Next Article in Journal
A New Gelling Agent and Rheology Modifier in Cosmetics: Caesalpinia spinosa Gum
Previous Article in Journal
Known and Unknown Features of Hair Cuticle Structure: A Brief Review
Previous Article in Special Issue
Noninvasive Skin Barrier Assessment: Multiparametric Approach and Pilot Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Epidermal Endocannabinoid System (EES) and its Cosmetic Application

1
Incospharm Corp., 328, Techno-2-ro, Yuseong-gu, Daejeon 34036, Korea
2
Cosfactory Co., Ltd., 87-12, Sandan-ro, Pyeongtaek-si, Gyeonggi-do 17745, Korea
3
Lastella Co., Ltd., BVC-201, KRIBB, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
4
SphingoBrain Inc., 1000 3rd St. Unit 718, San Francisco, CA, 941587, USA
5
Dr. Raymond laboratories, Inc., 560 Sylvan Ave. Englewood cliffs, NJ 07632, USA
*
Author to whom correspondence should be addressed.
Cosmetics 2019, 6(2), 33; https://doi.org/10.3390/cosmetics6020033
Submission received: 23 April 2019 / Revised: 14 May 2019 / Accepted: 14 May 2019 / Published: 16 May 2019
(This article belongs to the Special Issue Skin Barrier Function)

Abstract

:
Recently, cannabis, or its major constituent cannabidiol (CBD), has emerged as an attractive cosmetic ingredient. Initiated as a basic investigation of the physiological roles of cannabinoid receptors and their endogenous ligands, endocannabinoids’ diverse potential benefits have been proposed for using cannabinoid receptor modulating compounds in skin health. Improvement in skin barrier functions, alleviating inflammatory responses, and the relief of itching sensations are some commonly expected therapeutic benefits, which have been supported by many in vitro, in vivo, and clinical studies. While hemp seed oils or hemp extracts might be used for the cosmetic formulation, the potential for contamination with a psychoactive cannabinoid, such as 9-THC, should be carefully checked. Instead of using hemp-derived ingredients, the use of cannabinomimetics, synthetic ligands on cannabinoid receptors, or entourage compounds (which modulate intracellular synthesis and the degradation of endocannabinoids), have been tried. In this review, a brief introduction of the epidermal endocannabinoid system (EES) and its physiological roles will be followed by a review of the cosmetic and dermatologic application of cannabinomimetics and entourage compounds. The practical application of newly developed endocannabinomimetics will be discussed as well.

1. Endocannabinoid and Epidermal Endocannabinoid System (EES)

From the identification of Δ9-tetrahydrocannabinol (Δ9-THC; Figure 1) in the cannabis plant as a bioactive ingredient of psychological activity and the chemical synthesis of its analogues [1], cannabinoid research paved the way to the discovery of the endocannabinoid system (ECS). Along with the endogenous cannabinoid receptor ligands (endocannabinoids), the endocannabinoid system (ECS) consists of cannabinoid receptors and enzymes responsible for the production and degradation of endocannabinoids [2]. Recently, intracellular transport and uptake of endocannabinoids have also been reported as having modulatory activity on ECS tones, which has emerged as a new potential target for ECS activity modulation [3]. Among the diverse endocannabinoids, AEA (anandamide [N-arachidonoylethanolamine]) and 2-AG (2-arachidonoylglycerol) were the first molecules identified, followed by the further identification of a family of N-acyl-ethanolamides (NAEs) and monoacylglycerols (MAGs), respectively (Figure 1) [4]. Two different cannabinoid receptors, cannabinoid receptor-1 (CB1R) and cannabinoid receptor-2 (CB2R), have been identified and cloned from mammalian tissue. Both receptors belong to the seven-transmembrane G-protein-coupled receptor (GPCR), coupled to Gi-Go heterotrimeric G proteins [5]. While CB1R is typically expressed in the central nervous system, including in the brain, the spinal cord, and in peripheral nerve terminals [6], it also exists in immune cells and skin as well [7]. While its exact roles and functions remain controversial, the expression of CB1R and CB2R in cultured keratinocytes and skin have been repeatedly reported [8]. Recently, two orphan-GPCRs, GPR55 and GPR119, have been suggested as cannabinoid receptors. AEA, palmitoylethanolamine (PEA), Virodhamine, and 2-arachidonylethanolamie (2-AG) is identified as binding to GPR55 and oleoylethanolmide (OEA) as a ligand for GPR119 [9], respectively. While the exact roles of these two receptors are not yet clarified, potential involvement in carcinogenesis was previously reported for GPR55 [10,11]. Recent studies using functional metagenomics for gut microbiome resulted in an identification of unique proteins from commensal bacteria, mediating the production of N-acyl-3-hydroxypalmitoyl-glycine (NAHPG). It was postulated that NAHPG can bind to GPR132 and that it mediates the potential effects of gut microbiome on inflammation and immune function. Considering the abundance of skin microbiome and the expression of CB receptors in skin, it might be also postulated that cannabinoid signaling might be involved between the cross-talk of skin microbiome and skin homeostasis [12].

