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:

Discovery of a NAPE-PLD inhibitor that modulates emotional behavior in mice

Abstract

N-acylethanolamines (NAEs), which include the endocannabinoid anandamide, represent an important family of signaling lipids in the brain. The lack of chemical probes that modulate NAE biosynthesis in living systems hamper the understanding of the biological role of these lipids. Using a high-throughput screen, chemical proteomics and targeted lipidomics, we report here the discovery and characterization of LEI-401 as a CNS-active N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD) inhibitor. LEI-401 reduced NAE levels in neuroblastoma cells and in the brain of freely moving mice, but not in NAPE-PLD KO cells and mice, respectively. LEI-401 activated the hypothalamus–pituitary–adrenal axis and impaired fear extinction, thereby emulating the effect of a cannabinoid CB1 receptor antagonist, which could be reversed by a fatty acid amide hydrolase inhibitor. Our findings highlight the distinctive role of NAPE-PLD in NAE biosynthesis in the brain and suggest the presence of an endogenous NAE tone controlling emotional 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: HTS of a NAPE-PLD activity assay provided potent new inhibitors 26.
Fig. 2: Hit optimization affords nanomolar potent inhibitor LEI-401.
Fig. 3: Photoprobe 10 visualizes target engagement of LEI-401 with NAPE-PLD in situ.
Fig. 4: LEI-401 reduces NAE levels in Neuro-2a cells, but not in NAPE-PLD KO cells.
Fig. 5: LEI-401 decreases AEA in mouse brain.
Fig. 6: LEI-401 activates HPA axis signaling and diminishes fear extinction in vivo, which can be abolished by inhibition of FAAH.

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in this published article (and its Supplementary Information files) or are available from the corresponding author on reasonable request. The mass spectrometry proteomics data (raw data and ISOQuant output tables for proteins groups and peptides) have been deposited in the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org) via the PRIDE partner repository with the dataset identifier PXD017586.

References

  1. Hannun, Y. A. & Obeid, L. M. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat. Rev. Mol. Cell Biol. 9, 139 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Kano, M., Ohno-Shosaku, T., Hashimotodani, Y., Uchigashima, M. & Watanabe, M. Endocannabinoid-mediated control of synaptic transmission. Physiol. Rev. 89, 309–380 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Alger, B. E. & Kim, J. Supply and demand for endocannabinoids. Trends Neurosci. 34, 304–315 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Piomelli, D. & Sasso, O. Peripheral gating of pain signals by endogenous lipid mediators. Nat. Neurosci. 17, 164 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Morena, M., Patel, S., Bains, J. S. & Hill, M. N. Neurobiological interactions between stress and the endocannabinoid system. Neuropsychopharmacology 41, 80 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Lutz, B., Marsicano, G., Maldonado, R. & Hillard, C. J. The endocannabinoid system in guarding against fear, anxiety and stress. Nat. Rev. Neurosci. 16, 705 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pacher, P., Steffens, S., Haskó, G., Schindler, T. H. & Kunos, G. Cardiovascular effects of marijuana and synthetic cannabinoids: the good, the bad, and the ugly. Nat. Rev. Cardiol. 15, 151–166 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Maccarrone, M. Metabolism of the endocannabinoid anandamide: open questions after 25 years. Front. Mol. Neurosci. 10, https://doi.org/10.3389/fnmol.2017.00166 (2017).

  9. Okamoto, Y., Morishita, J., Tsuboi, K., Tonai, T. & Ueda, N. Molecular characterization of a phospholipase D generating anandamide and its congeners. J. Biol. Chem. 279, 5298–5305 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Hussain, Z., Uyama, T., Tsuboi, K. & Ueda, N. Mammalian enzymes responsible for the biosynthesis of N-acylethanolamines. Biochim. Biophys. Acta 1862, 1546–1561 (2017).

