholesterol regulates μ-opioid receptor-induced β-arrestin 2 translocation to membrane lipid rafts. Mol. Pharmacol. 80, 210–218 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Barter, P. J., Brandrup-Wognsen, G., Palmer, M. K. & Nicholls, S. J. Effect of statins on HDL-C: a complex process unrelated to changes in LDL-C: analysis of the VOYAGER Database. J. Lipid Res. 51, 1546–1553 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. McTaggart, F. & Jones, P. Effects of statins on high-density lipoproteins: a potential contribution to cardiovascular benefit. Cardiovasc. Drugs Ther. 22, 321–338 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Niu, Z. et al. Melanocortin 4 receptor antagonists attenuates morphine antinociceptive tolerance, astroglial activation and cytokines expression in the spinal cord of rat. Neurosci. Lett. 529, 112–117 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Lamberts, J. T., Rosenthal, L. D., Jutkiewicz, E. M. & Traynor, J. R. Role of the guanine nucleotide binding protein, Gαo, in the development of morphine tolerance and dependence. Psychopharmacol. (Berl.) 235, 71–82 (2018).

    Article  CAS  Google Scholar 

  37. Sanna, M. D., Borgonetti, V. & Galeotti, N. μ Opioid receptor-triggered Notch-1 activation contributes to morphine tolerance: role of neuron–glia communication. Mol. Neurobiol. 57, 331–345 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Toda, N., Kishioka, S., Hatano, Y. & Toda, H. Modulation of opioid actions by nitric oxide signaling. Anesthesiology 110, 166–181 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Vidal, B. et al. An atlas of Caenorhabditis elegans chemoreceptor expression. PLoS Biol. 16, e2004218 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Beets, I. et al. Vasopressin/oxytocin-related signaling regulates gustatory associative learning in C. elegans. Science 338, 543–545 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Fernandez, R. W. et al. Cellular expression and functional roles of all 26 neurotransmitter GPCRs in the C. elegans egg-laying circuit. J. Neurosci. 40, 7475–7488 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Flavell, S. W. et al. Serotonin and the neuropeptide PDF initiate and extend opposing behavioral states in C. elegans. Cell 154, 1023–1035 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Quillinan, N., Lau, E. K., Virk, M., von Zastrow, M. & Williams, J. T. Recovery from μ-opioid receptor desensitization after chronic treatment with morphine and methadone. J. Neurosci. 31, 4434–4443 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Whistler, J. L., Chuang, H. H., Chu, P., Jan, L. Y. & von Zastrow, M. Functional dissociation of μ opioid receptor signaling and endocytosis: implications for the biology of opiate tolerance and addiction. Neuron 23, 737–746 (1999).

    Article  CAS  PubMed  Google Scholar 

  45. Zachariou, V. et al. Essential role for RGS9 in opiate action. Proc. Natl Acad Sci. U.S.A. 100, 13656–13661 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gimpl, G., Burger, K. & Fahrenholz, F. Cholesterol as modulator of receptor function. Biochemistry 36, 10959–10974 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Sejdiu, B. I. & Tieleman, D. P. Lipid-protein interactions are a unique property and defining feature of G protein-coupled receptors. Biophys. J. 118, 1887–1900 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Manglik, A. et al. Crystal structure of the μ-opioid receptor bound to a morphinan antagonist. Nature 485, 321–326 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zheng, H. et al. Palmitoylation and membrane cholesterol stabilize μ-opioid receptor homodimerization and G protein coupling. BMC Cell Biol. 13, 6 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zheng, H. et al. Cholesterol level influences opioid signaling in cell models and analgesia in mice and humans. J. Lipid Res. 53, 1153–1162 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Mansouri, M. T., Khodayar, M. J., Tabatabaee, A., Ghorbanzadeh, B. & Naghizadeh, B. Modulation of morphine antinociceptive tolerance and physical dependence by co-administration of simvastatin. Pharmacol. Biochem. Behav. 137, 38–43 (2015).

    Article  CAS  PubMed  Google Scholar 

  52. Pedersen, L. B., Mogensen, J. B. & Christensen, S. T. Endocytic control of cellular signaling at the primary cilium. Trends Biochem. Sci. 41, 784–797 (2016).