2. Cannabinoid Receptor Modulation: Direct and Indirect Signaling Pathways

As a Gi-coupled GPCR, the binding of an agonistic ligand on a cannabinoid receptor (CBR) inhibits receptor-coupled adenylyl cyclase activity and the cyclic AMP (cAMP) cascade, followed by the stimulation of mitogen-activated protein kinase (MAPK) activity and consequent secondary signaling. Considering the mechanism of Gi protein, the binding of an antagonistic ligand alone is not able to induce secondary cellular signaling. However, changes in cellular environments, such as the availability of the G proteins subunits, may result in the binding of different types of G proteins, such as Gs or G1/11, which could have implications for different types of signaling (reviewed in reference [13]. One type of cellular signaling induced by cannabinoid receptor activation is the stimulation of de novo ceramide synthesis. Pharmacological effects of cannabinoids include immunosuppressive and anti-inflammatory activities against various immune-mediated diseases such as diabetes, allergic asthma, and atopic dermatitis. Among the several proposed modes of action, cannabinoids-induced apoptosis in immune cells through the ceramide signaling was proposed [14]. Previously, increased synthesis of ceramide by activation of serine palmitoyltransferase (SPT), an enzyme responsible for the rate-limiting step of de novo ceramide synthesis, was reported in glioma cells [15], and induction of ceramide synthase (CerS) was also observed in endocannabinoid-analogue treated cells [16]. While there is no report about the direct effects of endocannabinoid on ceramide synthesis in epidermal keratinocyte, a recent study using Echinacea-derived alkylamides, which are putative CB2R ligands [17], showed that topical application of Echinacea-derived alkylamides not only improves atopic eczema symptoms, but also increases overall epidermal lipids contents. This includes epidermal ceramide EOS (esterified ω-hydroxy acyl-sphingosine), which is considered to be one of the most important ceramide subtypes for the epidermal permeability barrier’s function as structural components for stratum corneum intercellular lipids [18]. In addition to the crucial structural roles, increased ceramide can also induce the expression of anti-microbial peptides in epidermis through endoplasmic reticulum-mediated signaling, and further help to improve skin barrier functions, in terms of antimicrobial barriers [19] (Figure 2).
In addition to the expression of both receptors, the whole biochemical machinery of ECS, including cellular transport and intracellular uptake, synthesizing, and metabolizing endocannabinoids, was also identified in keratinocytes [8], which suggests that ECS plays important physiological and pathological roles in skin. Accordingly, modulation of CBR activity in skin can be induced not only by the direct binding of ligand molecules, but also by the indirect change of endocannabinoid levels through the modulation of biochemical machinery. For example, stimulation of N-acylphosphatidylethanolamines (NAPEs)-hydrolyzing phospholipase D (PLD) activity, which catalyzes the cellular synthesis of AEA, stimulation of cellular uptake of AEA by a specific AEA membrane transporter (AMT), or inhibition of AEA-hydrolyzing enzyme fatty acid amide hydrolase (FAAH), can indirectly activate CBRs and further induce cellular responses. This kind of indirect activation of CBRs through the modulation of related enzymes or transporter activity is called an “entourage effect” [20]. While the “entourage effect” was first used for describing the modulating effects of fatty acid glycerol esters on 2-AG activity on CB receptors, it was then expanded to explain the biological activity of entourage compounds’ ability to interfere with the degradation of biologically active (target receptor binding) compounds, or to increase the affinity of active compounds on the target receptor [21]. This indirect modulation of CBRs underlies, at least in part, the mode of action for the current use of palmitoylethanolamine (PEA) as a topical anti-inflammatory and anti-pruritic ingredient [22,23]. It can be also suggested that, even with a CBR agonist with lower efficacy, a combination of FAAH inhibitors or other entourage compounds can provide sufficient therapeutic efficacy [24].