    Article  CAS  Google Scholar 

  11. Magotti, P. et al. Structure of human N-acylphosphatidylethanolamine-hydrolyzing phospholipase D: regulation of fatty acid ethanolamide biosynthesis by bile acids. Structure 23, 598–604 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang, J. et al. Functional analysis of the purified anandamide-generating phospholipase D as a member of the metallo-β-lactamase family. J. Biol. Chem. 281, 12325–12335 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Leung, D., Saghatelian, A., Simon, G. M. & Cravatt, B. F. Inactivation of N-acyl phosphatidylethanolamine phospholipase D reveals multiple mechanisms for the biosynthesis of endocannabinoids. Biochemistry 45, 4720–4726 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tsuboi, K. et al. Enzymatic formation of N-acylethanolamines from N-acylethanolamine plasmalogen through N-acylphosphatidylethanolamine-hydrolyzing phospholipase D-dependent and -independent pathways. Biochim. Biophys. Acta 1811, 565–577 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Leishman, E., Mackie, K., Luquet, S. & Bradshaw, H. B. Lipidomics profile of a NAPE-PLD KO mouse provides evidence of a broader role of this enzyme in lipid metabolism in the brain. Biochim. Biophys. Acta 1861, 491–500 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Petersen, G., Pedersen, A. H., Pickering, D. S., Begtrup, M. & Hansen, H. S. Effect of synthetic and natural phospholipids on N-acylphosphatidylethanolamine-hydrolyzing phospholipase D activity. Chem. Phys. Lipids 162, 53–61 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Scott, S. A. et al. Discovery of desketoraloxifene analogues as inhibitors of mammalian, Pseudomonas aeruginosa, and NAPE phospholipase D enzymes. ACS Chem. Biol. 10, 421–432 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Castellani, B. et al. Synthesis and characterization of the first inhibitor of N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD). Chem. Comm. 53, 12814–12817 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Karawajczyk, A., Orrling, K. M., de Vlieger, J. S. B., Rijnders, T. & Tzalis, D. The European Lead Factory: a blueprint for public–private partnerships in early drug discovery. Front. Med. 3, https://doi.org/10.3389/fmed.2016.00075 (2017).

  20. Peppard, J.V., Mehdi, S., Li, Z. & Duguid, M.S. Assay methods for identifying agents that modify the activity of NAPE-PLD or ABH4. US patent WO2008150832A1 (2010).

  21. Fu, J. et al. Food intake regulates oleoylethanolamide formation and degradation in the proximal small intestine. J. Biol. Chem. 282, 1518–1528 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Saghatelian, A., Jessani, N., Joseph, A., Humphrey, M. & Cravatt, B. F. Activity-based probes for the proteomic profiling of metalloproteases. Proc. Natl Acad. Sci. USA 101, 10000–10005 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. van Rooden, E. J. et al. Mapping in vivo target interaction profiles of covalent inhibitors using chemical proteomics with label-free quantification. Nat. Protoc. 13, 752 (2018).

    Article  PubMed  CAS  Google Scholar 

  24. Lin, L. et al. Dietary fatty acids augment tissue levels of N-acylethanolamines in N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD) knockout mice. J. Nutr. Biochem. 62, 134–142 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Hill, M. N., Campolongo, P., Yehuda, R. & Patel, S. Integrating endocannabinoid signaling and cannabinoids into the biology and treatment of posttraumatic stress disorder. Neuropsychopharmacology 43, 80 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Locci, A. & Pinna, G. Stimulation of peroxisome proliferator-activated receptor-α by N-palmitoylethanolamine engages allopregnanolone biosynthesis to modulate emotional behavior. Biol. Psychiatry 85, 1036–1045 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Kathuria, S. et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nat. Med. 9, 76 (2002).

    Article  PubMed  CAS  Google Scholar 

  28. Marsicano, G. et al. The endogenous cannabinoid system controls extinction of aversive memories. Nature 418, 530–534 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Hill, M. N. et al. Endogenous cannabinoid signaling is essential for stress adaptation. Proc. Natl Acad. Sci. USA 107, 9406–9411 (2010).