    Article  CAS  PubMed  Google Scholar 

  53. Qi, X. & Li, X. Mechanistic insights into the generation and transduction of hedgehog signaling. Trends Biochem. Sci. 45, 397–410 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kowatsch, C., Woolley, R. E., Kinnebrew, M., Rohatgi, R. & Siebold, C. Structures of vertebrate Patched and Smoothened reveal intimate links between cholesterol and Hedgehog signalling. Curr. Opin. Struct. Biol. 57, 204–214 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Giles, A. C. et al. A complex containing the O-GlcNAc transferase OGT-1 and the ubiquitin ligase EEL-1 regulates GABA neuron function. J. Biol. Chem. 294, 6843–6856 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Erbs, E. et al. A mu–delta opioid receptor brain atlas reveals neuronal co-occurrence in subcortical networks. Brain Struct. Funct. 220, 677–702 (2015).

    Article  CAS  PubMed  Google Scholar 

  57. Haley, T. J. & McCormick, W. G. Pharmacological effects produced by intracerebral injection of drugs in the conscious mouse. Br. J. Pharm. Chemother. 12, 12–15 (1957).

    Article  CAS  Google Scholar 

  58. Perner, C. & Sokol, C. L. Protocol for dissection and culture of murine dorsal root ganglia neurons to study neuropeptide release. STAR Protoc. 2, 100333 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Petruska, J. C., Napaporn, J., Johnson, R. D., Gu, J. G. & Cooper, B. Y. Subclassified acutely dissociated cells of rat DRG: histochemistry and patterns of capsaicin-, proton-, and ATP-activated currents. J. Neurophysiol. 84, 2365–2379 (2000).

    Article  CAS  PubMed  Google Scholar 

  60. Oner, S. S., Blumer, J. B. & Lanier, S. M. Group II activators of G-protein signaling: monitoring the interaction of Gα with the G-protein regulatory motif in the intact cell. Methods Enzymol. 522, 153–167 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Liu, S. L. et al. Orthogonal lipid sensors identify transbilayer asymmetry of plasma membrane cholesterol. Nat. Chem. Biol. 13, 268–274 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank N. Martemyanova for producing and maintaining mice examined in this study and members of the Grill and Martemyanov laboratories for valuable input. This work was supported by NIH grants no. DA040406 and no. DA048036 (to B.G. and K.A.M.), no. DA036596 and no. EY028033 (to K.A.M.), and no. DA047771 (to H.M.S.). B.G.’s work is made possible in part by a generous gift in honor of Timothy Jackson. We thank the University of Kansas Genome Sequencing Core, which is supported by an NIH Center of Biomedical Research Excellence Grant (no. GM103638).

Author information

Authors and Affiliations

Authors

Contributions

N.M., D.W., C.K., H.M.S., M.D., O.K.S. and A.C.G. performed experiments. B.G. and K.A.M. conceived the project, oversaw implementation and wrote the manuscript. All authors participated in editing the manuscript.

Corresponding authors

Correspondence to Brock Grill or Kirill A. Martemyanov.

Ethics declarations

Competing interests

B.G. and K.A.M. have filed a patent on the utility of Ptchd1 and modulation of neuronal cholesterol for therapeutic purposes. They are also stockholders in Evodenovo Inc., which has financial interests in intellectual property associated with this study.

Peer review

Peer review information

Nature Neuroscience thanks Gregory Scherrer and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 TgMOR; bgg10 mutants show impaired tolerance to fentanyl and morphine.

a, Time course showing animal speed after application of different concentrations of fentanyl (arrow) for tgMOR animals at 1st exposure (naïve animals). b, Time course of fentanyl doses for tgMOR after repeated (5x) exposure to negative control vehicle. c, Time course of fentanyl doses for tgMOR after repeated (5x) exposure to fentanyl (10 μM). d, Time course of fentanyl doses for tgMOR; bgg10 mutants at 1st exposure (naïve animals). e, Time course of fentanyl doses for tgMOR; bgg10 after repeated (5x) exposure to vehicle. f, Time course of fentanyl doses for tgMOR; bgg10 after repeated (5x) exposure to fentanyl (10 μM). g, Dose-response curves showing tgMOR animals develop tolerance after repeated (5x) exposure to morphine (300 μM). h, Quantitation of opioid efficacy (EC50 values from g) shows decreased morphine efficacy and development of tolerance by tgMOR animals after repeated (5x) exposure to morphine. For a-h, automated MWT was used to acquire all data. Plotted is mean data for n = 12 wells (4-5 animals/well) for all conditions and genotypes. Data for 12 wells were derived from 3 independent experiments. For g and h, data points and histograms are means. n = 3 independent experiments with each experiment derived from 4 wells (4-5 animals/well) per dose. For g, error bars are ± SEM. For h, error bars are ± SD and data were analyzed using one-way ANOVA and Bonferroni’s post hoc test. ***p < 0.001.

