Skip to main content
Log in

Serum endocannabinoids and N-acyl ethanolamines and the influence of simulated solar UVR exposure in humans in vivo

  • Published:
Photochemical & Photobiological Sciences Aims and scope Submit manuscript

Abstract

Solar ultraviolet radiation (UVR) exposure of human skin has beneficial and harmful effects on health, including impact on immune function, inflammation and reportedly mood, but these are not fully elucidated. Since the endocannabinoid system is implicated in many activities including mood alteration, our objective was to (i) determine and quantify circulating levels of a wide range of endocannabinoid and N-acyl ethanolamine (NAE) species (ii) evaluate whether these are modulated by cutaneous UVR exposures, as attained through repeated low level summer sunlight exposure. Wearing goggles to prevent eye exposure, 16 healthy volunteers (23–59 y; 10 light skin, phototype II, and 6 dark skin, phototype V) received the same UVR exposures (1.3 SED, 95% UVA/5% UVB) thrice weekly for 6 weeks, whilst casually dressed to expose ∼35% skin surface area. Blood samples were taken at baseline, days 1, 3 and 5 of week one, then at weekly intervals, and analysed by LC-MS/MS. Eleven endocannabinoids and NAEs were detected and quantified at baseline, with N-palmitoyl ethanolamine the most abundant (30% of total). Levels did not vary according to phototype (p > 0.05), except for the NAE docosapentaenoyl ethanolamide, which was higher in phototype II than V (p = 0.0002). Level of the endocannabinoid, 2-AG, was elevated during the UVR exposure course (p < 0.05 vs. baseline for all subjects; p < 0.01 for each phototype group), with maximum levels reached by week 2–3, while NAE species did not significantly alter. These findings suggest differential involvement of the cutaneous endocannabinoid system in low dose solar UVR responses in humans.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. NRPB, Health Effects from Ultraviolet Radiation: Report of an Advisory Group on Non-Ionising Radiation, Documents of the NRPB, 2002, 13, 207–217.

    Google Scholar 

  2. J. K. Robinson, J. Kim, S. Rosenbaum, and S. Ortiz, Indoor tanning knowledge, attitudes, and behavior among young adults from 1988–2007, Arch. Dermatol., 2008, 144, 484–488.

    Google Scholar 

  3. C. R. Harrington, T. C. Beswick, J. Leitenberger, A. Minhajuddin, H. T. Jacobe, and B. Adinoff, Addictive-like behaviours to ultraviolet light among frequent indoor tanners, Clin. Exp. Dermatol., 2011, 36, 33–38.

    Google Scholar 

  4. S. Schneider, K. Diehl, C. Bock, M. M. Schluter, E. W. Breitbart, B. Volkmer, and R. Greinert, Sunbed use, user characteristics, and motivations for tanning: results from the German population-based SUN-Study 2012, JAMA Dermatol., 2013, 149, 43–49.

    Google Scholar 

  5. S. Felton, B. Adinoff, H. Jeon-slaughter, and H. Jacobe, The significant health threat from tanning bed use as a self-treatment for psoriasis, J. Am. Acad. Dermatol., 2016, 74, 1015–1017.

    Google Scholar 

  6. D. B. Carr, B. A. Bullen, G. S. Skinar, M. A. Arnold, M. Rosenblatt, I. Z. Beitins, J. B. Martin, and J. W. McArthur, Physical conditioning facilitates the exercise-induced secretion of beta-endorphin and beta-lipotropin in women, N. Engl. J. Med., 1981, 305, 560–563.

    Google Scholar 

  7. R. A. Houghten, R. W. Swann, and C. H. Li, beta-Endorphin: stability, clearance behavior, and entry into the central nervous system after intravenous injection of the tritiated peptide in rats and rabbits, Proc. Natl. Acad. Sci. U. S. A., 1980, 77, 4588–4591.

    Google Scholar 

  8. P. C. Levins, D. B. Carr, J. E. Fisher, K. Momtaz, and J. A. Parrish, Plasma beta-endorphin and beta-lipoprotein response to ultraviolet radiation, Lancet., 1983, 2, 8342, 166.

    Google Scholar 

  9. P. E. Belon, UVA exposure and pituitary secretion. Variations of human lipotropin concentrations (beta LPH) after UVA exposure, Photochem. Photobiol., 1985, 42, 327–329.