3. Physiological and Pathophysiological Roles of Cannabinoid Receptors in Skin

Among the diverse biological effects of CBR agonists in skin, the inhibition of epidermal keratinocyte proliferation, stimulation of epidermal keratinocytes differentiation, and anti-inflammatory activity suggest potential therapeutic applications for inflammatory skin diseases, including allergic contact dermatitis [25], atopic dermatitis [26], and psoriasis [27,28]. As summarized in Table 1, in vivo studies using various animal models and clinical studies, anti-inflammatory activities, anti-pruritic activities, and effects on epidermal permeability barrier functions were also explained (reviewed in [29] and [30]). Recently, using a CB1R and CB2R knockout mice model, Roelandt et al. showed a contradictory role of CB1R and CB2R on skin barrier function. While the absence of CB1R negatively impacted the recovery of the epidermal permeability barrier function after acute disruption, the knockout (-/-) of CB2R accelerated recovery. Secretion of lamellar body and formation of stratum corneum intercellular lipid bilayer structures were altered accordingly [31]. In both the fluorescein isothiocyanate-induced Th2-type contact hypersensitivity model and the 2,4-dinitrofluorobenzene-induced contact hypersensitivity model, protective activity of CB1R on skin barrier disruption, as well as inflammation, was also reported [28,32]. In addition to epidermal keratinocytes, CBRs in the mast cell are also reported to be involved in the EEC system. Ligand activation of CB1R reduced both the degranulation of mast cells and the maturation of mast cells from the progenitor cells in situ, which suggested a potential application of CB1R stimulation for allergic and mast cell-dependent skin diseases [33]. Additional mechanisms related with ECS signaling include transient receptor potential vanilloid-1 (TRPV-1) and peroxisome proliferator-activated receptor (PPAR) [34]. While detailed explanation about the roles of these receptors in skin is beyond the scope of this review, anti-pruritic effects of TRPV-1 agonist and anti-inflammatory activities of PPAR agonist are potentially related with CBR signaling (Figure 2) [35,36].