Source data

Extended Data Fig. 2 RSBP-1/R7BP and FRPR-13 show differential effects on opioid tolerance.

a, Dose-response curves showing tgMOR; rsbp-1 mutants do not develop tolerance after repeated (5x) exposure to fentanyl (10 μM). b, Quantitation of opioid efficacy (EC50 values from a) shows similar fentanyl efficacy and absence of tolerance in tgMOR; rsbp-1 mutants after repeated (5x) exposure to fentanyl. c, Dose-response curves showing tgMOR; frpr-13 mutants develop tolerance after repeated (5x) exposure to fentanyl (10 μM). d, Quantitation of opioid efficacy (EC50 values from c) shows decreased fentanyl efficacy and development of tolerance by tgMOR; frpr-13 mutants after repeated (5x) exposure to fentanyl. For a-d, data points and histograms are means ± SD. n = 3 independent experiments with each experiment derived from 4 wells (4-5 animals/well) per dose for all genotypes. For b and d, data were analyzed using one-way ANOVA and Bonferroni’s post hoc test. ***p < 0.001.

Source data

Extended Data Fig. 3 Altered opioid responsiveness of tgMOR; bgg10.

a, Representative traces of individual animal movement acquired using MWT after application of fentanyl (10 μM) for indicated time and genotypes. b, Time course of fentanyl response (10 μM, arrow) for indicated genotypes. Arrow denotes fentanyl application. Note only tgMOR and tgMOR; bgg10 animals respond to fentanyl. TgMOR; bgg10 mutants are hypersensitive reaching maximum paralysis after 45 minutes in fentanyl. For b, plotted is mean data for n = 12 wells (4–5 animals/well) for all genotypes. Data for 12 wells were derived from three independent experiments. Data were analyzed using two-way ANOVA and Bonferroni’s post hoc test. ***p < 0.001.

Source data

Extended Data Fig. 4 Mutations in tgMOR; bgg10 occur in PTR-25/F43D9.1, a conserved member of the Patched protein family.

a, Plot showing analysis of whole genome sequence data with mapped region of chromosome III (red and grey bars) containing bgg10. b, Gene diagram showing Q1032stop nonsense mutation (red arrow) in ptr-25 (F43D9.1) contained in mapped region from tgMOR; bgg10 mutants. c, PTR-25 protein with Sterol Sensing Domain (SSD, yellow), Patched family domain (blue) and arginine rich domain (grey). bgg10 mutation, Q1032stop, highlighted (red arrow). d, Phylogenetic analysis of Patched family proteins including all major subfamilies: PTC/PTCH, NCR/NPC, DISP/PTCHD2, PTCHD1/4 (shaded in green) and the PTR group which is heavily expanded in C. elegans. Note PTR-25 has closest homology to mammalian PTCHD1/4. Protein sequences from human (black), mouse (light blue), fly (dark blue) and C. elegans (red) were aligned with Clustal W. Phylogenetic tree was constructed using maximum likelihood method with 1000 bootstrap replicates.

Extended Data Fig. 5 Validation of PTR-25/PTCHD1 as causal gene mutated in tgMOR; bgg10 animals that results in opioid hypersensitivity.

a, Time course of fentanyl application for indicated genotypes. Arrow indicates application of fentanyl (10 μM). TgMOR; ptr-25 CRISPR animals phenocopied hypersensitivity in tgMOR; bgg10 mutants. b, Native rescue with PTR-25 restored opioid sensitivity of tgMOR; bgg10 animals. c, Pan-neuronal rescue (rab-3 promoter) with PTR-25 reversed opioid sensitivity of tgMOR; bgg10 mutants. d, Time course of fentanyl application (5 μM, arrow) for indicated genotypes. Note transgenic expression of human PTCHD1 using the native ptr-25 promoter rescues opioid hypersensitivity of tgMOR; bgg10 animals. e, Dose-response curves with fentanyl showing transgenic human PTCHD1 decreased opioid sensitivity of tgMOR; bgg10 animals. For a-d, plotted is mean data for n = 12 wells (4-5 animals/well) for all genotypes. For e, n = 3 independent experiments with each experiment derived from 4 wells (4-5 animals/well) per dose for all genotypes. Data points are means ± SD. For a-d, data were analyzed by two-way ANOVA with Bonferroni’s post hoc test. ***p < 0.001, * p < 0.05.