    Google Scholar 

  10. M. Wintzen, D. M. Ostijn, M. C. Polderman, S. le Cessie, J. P. Burbach, and B. J. Vermeer, Total body exposure to ultraviolet radiation does not influence plasma levels of immunoreactive beta-endorphin in man, Photodermatol., Photoimmunol. Photomed., 2001, 17, 256–260.

    Google Scholar 

  11. T. Gambichler, A. Bader, M. Vojvodic, A. Avermaete, M. Schenk, P. Altmeyer, and K. Hoffmann, Plasma levels of opioid peptides after sunbed exposures, Br. J. Dermatol., 2002, 147, 1207–1211.

    Google Scholar 

  12. G. L. Fell, K. C. Robinson, J. Mao, C. J. Woolf, and D. E. Fisher, Skin β-endorphin mediates addiction to UV light, Cell., 2014, 157, 1527–1534.

    Google Scholar 

  13. M. Kaur, A. Liguori, W. Lang, S. R. Rapp, A. B. Fleischer, and S. R. Feldman, Induction of withdrawal-like symptoms in a small randomized controlled trial of opioid blockade in frequent tanners, J. Am. Acad. Dermatol., 2006, 54, 709–711.

    Google Scholar 

  14. P. B. Sparling, A. Giuffrida, D. Piomelli, L. Rosskopf, and A. Dietrich, Exercise activates the endocannabinoid system, NeuroReport., 2003, 14, 2209–2211.

    Google Scholar 

  15. D. A. Raichlen, A. D. Foster, G. L. Gerdeman, A. Seillier, and A. Giuffrida, Wired to run: exercise-induced endocannabinoid signaling in humans and cursorial mammals with implications for the ‘runner’s high’, J. Exp. Biol., 2012, 215, Pt 8, 1331–1336.

    Google Scholar 

  16. E. Heyman, F. X. Gamelin, M. Goekint, F. Piscitelli, B. Roelands, E. Levalir, V. DiMarzo, and R. Meeusen, Intense exercise increases circulating endocannabinoid and BDNF levels in humans-possible implications for reward and depression, Psychoneuroendocrinology., 2012, 37, 844–851.

    Google Scholar 

  17. T. Bíró, B. Tóth, G. Haskó, R. Paus, and P. Pacher, The endocannabinoid system of the skin in health and disease: novel perspectives and therapeutic opportunities, Trends Pharmacol. Sci., 2009, 30, 411–420.

    Google Scholar 

  18. J. van Smeden, L. Hoppel, R. van der Heijden, T. Hankemeier, R. J. Vreeken, and J. A. Bouwstra, LC/MS analysis of stratum corneum lipids: ceramide profiling and discovery, J. Lipid Res., 2011, 52, 1211–1221.

    Google Scholar 

  19. A. Nicolaou, Eicosanoids in skin inflammation, Prostaglandins, Leukotrienes Essent. Fatty Acids., 2013, 88, 131–138.

    Google Scholar 

  20. B. Breiden, and K. Sandhoff, The role of sphingolipid metabolism in cutaneous permeability barrier formation, Biochim. Biophys. Acta., 2014, 1841, 441–452.

    Google Scholar 

  21. A. C. Kendall, S. M. Pilkington, K. A. Massey, G. Sassano, L. E. Rhodes, and A. Nicolaou, Distribution of bioactive lipid mediators in human skin, J. Invest. Dermatol., 2015, 135, 1510–1520.

    Google Scholar 

  22. S. M. Pilkington, L. E. Rhodes, N. M. Al-Aasswad, K. A. Massey, and A. Nicolaou, Impact of EPA ingestion on COX- and LOX-mediated eicosanoid synthesis in skin with and without a pro-inflammatory UVR challenge − report of a randomised controlled study in humans, Mol. Nutr. Food Res., 2014, 58, 580–590.

    Google Scholar 

  23. A. C. Kendall, and A. Nicolaou, Bioactive lipid mediators in skin inflammation and immunity, Prog. Lipid Res., 2013, 52, 141–164.