4. Development of Endocannabinomimetics and its Cosmetic Application

Focusing on the expected potential benefits, the dermatological and cosmetic application of the topical cannabinoid receptor agonist have been repeatedly reported. The development of cosmetic ingredients requires not only sufficient efficacy, but also strict safety without local and systemic adverse effects. Hence, cannabinomimetics as cosmetic ingredients might be based on endocannabinoid-mimicking compounds, considering the potential undesirable psycho-activity possibly associated with phyto-cannabinoid mimetics. Most of the currently identified endocannabinoids consist of two functional chemical groups: long chain-fatty acids either in saturated or unsaturated form, and hydrophilic moieties, including ethanolamine, dopamine, and glycerol (Figure 1) [2]. Considering these two groups are commonly conjugated via an amide linkage-forming fatty acid amide compound, it can be hypothesized that the conjugation of amino acid as hydrophilic moiety into long chain-fatty acid might introduce CBR agonistic or antagonistic activity. Based on this hypothesis, previously we have reported the synthesis of a cannabinoid receptor agonist and its effects on skin barrier function. A series of fatty acid amide molecules were synthesized and their CBR modulating activity was screened. As a result, two compounds, α-oleoyl oleoylamine ethanolamine (α-OOE) and α-oleoyl oleoylamine serinol (α-OOS), were identified as having CB1R agonistic effects, while no effects were observed on CB2R. A series of in vitro studies showed that significant anti-inflammatory activities are exerted by these agonists, which were further confirmed by investigations using an oxazolone-induced atopic dermatitis model [39]. In addition to anti-inflammatory activity, an accelerated recovery of epidermal permeability barrier function after acute disruption was also observed, which suggests that a topical CBR agonist can improve skin barrier function as well [40]. Since the conjugation of serinol as a hydrophilic moiety can also incorporate CBR modulating activity into fatty acids, new fatty acid amide molecules conjugated with serinol were synthesized and their CBR agonistic activity was tested. As a result, N-palmitoyl serinol was identified as a CB1R agonistic ligand, whose binding activity was comparable to ACEA in the test system (Figure 3). Interestingly, increased endogenous ceramide concentration by N-palmitoyl serinol treatment and the consequent stimulation of the apoptosis signal was reported in neuroblastoma cells [48]. Clinical efficacy, as tested on small number of healthy volunteers, showed a slight reduction of trans-epidermal water loss (TEWL) and significant increases in skin hydration immediately after one application (Figure 3). These results suggest that endocannabinomimetics can be used for improving skin barrier functions, alleviating inflammatory responses, and relieving itching sensations, and can be a new candidate for new cosmeceutical ingredients for both sensitive and diseased skin.

5. Future Perspectives

While the cannabinoid-related studies are mostly focused on the modulation of neurological functions, the topical application of cannabinoids for local effects has also been receiving significant attention recently. The use of a phytocannabinoid, such as cannabidiol, is getting more popular in the cosmetic industry, and these are commonly labeled as hemp-containing products [49]. However, the use of phytocannabinoid might draw potential safety issues related to its impurities with psychoactivity or legal concerns in certain countries. Hence, the use of endocannabinoid or endocannabinomimetics directly modulating CBRs in skin, or the use of “entourage effects” ingredients which indirectly regulate the activity of endocannabinoids, can be a possible option. Development of endocannabinoidomimetics based on natural or unnatural amino acids as hydrophilic moiety and their practical application has been tried, and a few candidates are already in cosmetic use. The further combination of endocannabinoidomimitics and entourage ingredients might provide sufficient therapeutic efficacy for skin diseases, including atopic dermatitis or psoriasis.

Author Contributions

For research articles with several authors, a short paragraph specifying their individual contributions must be provided. conceptualization, S.J. and B.D.P.; writing—original draft preparation, S.J.; writing—review and editing, M.S.K. and S.H.L.; supervision, S.J.; project administration, S.J.

Funding

This research received no external funding.