    Article  CAS  PubMed  Google Scholar 

  30. Gunduz-Cinar, O. et al. Convergent translational evidence of a role for anandamide in amygdala-mediated fear extinction, threat processing and stress-reactivity. Mol. Psychiatry 18, 813–823 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Dincheva, I. et al. FAAH genetic variation enhances fronto-amygdala function in mouse and human. Nat. Commun. 6, 6395 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Morena, M. et al. Upregulation of anandamide hydrolysis in the basolateral complex of amygdala reduces fear memory expression and indices of stress and anxiety. J. Neurosci. 39, 1275–1292 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zimmermann, T. et al. Impaired anandamide/palmitoylethanolamide signaling in hippocampal glutamatergic neurons alters synaptic plasticity, learning, and emotional responses. Neuropsychopharmacology 44, 1377–1388 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Hill, M. N. et al. Suppression of amygdalar endocannabinoid signaling by stress contributes to activation of the hypothalamic–pituitary–adrenal axis. Neuropsychopharmacology 34, 2733 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Bluett, R. J. et al. Central anandamide deficiency predicts stress-induced anxiety: behavioral reversal through endocannabinoid augmentation. Transl. Psychiatry 4, e408–e408 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gray, J. M. et al. Corticotropin-releasing hormone drives anandamide hydrolysis in the amygdala to promote anxiety. J. Neurosci. 35, 3879 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gunduz-Cinar, O., Hill, M. N., McEwen, B. S. & Holmes, A. Amygdala FAAH and anandamide: mediating protection and recovery from stress. Trends Pharmacol. Sci. 34, 637–644 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Tsuboi, K., Uyama, T., Okamoto, Y. & Ueda, N. Endocannabinoids and related N-acylethanolamines: biological activities and metabolism. Inflamm. Regen. 38, 28 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ahn, K. et al. Discovery and characterization of a highly selective FAAH inhibitor that reduces inflammatory pain. Chem. Biol. 16, 411–420 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Fu, J. et al. Oleylethanolamide regulates feeding and body weight through activation of the nuclear receptor PPAR-α. Nature 425, 90 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Solorzano, C. et al. Selective N-acylethanolamine-hydrolyzing acid amidase inhibition reveals a key role for endogenous palmitoylethanolamide in inflammation. Proc. Natl Acad. Sci. USA 106, 20966–20971 (2009).

    Article  CAS  PubMed  Google Scholar 

  42. Cannich, A. et al. CB1 cannabinoid receptors modulate kinase and phosphatase activity during extinction of conditioned fear in mice. Learn. Mem. 11, 625–632 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Suzuki, A. et al. Memory reconsolidation and extinction have distinct temporal and biochemical signatures. J. Neurosci. 24, 4787 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chhatwal, J. P. et al. Functional Interactions between endocannabinoid and CCK neurotransmitter systems may be critical for extinction learning. Neuropsychopharmacology 34, 509–521 (2009).

    Article  CAS  PubMed  Google Scholar 

  45. Mayo, L. M. et al. Elevated anandamide, enhanced recall of fear extinction, and attenuated stress responses following inhibition of fatty acid amide hydrolase: a randomized, controlled experimental medicine trial. Biol. Psychiatry 87, 538–547 (2019).

    Article  PubMed  Google Scholar 

  46. Neumeister, A. et al. Elevated brain cannabinoid CB1 receptor availability in post-traumatic stress disorder: a positron emission tomography study. Mol. Psychiatry 18, 1034–1040 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Geurts, L. et al. Adipose tissue NAPE-PLD controls fat mass development by altering the browning process and gut microbiota. Nat. Commun. 6, 6495 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Jourdan, T. et al. Activation of the Nlrp3 inflammasome in infiltrating macrophages by endocannabinoids mediates beta cell loss in type 2 diabetes. Nat. Med. 19, 1132 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hansen, H. H., Ikonomidou, C., Bittigau, P., Hansen, S. H. & Hansen, H. S. Accumulation of the anandamide precursor and other N-acylethanolamine phospholipids in infant rat models of in vivo necrotic and apoptotic neuronal death. J. Neurochem. 76, 39–46 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Sousa-Valente, J. et al. Inflammation of peripheral tissues and injury to peripheral nerves induce differing effects in the expression of the calcium-sensitive N-arachydonoylethanolamine-synthesizing enzyme and related molecules in rat primary sensory neurons. J. Comp. Neurol. 525, 1778–1796 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. van der Wel, T. et al. A natural substrate-based fluorescence assay for inhibitor screening on diacylglycerol lipase α. J. Lipid Res. 56, 927–935 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Baggelaar, M. P. et al. Development of an activity-based probe and in silico design reveal highly selective inhibitors for diacylglycerol lipase-α in brain. Angew. Chem. Int. Ed. 52, 12081–12085 (2013).

    Article  CAS  Google Scholar 

  53. Navia-Paldanius, D., Savinainen, J. R. & Laitinen, J. T. Biochemical and pharmacological characterization of human α/β-hydrolase domain containing 6 (ABHD6) and 12 (ABHD12). J. Lipid Res. 53, 2413–2424 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Soethoudt, M. et al. Cannabinoid CB2 receptor ligand profiling reveals biased signalling and off-target activity. Nat. Commun. 8, 13958 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. van Esbroeck, A. C. M. et al. Activity-based protein profiling reveals off-target proteins of the FAAH inhibitor BIA 10-2474. Science 356, 1084–1087 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Council, N. R. Guide for the Care and Use of Laboratory Animals 8th edn (The National Academies Press, 2011).