    Google Scholar 

  24. R. Mechoulam, E. Fride, and V. Di Marzo, Endocannabinoids, Eur. J. Pharmacol., 1998, 359, 1–18.

    Google Scholar 

  25. R. Mechoulam, and L. Hanus, A historical overview of chemical research on cannabinoids, Chem. Phys. Lipids., 2000, 108, 1–13.

    Google Scholar 

  26. T. Bisogno, Endogenous cannabinoids: structure and metabolism, J. Neuroendocrinol., 2008, 20, Suppl 1, 1–9.

    Google Scholar 

  27. L. A. Matsuda, S. J. Lolait, M. J. Brownstein, A. C. Young, and T. I. Bonner, Structure of a cannabinoid receptor and functional expression of the cloned cDNA, Nature., 1990, 346, 561–564.

    Google Scholar 

  28. S. Munro, K. L. Thomas, and M. Abu-shaar, Molecular characterization of a peripheral receptor for cannabinoids, Nature., 1993, 365, 61–65.

    Google Scholar 

  29. A. C. Howlett, The cannabinoid receptors, Prostaglandins Other Lipid Mediators., 2002, 68–69, 619–631.

    Google Scholar 

  30. J. Guindon, and A. G. Hohmann, The endocannabinoid system and pain, CNS Neurol. Disord.: Drug Targets., 2009, 8, 403–421.

    Google Scholar 

  31. E. Boorman, Z. Zajkowska, R. Ahmed, C. M. Pariante, and P. A. Zunszain, Crosstalk between endocannabinoid and immune systems: a potential dysregulation in depression?, Psychopharmacology., 2016, 233, 1591–1604.

    Google Scholar 

  32. H. S. Hansen, and A. Artmann, Endocannabinoids and nutrition, J. Neuroendocrinol., 2008, 20, 94–99.

    Google Scholar 

  33. N. Ghafouri, B. Ghafouri, M. V. Turkina, L. Karlsson, C. J. Fowler, and B. Gerdle, High levels of N-palmitoylethanolamide and N-stearoylethanolamide in microdialysate from myalgic trapezius muscle in women, PLoS One., 2011, 6, e27257.

    Google Scholar 

  34. M. Karsak, E. Gaffal, R. Date, L. Wang-Eckhardt, J. Rehnelt, S. Petrosino, K. Starowicz, R. Steuder, E. Schlicker, B. Cravatt, R. Mechoulam, R. Buettner, S. Werner, V. Di Marzo, T. Tüting, and A. Zimmer, Attenuation of allergic contact dermatitis through the endocannabinoid system, Science., 2007, 316, 1494–1497.

    Google Scholar 

  35. D. Zheng, A. M. Bode, Q. Zhao, Y. Y. Cho, W. Y. Ma, and Z. Dong, The cannabinoid receptors are required for ultraviolet-induced inflammation and skin cancer development, Cancer Res., 2008, 68, 3992–3998.

    Google Scholar 

  36. S. J. Felton, M. S. Cooke, R. Kift, J. L. Berry, A. R. Webb, P. M. Lam, F. R. de Gruijl, A. Vail, and L. E. Rhodes, Concurrent beneficial (vitamin D production) and hazardous (cutaneous DNA damage) impact of repeated low-level summer sunlight exposures, Br. J. Dermatol., 2016, 175, 1320–1328.

    Google Scholar 

  37. T. B. Fitzpatrick, The validity and practicality of sun-reactive skin types I through VI, Arch. Dermatol., 1988, 124, 869–871.

    Google Scholar 

  38. L. E. Rhodes, A. R. Webb, H. I. Fraser, R. Kift, M. T. Durkin, D. Allan, S. J. O’Brien, A. Vail, and J. L. Berry, Recommended summer sunlight exposure levels can produce sufficient (> or =20 ng ml(-1)) but not the proposed optimal (> or =32 ng ml(-1)) 25(OH)D levels at UK latitudes, J. Invest. Dermatol., 2010, 130, 1411–1418.

    Google Scholar 

  39. B. L. Diffey, C. T. Jansén, F. Urbach, and H. C. Wulf, The standard erythema dose: a new photobiological concept, Photodermatol., Photoimmunol. Photomed., 1997, 13, 64–66.