Conflicts of Interest

S. Jeong and B. Park are stockholders of Sphingobrain Inc. B. Park is stockholder of Dr. Raymond laboratories Inc.

References

  1. Gaoni, Y.; Mechoulam, R. The isolation and structure of delta-1-tetrahydrocannabinol and other neutral cannabinoids from hashish. J. Am. Chem. Soc. 1971, 93, 217–224. [Google Scholar] [CrossRef]
  2. Lu, H.C.; Mackie, K. An Introduction to the endogenous cannabinoid system. Biol. Psychiatry 2016, 79, 516–525. [Google Scholar] [CrossRef] [PubMed]
  3. Chicca, A.; Arena, C.; Manera, C. Beyond the direct activation of cannabinoid receptors: New strategies to modulate the endocannabinoid system in CNS-related diseases. Recent Pat. CNS Drug Discov. 2016, 10, 122–141. [Google Scholar] [CrossRef]
  4. Piscitelli, F.; Bradshaw, H.B. Endocannabinoid analytical methodologies: Techniques that drive discoveries that drive techniques. Adv. Pharmacol. 2017, 80, 1–30. [Google Scholar] [CrossRef]
  5. Howlett, A.C.; Abood, M.E. CB1 and CB2 receptor pharmacology. Adv. Pharmacol. 2017, 80, 169–206. [Google Scholar] [CrossRef]
  6. Svízenská, I.; Dubový, P.; Sulcová, A. Cannabinoid receptors 1 and 2 (CB1 and CB2), their distribution, ligands and functional involvement in nervous system structures—A short review. Pharmacol. Biochem. Behav. 2008, 90, 501–511. [Google Scholar] [CrossRef]
  7. Río, C.D.; Millán, E.; García, V.; Appendino, G.; DeMesa, J.; Muñoz, E. The endocannabinoid system of the skin. A potential approach for the treatment of skin disorders. Biochem. Pharmacol. 2018, 157, 122–133. [Google Scholar] [CrossRef] [PubMed]
  8. Maccarrone, M.; Di Rienzo, M.; Battista, N.; Gasperi, V.; Guerrieri, P.; Rossi, A.; Finazzi-Agrò, A. The endocannabinoid system in human keratinocytes. Evidence that anandamide inhibits epidermal differentiation through CB1 receptor-dependent inhibition of protein kinase C, activation protein-1, and transglutaminase. J. Biol. Chem. 2003, 278, 33896–33903. [Google Scholar] [CrossRef]
  9. Brown, A.J. Novel cannabinoid receptors. Br. J. Pharmacol. 2007, 152, 567–575. [Google Scholar] [CrossRef] [Green Version]
  10. Pérez-Gómez, E.; Andradas, C.; Flores, J.M.; Quintanilla, M.; Paramio, J.M.; Guzmán, M.; Sánchez, C. The orphan receptor GPR55 drives skin carcinogenesis and is upregulated in human squamous cell carcinomas. Oncogene 2013, 32, 2534–2542. [Google Scholar] [CrossRef]
  11. Adinolfi, B.; Romanini, A.; Vanni, A.; Martinotti, E.; Chicca, A.; Fogli, S.; Nieri, P. Anticancer activity of anandamide in human cutaneous melanoma cells. Eur. J. Pharmacol. 2013, 718, 154–159. [Google Scholar] [CrossRef]
  12. Cohen, L.J.; Kang, H.S.; Chu, J.; Huang, Y.H.; Gordon, E.A.; Reddy, B.V.; Ternei, M.A.; Craig, J.W.; Brady, S.F. Functional metagenomic discovery of bacterial effectors in the human microbiome and isolation of commendamide, a GPCR G2A/132 agonist. Proc. Natl. Acad. Sci. USA 2015, 112, E4825–E4834. [Google Scholar] [CrossRef] [Green Version]
  13. Mallipeddi, S.; Janero, D.R.; Zvonok, N.; Makriyannis, A. Functional selectivity at G-protein coupled receptors: Advancing cannabinoid receptors as drug targets. Biochem. Pharmacol. 2017, 128, 1–11. [Google Scholar] [CrossRef] [Green Version]
  14. Rieder, S.A.; Chauhan, A.; Singh, U.; Nagarkatti, M.; Nagarkatti, P. Cannabinoid-induced apoptosis in immune cells as a pathway to immunosuppression. Immunobiology 2010, 215, 598–605. [Google Scholar] [CrossRef] [PubMed]