  57. Kilkenny, C., Browne, W., Cuthill, I. C., Emerson, M. & Altman, D. G. Animal research: reporting in vivo experiments: the ARRIVE guidelines. Br. J. Pharmacol. 160, 1577–1579 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Cinar, R. et al. Hybrid inhibitor of peripheral cannabinoid-1 receptors and inducible nitric oxide synthase mitigates liver fibrosis. JCI Insight 1, e87336 (2016).

    Article  PubMed Central  Google Scholar 

  59. Mukhopadhyay, B. et al. Hyperactivation of anandamide synthesis and regulation of cell-cycle progression via cannabinoid type 1 (CB1) receptors in the regenerating liver. Proc. Natl Acad. Sci. USA 108, 6323–6328 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The research leading to these results has received support from the Innovative Medicines Initiative Joint Undertaking under grant agreement no. 115489, resources of which are composed of financial contribution from the European Union’s Seventh Framework Programme (no. FP7/2007-2013) and EFPIA companies’ in kind contribution. M.v.d.S. was supported by a VICI-grant from the Netherlands Organization for Scientific Research and funding from Oncode Institute. Leiden University, Faculty of Science ‘Profiling Programme: Endocannabinoids’ is also acknowledged for financial support to E.D.M., V.K., T.H. and M.v.d.S. This research was also supported by National Institutes of Health National Institute on Drug Abuse (grant nos. R01DA039942 and P30DA0339340), Canadian Institutes of Health Research (grant no. FDN-143329 to M.N.H.) as well as start-up funds from the VCU School of Pharmacy to A.H.L. and the Intramural Research Program of NIAAA/NIH (to P.P., O.G.-C., L.I.C., C.M.D. and A.H.). We kindly acknowledge the Pharmaceutical Sciences division of F. Hoffman-La Roche Ltd for their technical assistance with the drug metabolism and pharmacokinetics experiments.

Author information

Authors and Affiliations

Authors

Contributions

E.D.M., T.H., M.N.H, P.P., A.H.L. and M.v.d.S. conceived the project and designed the experiments. J.W., H.v.d.H. and C.A.A.v.B. performed the HTS study. E.D.M. and I.K. synthesized the compounds. E.D.M., A.C.M.v.E., A.M.F.v.d.G., A.M., T.v.d.W, M.S., M.J., T.J.W. and A.P.A.J., performed biochemical and cellular experiments. A.T.B. and B.I.F. performed the proteomics measurements. V.K. and X.D. performed the in vitro and cellular lipidomics measurements. E.D.M., M.M., R.C., G.N.P., D.O., Z.V.V., J.P., C.M., G.D., J.K.P., A.P.A.J., B.I.F., M.W., U.G. and M.N.H. performed in vivo pharmacokinetics, lipidomics and behavioral studies. O.G.-C., L.I.C., C.M.D. and A.H. designed, performed and analyzed fear extinction behavior. E.D.M., M.W., U.G., B.F.C., M.W.B., H.v.d.H., C.A.A.v.B., P.P., A.H.L. and M.v.d.S analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Mario van der Stelt.

Ethics declarations

Competing interests

E.M., I.K. C.B. and M.v.d.S. are listed as inventors on patent application WO 2019/229250 A1 filed by Leiden University in which inhibitors of NAPE-PLD are disclosed.

Additional information

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

Supplementary information

Supplemental Information

Supplementary Tables 1–7, Figs. 1–21 and Note.

Reporting Summary

Supplementary Dataset 1

Label-free quantification of mock- and hNAPE-PLD-FLAG transfected HEK293T cells treated with DMSO or LEI-401 (10 µM). Three biological replicates per condition. P values were calculated with a Student’s t-test (two-tailed, unpaired) and Benjamini–Hochberg correction (10% FDR).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mock, E.D., Mustafa, M., Gunduz-Cinar, O. et al. Discovery of a NAPE-PLD inhibitor that modulates emotional behavior in mice. Nat Chem Biol 16, 667–675 (2020). https://doi.org/10.1038/s41589-020-0528-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0528-7

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research