Source data

Extended Data Fig. 6 Behavioral characterization of opioid responsiveness of Ptchd1 KO mice.

a, Evaluation of activity levels by tracking distance traveled in an open field. Data are from 7 WT and 6 Ptchd1 KO mice. b, Evaluation of activity levels by calculating total distance traveled in an open field over 120 minutes (from panel a). Data are from 7 WT and 6 Ptchd1 KO mice. c, Evaluation of activity levels by tracking distance traveled in an open field in first 10 minutes and last 10 minutes from panel a. Data are from 7 WT and 6 Ptchd1 KO mice. d, Evaluation of activity levels by tracking distance traveled in an open field after naltrexone or saline injection. Data are from 7 WT and 6 Ptchd1 KO mice. e, Analysis of time course of morphine analgesia by hot plate assay upon systemic morphine administration. Data are from 7 WT and 6 Ptchd1 KO mice. f, Evaluation of morphine induced analgesia in hot plate assay upon intracerebroventricular (ICV) injection of morphine. Data are from 7 male mice per genotype. g, Analysis of time course of morphine analgesia in the hot plate assay following ICV injection of morphine. Data are from 7 male mice per genotype. h, Evaluation of analgesia by hot plate assay with systemic fentanyl administration. Data are from 7 male mice per genotype. i, Analysis of time course of fentanyl analgesia by hot plate assay from the experiment in panel d. j, Evaluation of chronic analgesic tolerance by hot plate assay. Data are from 7 male mice per genotype. k, Quantification of analgesic efficacy reduction as a difference in the MPE between session 1 and 10 from panel j. Data are from 7 male mice per genotype. l, Evaluation of opioid induced hyperalgesia by hot plate assay. Baseline response latencies of mice receiving repeated fentanyl injections (in panel j) prior to drug administration. m, Quantification of opioid induced hyperalgesia as a difference in the baseline latencies between session 1 and 10 from panel l. Data are from 7 male mice per genotype. n, Evaluation of acute analgesic tolerance by hot plate assay. Mice received repeated morphine injections (20 mg/kg) 120 minutes apart. Data are from 5 WT and 7 Ptchd1 KO mice. o, Quantification of analgesic efficacy reduction as a difference in the MPE between session 1 and 2 from panel n. Data are from 5 WT and 7 Ptchd1 KO mice. p, Evaluation of analgesia by tail immersion assay after intrathecal administration of morphine. Data are from 9 WT and 5 Ptchd1 KO mice. q, Evaluation of analgesic tolerance by tail immersion assay. Mice received repeated intrathecal morphine injections (3.5 μg) over 5 days. Data are from 9 WT and 5 Ptchd1 KO mice. r, Quantification of analgesic efficacy reduction as a difference in the MPE between session 1 and 5 from panel q. Data are from 9 WT and 5 Ptchd1 KO mice. For panel a, d, and q, statistical comparisons were performed by 2-way ANOVA and Šídák post-hoc test. For panel c, e, g, i, j, l, and p, statistical comparisons were performed by 2-way ANOVA and Bonferroni’s post hoc comparison. For panels f, h, and n, statistical comparisons were performed by multiple unpaired two tailed Student’s t-test with Bonferroni’s post hoc. For b, k, m, o, and r, statistical comparisons were performed by unpaired two tailed Student’s t-test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. Mean values with S.E.M. errors are shown. MPE, maximum possible effect.

Source data

Extended Data Fig. 7 Characterization of Ptchd1 – MOR co-expression across the brain.

a, Schematic of Ptchd1:YFP and MOR-mCherry mouse genetic manipulations. In the Ptchd1-YFP mouse, exon 1 of the Ptchd1 gene has been replaced with YFP with a YFP-bovine growth hormone poly-A tail (BGH) cassette. In the MOR-mCherry mouse, mCherry was inserted just before the stop codon on exon 4. For electrophysiology and expression studies, the Ptchd1-YFP mouse was crossed with the MOR-mCherry mouse. All mice imaged were heterozygous for the MOR-mCherry allele and homozygous for the Ptchd1-YFP allele. For electrophysiology experiments, control mice were heterozygous the Ptchd1-YFP allele and the MOR-mCherry allele. Ptchd1 KO mice were homozygous for the Ptchd1-YFP allele and heterozygous for the MOR-mCherry allele. ‘−’ denotes genetically modified allele, ‘+’ is the wild-type allele. b-g, Co-expression of MOR and PTCHD1 in locus coeruleus (b-c), nucleus accumbens (d-e), and thalamus (f-g). Representative immunohistochemistry images of brain sections from Ptchd1−/− /MOR-mCherry−/+ mice showing PTCHD1-YFP and MOR-mCherry co-expression. Scale bar is 250 µm in panel b and 100 µm in d and f. Quantification was performed by counting neurons with identified presence of indicated markers in each region from n = 3 (panel c and e) and n = 4 (panel g) mice per genotype. Mean values with S.E.M. errors are shown in all bar graphs.