    Google Scholar 

  40. A. R. Webb, R. Kift, J. L. Berry, and L. E. Rhodes, The vitamin D debate: translating controlled experiments into reality for human sun exposure times, Photochem. Photobiol., 2011, 87, 741–745.

    Google Scholar 

  41. M. Feuerecker, D. Hauer, T. Gresset, S. Lassas, I. Kaufmann, M. Vogeser, J. Briegel, C. Hornuss, A. Chouker, and G. Schelling, Effect of an acute consumption of a moderate amount of ethanol on plasma endocannabinoid levels in humans, Alcohol Alcohol., 2012, 47, 226–232.

    Google Scholar 

  42. F. J. Pavón, P. Araos, A. Pastor, M. Calado, M. Pedraz, R. Campos-Cloute, J. J. Ruiz, A. Serrano, E. Blanco, P. Rivera, J. Suárez, M. Romero-Cuevas, M. Pujadas, E. Vergara-Moragues, I. Gornemann, M. Torrens, R. de la Torre, and F. de Rodríguez Fonseca, Evaluation of plasma-free endocannabinoids and their congeners in abstinent cocaine addicts seeking outpatient treatment: impact of psychiatric co-morbidity, Addict. Biol., 2013, 18, 955–969.

    Google Scholar 

  43. P. J. Jones, L. Lin, L. G. Gillingham, H. Yang, and J. M. Omar, Modulation of plasma N-acylethanolamine levels and physiological parameters by dietary fatty acid composition in humans, J. Lipid Res., 2014, 55, 2655–2664.

    Google Scholar 

  44. J. A. Lovegrove, S. S. Lovegrove, S. V. Lesauvage, L. M. Brady, N. Saini, A. M. Minihane, and C. M. Williams, Moderate fish-oil supplementation reverses low-platelet, long-chain n-3 polyunsaturated fatty acid status and reduced plasma triacylglyceral concentration in British Indo-Asians, Am. J. Clin. Nutr., 2004, 79, 974–982.

    Google Scholar 

  45. M. Manav, J. Su, K. Hughes, H. P. Lee, and C. N. Ong, Omega-3 fatty acids and selenium as coronary heart disease risk modifying factors in Asian Indian and Chinese males, Nutrition., 2004, 20, 967–973.

    Google Scholar 

  46. C. Turcotte, F. Chouinard, J. S. Lefebvre, and N. Flamand, Regulation of inflammation by cannabinoids, the endocannabinoids 2-arachidonoyl-glycerol and arachidonoyl-ethanolamide, and their metabolites, J. Leukocyte Biol., 2015, 97, 6, 1049–1070.

    Google Scholar 

  47. C. J. Carsberg, J. Ohanian, and P. S. Friedmann, Ultraviolet radiation stimulates a biphasic pattern of 1,2-diacylglycerol formation in cultured human melanocytes and keratinocytes by activation of phospholipases C and D, Biochem. J., 1995, 305, Pt 2, 471–477.

    Google Scholar 

  48. H. Hall, E. J. Williams, S. E. Moore, F. S. Walsh, A. Prochiantz, and P. Doherty, Inhibition of FGF-stimulated phosphatidylinositol hydrolysis and neurite outgrowth by a cell-membrane permeable phosphopeptide, Curr. Biol., 1996, 6, 5, 580–587.

    Google Scholar 

  49. P. Urquhart, A. Nicolaou, and D. F. Woodward, Endocannabinoids and their oxygenation by cyclo-oxygenases, lipoxygenses and other oxygenase, Biochim. Biophys. Acta., 2015, 1851, 366–376.

    Google Scholar 

  50. A. C. Kendall, S. M. Pilkington, G. Sassano, L. E. Rhodes, and A. Nicolaou, N-Acyl ethanolamide and eicosanoid involvement in irritant dermatitis, Br. J. Dermatol., 2016, 175, 1, 163–171.

    Google Scholar 

  51. S. Magina, M. A. Vieira-Coelho, E. Moura, M. P. Serrão, F. Piscitelli, D. Moura, and V. DiMarzo, Effect of narrowband ultraviolet B treatment on endocannabinoid plasma levels in psoriasis patients, Br. J. Dermatol., 2014, 171, 198–201.