  15. Gómez del Pulgar, T.; Velasco, G.; Sánchez, C.; Haro, A.; Guzmán, M. De novo-synthesized ceramide is involved in cannabinoid-induced apoptosis. Biochem. J. 2002, 363, 183–188. [Google Scholar] [CrossRef] [PubMed]
  16. Gustafsson, K.; Sander, B.; Bielawski, J.; Hannun, Y.A.; Flygare, J. Potentiation of cannabinoid-induced cytotoxicity in mantle cell lymphoma through modulation of ceramide metabolism. Mol. Cancer Res. 2009, 7, 1086–1098. [Google Scholar] [CrossRef] [PubMed]
  17. Raduner, S.; Majewska, A.; Chen, J.Z.; Xie, X.Q.; Hamon, J.; Faller, B.; Altmann, K.H.; Gertsch, J. Alkylamides from Echinacea are a new class of cannabinomimetics. Cannabinoid type 2 receptor-dependent and -independent immunomodulatory effects. J. Biol. Chem. 2006, 281, 14192–14206. [Google Scholar] [CrossRef]
  18. Moore, D.J.; Rawlings, A.V. The chemistry, function and (patho)physiology of stratum corneum barrier ceramides. Int. J. Cosmet. Sci. 2017, 39, 366–372. [Google Scholar] [CrossRef] [Green Version]
  19. Park, K.; Elias, P.M.; Hupe, M.; Borkowski, A.W.; Gallo, R.L.; Shin, K.O.; Lee, Y.M.; Holleran, W.M.; Uchida, Y. Resveratrol stimulates sphingosine-1-phosphate signaling of cathelicidin production. J. Investig. Dermatol. 2013, 133, 1942–1949. [Google Scholar] [CrossRef]
  20. Ben-Shabat, S.; Fride, E.; Sheskin, T.; Tamiri, T.; Rhee, M.H.; Vogel, Z.; Bisogno, T.; De Petrocellis, L.; Di Marzo, V.; Mechoulam, R. An entourage effect: Inactive endogenous fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity. Eur. J. Pharmacol. 1998, 353, 23–31. [Google Scholar] [CrossRef]
  21. Fowler, C.J. Plant-derived, synthetic and endogenous cannabinoids as neuroprotective agents. Non-psychoactive cannabinoids, ‘entourage’ compounds and inhibitors of N-acyl ethanolamine breakdown as therapeutic strategies to avoid pyschotropic effects. Brain Res. Brain Res. Rev. 2003, 41, 26–43. [Google Scholar] [CrossRef]
  22. Petrosino, S.; Cristino, L.; Karsak, M.; Gaffal, E.; Ueda, N.; Tüting, T.; Bisogno, T.; De Filippis, D.; D’Amico, A.; Saturnino, C.; et al. Protective role of palmitoylethanolamide in contact allergic dermatitis. Allergy 2010, 65, 698–711. [Google Scholar] [CrossRef]
  23. Alhouayek, M.; Muccioli, G.G. Harnessing the anti-inflammatory potential of palmitoylethanolamide. Drug Discov. Today 2014, 19, 1632–1639. [Google Scholar] [CrossRef]
  24. Di Marzo, V.; Melck, D.; Orlando, P.; Bisogno, T.; Zagoory, O.; Bifulco, M.; Vogel, Z.; De Petrocellis, L. Palmitoylethanolamide inhibits the expression of fatty acid amide hydrolase and enhances the anti-proliferative effect of anandamide in human breast cancer cells. Biochem. J. 2001, 358, 249–255. [Google Scholar] [CrossRef] [Green Version]
  25. Karsak, M.; Gaffal, E.; Date, R.; Wang-Eckhardt, L.; Rehnelt, J.; Petrosino, S.; Starowicz, K.; Steuder, R.; Schlicker, E.; Cravatt, B.; et al. Attenuation of allergic contact dermatitis through the endocannabinoid system. Science 2007, 316, 1494–1497. [Google Scholar] [CrossRef] [PubMed]
  26. Haruna, T.; Soga, M.; Morioka, Y.; Imura, K.; Furue, Y.; Yamamoto, M.; Hayakawa, J.; Deguchi, M.; Arimura, A.; Yasui, K. The inhibitory effect of S-777469, a cannabinoid type 2 receptor agonist, on skin inflammation in mice. Pharmacology 2017, 99, 259–267. [Google Scholar] [CrossRef] [PubMed]
  27. Wilkinson, J.D.; Williamson, E.M. 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] [CrossRef] [PubMed]