Source data

Extended Data Fig. 8 Analysis of MOR co-expression with Ptchd1 and morphological effects of Ptchd1 loss.

a, In situ hybridization in brain sections from WT animals. Slices were processed for the detection of the indicated probes in dorsal root ganglia (DRG), striatum (STR) and ventral tegmental area (VTA) using RNAScope. Neuronal marker NeuN was used to identify neurons. b, Quantifications of regional size and cell density in VTA and NAc between Ptchd1 KO and control genotypes. VTA included 8,071 cells from n = 3 in control and 7,812 cells from n = 3 in Ptchd1 KO mice. NAc included 46,979 cells from n = 3 in control and 41,111 cells from n = 3 in Ptchd1 KO mice. c, Quantification of membrane capacitance from recorded DRG nociceptors (control n = 26 cells, 8 mice; Ptchd1 KO n = 34 cells, 7 mice), VTA GABAergic interneurons (control n = 22 cells, 8 mice; Ptchd1 KO n = 25 cells, 12 mice), VTA dopamine projection neurons (control n = 6 cells, 3 mice; Ptchd1 KO n = 7 cells, 4 mice), and NAc medium spiny neurons (control n = 12 cells, 4 mice; Ptchd1 KO n = 12 cells, 5 mice) between Ptchd1 KO and control genotypes. For panel b and c, the statistical comparisons were performed by unpaired two-tailed student’s t-test. *p < 0.05, n.s. is p > 0.05. Mean values with S.E.M. errors are shown.

Source data

Extended Data Fig. 9 Electrophysiological characterization of opioid responses in Ptchd1 KO neurons.

a, Representative whole-cell current recordings of Ptchd1 KO and control DA neurons illustrating inhibition of sIPSC frequency by application of 10 μM DAMGO. b, Quantification of both baseline sIPSC frequencies and their inhibition in DA neurons upon sequential applications of increasing concentrations of DAMGO. Control n = 6 cells, 3 mice; Ptchd1 KO n = 7 cells, 4 mice. c, Schematic of recordings from medium spiny neurons (MSN) in the NAc. Activation of MOR on MSNs depresses their excitability. d, Quantification of rheobase from baseline, through two applications of 10 μM morphine and recovery. Control n = 8 cells, 4 mice and 3 cells, 3 mice; Ptchd1 KO n = 6 cells, 4 mice and 3 cells, 3 mice for the first and second application, respectively. e, Quantification of 10 μM morphine responses as the difference in rheobase. f, Quantification of desensitization between first and second morphine responses. n = 3 cells from 3 mice. For panel b, the statistical comparison was performed by 2-way ANOVA with Holm-Šídák post-hoc test. For panel e, the statistical comparison was performed by a mixed-model ANOVA with Holm-Šídák post-hoc test. For panel f, the statistical comparison was performed by an unpaired two-tailed student’s t-test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. Mean values with S.E.M. errors are shown.

Source data

Extended Data Fig. 10 Cell biological characterization of Ptchd1 actions.