    Google Scholar 

  52. J. D. Wilkinson, and E. M. Williamson, Cannabinoids inhibit human keratinocyte proliferation through a non-CB1/CB2 mechanism and have a potential therapeutic value in the treatment of psoriasis, J. Dermatol. Sci., 2007, 45, 87–92.

    Google Scholar 

  53. K. Sugawara, T. Biro, D. Tsuruta, B. I. Toth, A. Kromminha, N. Zakany, A. Zimmer, W. Funk, B. F. Gibbs, A. Zimmer, and R. Paus, Endocannabinoids limit excessive mast cell maturation and activation in human skin, J. Allergy Clin. Immunol., 2012, 129, 726–738.

    Google Scholar 

  54. F. Montecucco, and V. Di Marzo, At the heart of the matter: the endocannabinoid system in cardiovascular function and dysfunction, Trends Pharmacol. Sci., 2012, 33, 331–340.

    Google Scholar 

  55. B. Horváth, P. Mukhopadhyay, G. Haskó, and P. Pacher, The endocannabinoid system and plant-derived cannabinoids in diabetes and diabetic complications, Am. J. Pathol., 2012, 180, 432–442.

    Google Scholar 

  56. D. M. Sommer, S. Jenisch, M. Suchan, E. Christophers, and M. Weichenthal, Increased prevalence of the metabolic syndrome in patients with moderate to severe psoriasis, Arch. Dermatol. Res., 2006, 298, 321–328.

    Google Scholar 

  57. M. N. Hill, E. J. Carrier, R. J. McLaughlin, A. C. Morrish, S. E. Meier, C. J. Hillard, and B. B. Gorzalka, Regional alterations in the endocannabinoid system in an animal model of depression: effects of concurrent antidepressant treatment, J. Neurochem., 2008, 106, 2322–2336.

    Google Scholar 

  58. P. Adamczyk, A. Gołda, A. C. McCreary, M. Filip, and E. Przegalinski, Activation of endocannabinoid transmission induces antidepressant-like effects in rats, J. Physiol. Pharmacol., 2008, 59, 217–228.

    Google Scholar 

  59. M. N. Hill, G. E. Miller, W. S. Ho, B. B. Gorzalka, and C. J. Hillard, Serum endocannabinoid content is altered in females with depressive disorders: a preliminary report, Pharmacopsychiatry., 2008, 41, 48–53.

    Google Scholar 

  60. M. B. Hill, G. E. Miller, E. J. Carrier, B. B. Gorzalka, and C. J. Hillard, Circulating endocannabinoids and N-acyl ethanolamines are differentially regulated in major depression and following exposure to social stress, Psychonueroendocrinology., 2009, 34, 1257–1262.

    Google Scholar 

  61. C. R. Lupica, and A. C. Riegel, Endocannabinoid release from midbrain dopamine neurons: a potential substrate for cannabinoid receptor antagonist treatment of addiction, Neuropharmacology., 2005, 48, 1105–1116.

    Google Scholar 

  62. J. Fuss, J. Steinle, L. Bindila, M. K. Auer, H. Kircherr, B. Lutz, and P. Gass, A runner’s high depends on cannabinoid receptors in mice, Proc. Natl. Acad. Sci. U. S. A., 2015, 112, 13105–13108.

    Google Scholar 

  63. T. Schwarz, and A. Schwarz, Molecular mechanisms of ultraviolet radiation-induced immunosuppression, Eur. J. Cell Biol., 2011, 90, 560–564.

    Google Scholar 

  64. D. D. Taub, P. Proost, W. J. Murphy, M. Anver, D. L. Longo, J. van Damme, and J. J. Oppenheim, Monocyte chemotactic protein-1 (MCP-1), -2, and -3 are chemotactic for human T lymphocytes, J. Clin. Invest., 1995, 95, 1370–1376.

    Google Scholar 

Download references

Acknowledgments

The authors acknowledge the assistance of Marie Durkin in volunteer recruitment.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lesley E. Rhodes.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Felton, S.J., Kendall, A.C., Almaedani, A.F.M. et al. Serum endocannabinoids and N-acyl ethanolamines and the influence of simulated solar UVR exposure in humans in vivo. Photochem Photobiol Sci 16, 564–574 (2017). https://doi.org/10.1039/c6pp00337k

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1039/c6pp00337k

Navigation