a, Representative immunofluorescence images examining co-localization of MOR with Na+/K+ ATPase. n = 8 cells from each genotype. b, Quantification of the Ptchd1 effect on co-localization based on Pearson’s correlation coefficient in confocal images of fixed cells. 0x and 2x refer to the amount of DNA transfected, where x = 0.21 µg. n = 12 cells from each genotype. Significance determined using unpaired two tailed Student’s t-test. ***p < 0.001. c, Western blot of HEK293T cells expressing HiBiT-MOR alone or with PTCHD1-flag. d, Schematic of the assay design to study the effect of C. elegans protein PTR-25 on internalization of MOR. Addition of LgBit to cells expressing HiBit-MOR forms a nanoluciferase (nLuc) enzyme. DAMGO treatment causes internalization of the complex and quenching of the luminescence. e, Quantification of DAMGO-induced internalization of HiBiT-MOR in HEK293T cells with or without PTR-25 co-expression. 0, 1x, and 2x refer to the amount of DNA transfected, where x = 0.21 µg. n = 5 biologically independent experiments. Significance determined using one-way ANOVA and Dunnett’s post-hoc. *p < 0.05. f, Schematic of assay design to study the effect of Ptchd1 on internalization of β2-adrenergic receptor (ADRB2). Addition of LgBit to cells expressing HiBit-ADR2B forms a nanoluciferase (nLuc) enzyme. Isoproterenol treatment causes internalization of the complex and quenching of the luminescence. g, Quantification of isoproterenol induced internalization of HiBit-ADRB2 in HEK293T cells with or without PTCHD1 co-expression. 0x, 1x, and 2x refer to the amount of DNA transfected, where x = 0.21 µg. n = 4 biologically independent experiments. Significance determined using one-way ANOVA and Dunnett’s post-hoc. n.s. is p > 0.05. h, Experimental design of β-arrestin recruitment assay to study the effect of Ptchd1 on type 2 vasopressin receptor (VPR2). Addition of arginine vasopressin (AVP) induces recruitment of LgBiT-tagged β-arr2 to VPR2-SmBiT to form a functional nLuc enzyme. i, Time course of Ptchd1 effect on recruitment of β-arr2 to VPR2 determined in n = 5 biologically independent experiments. j, Quantification of the effect of Ptchd1 on the maximum fold-change of β-arr2 recruitment from data in panel i. Significance determined using one-way ANOVA. n.s. is p > 0.05. k, Quantification of the initial rate of β-arr2 recruitment to MOR from data in b. 0x, 1x, and 2x refer to the amount of DNA transfected, where x = 0.21 µg. n = 5 biologically independent experiments. Significance determined using one-way ANOVA and Dunnett’s post-hoc. n.s., p > 0.05. l, Schematic of the assay design of β-arrestin recruitment assay. Addition of DAMGO induces recruitment of LgBiT-tagged β-arr2 to MOR-SmBiT to form a functional nLuc enzyme. m, Modulation of DAMGO mediated- βarr2-LgBiT recruitment to MOR-SmBiT following incubation with 4 mM MBCD or 200 µg / mL cholesterol. n = 7 biologically independent experiments for control and MBCD and n = 6 biologically independent experiments for cholesterol. n, Quantification of the maximum fold change and o, the initial recruitment rate from panel n. Significance was determined using a one-way ANOVA and Dunnett’s post-hoc. ns, p > 0.05, *p < 0.05, ***p < 0.001, ****p < 0.0001. In all graphs mean values with S.E.M. errors are shown.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–3.

Reporting Summary

Supplementary Video 1

Evaluation of baseline nociception of WT mice in the hotplate test.

Supplementary Video 2

Evaluation of baseline nociception of Ptchd1 KO mice in the hotplate test.

Supplementary Video 3

Evaluation of morphine (10 mg kg−1) analgesia of WT mice in the hotplate test.

Supplementary Video 4

Evaluation of morphine (10 mg kg−1) analgesia of Ptchd1 KO mice in the hotplate test.

Source data

Source Data Fig. 1

Raw values and statistical source data.

Source Data Fig. 2

Raw values and statistical source data.

Source Data Fig. 3

Raw values and statistical source data.

Source Data Fig. 4

Raw values and statistical source data.

Source Data Fig. 5

Raw values and statistical source data.

Source Data Extended Data Fig. 1

Raw values and statistical source data.

Source Data Extended Data Fig. 2

Raw values and statistical source data.

Source Data Extended Data Fig. 3

Raw values and statistical source data.

Source Data Extended Data Fig. 5

Raw values and statistical source data.

Source Data Extended Data Fig. 6

Raw values and statistical source data.

Source Data Extended Data Fig. 7

Raw values and statistical source data.

Source Data Extended Data Fig. 8

Raw values and statistical source data.

Source Data Extended Data Fig. 9

Raw values and statistical source data.

Source Data Extended Data Fig. 10

Raw values and statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor 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

Maza, N., Wang, D., Kowalski, C. et al. Ptchd1 mediates opioid tolerance via cholesterol-dependent effects on μ-opioid receptor trafficking. Nat Neurosci 25, 1179–1190 (2022). https://doi.org/10.1038/s41593-022-01135-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-022-01135-0

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