  28. Gaffal, E.; Cron, M.; Glodde, N.; Bald, T.; Kuner, R.; Zimmer, A.; Lutz, B.; Tüting, T. Cannabinoid 1 receptors in keratinocytes modulate proinflammatory chemokine secretion and attenuate contact allergic inflammation. J. Immunol. 2013, 190, 4929–4936. [Google Scholar] [CrossRef]
  29. Marks, D.H.; Friedman, A. The Therapeutic Potential of Cannabinoids in Dermatology. Skin Ther. Lett. 2018, 23, 1–5. [Google Scholar]
  30. Oddi, S.; Maccarrone, M. Endocannabinoids and Skin Barrier Function: Molecular Pathways and Therapeutic Opportunities. In Skin Stress Response Pathways; Wondrak, G.T., Ed.; Springer International Publishing: Berlin, Germany, 2016; pp. 301–323. [Google Scholar] [CrossRef]
  31. Roelandt, T.; Heughebaert, C.; Bredif, S.; Giddelo, C.; Baudouin, C.; Msika, P.; Roseeuw, D.; Uchida, Y.; Elias, P.M.; Hachem, J.P. Cannabinoid receptors 1 and 2 oppositely regulate epidermal permeability barrier status and differentiation. Exp. Dermatol. 2012, 21, 688–693. [Google Scholar] [CrossRef] [PubMed]
  32. Gaffal, E.; Glodde, N.; Jakobs, M.; Bald, T.; Tüting, T. Cannabinoid 1 receptors in keratinocytes attenuate fluorescein isothiocyanate-induced mouse atopic-like dermatitis. Exp. Dermatol. 2014, 23, 401–406. [Google Scholar] [CrossRef]
  33. Sugawara, K.; Bíró, T.; Tsuruta, D.; Tóth, B.I.; Kromminga, A.; Zákány, N.; Zimmer, A.; Funk, W.; Gibbs, B.F.; Zimmer, A.; et al. Endocannabinoids limit excessive mast cell maturation and activation in human skin. J. Allergy Clin. Immunol. 2012, 129, 726–738. [Google Scholar] [CrossRef]
  34. Tóth, K.F.; Ádám, D.; Bíró, T.; Oláh, A. Cannabinoid Signaling in the Skin: Therapeutic Potential of the “C(ut)annabinoid” System. Molecules 2019, 24, 918. [Google Scholar] [CrossRef] [PubMed]
  35. Muller, C.; Morales, P.; Reggio, P.H. Cannabinoid ligands targeting TRP channels. Front. Mol. Neurosci. 2018, 11, 487. [Google Scholar] [CrossRef] [PubMed]
  36. O’Sullivan, S.E. An update on PPAR activation by cannabinoids. Br. J. Pharmacol. 2016, 173, 1899–1910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Oláh, A.; Szabó-Papp, J.; Soeberdt, M.; Knie, U.; Dähnhardt-Pfeiffer, S.; Abels, C.; Bíró, T. Echinacea purpurea-derived alkylamides exhibit potent anti-inflammatory effects and alleviate clinical symptoms of atopic eczema. J. Dermatol. Sci. 2017, 88, 67–77. [Google Scholar] [CrossRef]
  38. Bort, A.; Alvarado-Vazquez, P.A.; Moracho-Vilrriales, C.; Virga, K.G.; Gumina, G.; Romero-Sandoval, A.; Asbill, S. Effects of JWH015 in cytokine secretion in primary human keratinocytes and fibroblasts and its suitability for topical/transdermal delivery. Mol. Pain 2017, 13, 174480691668822. [Google Scholar] [CrossRef] [PubMed]
  39. Nam, G.; Jeong, S.K.; Park, B.M.; Lee, S.H.; Kim, H.J.; Hong, S.-P.; Kim, B.; Kim, B.-W. Selective Cannabinoid receptor-1 agonists regulate mast cell activation in an oxazolone-induced atopic dermatitis model. Ann. Dermatol. 2018, 28, 22–29. [Google Scholar] [CrossRef]
  40. Kim, H.J.; Kim, B.; Park, B.M.; Jeon, J.E.; Lee, S.H.; Mann, S.; Ahn, S.K.; Hong, S.P.; Jeong, S.K. Topical cannabinoid receptor 1 agonist attenuates the cutaneous inflammatory responses in oxazolone-induced atopic dermatitis model. Int. J. Dermatol. 2015, 54, e401–e408. [Google Scholar] [CrossRef] [PubMed]
  41. Gaffal, E.; Cron, M.; Glodde, N.; Tüting, T. Anti-inflammatory activity of topical THC in DNFB-mediated mouse allergic contact dermatitis independent of CB1 and CB2 receptors. Allergy 2013, 68, 994–1000. [Google Scholar] [CrossRef]
  42. De Petrocellis, L.; Guida, F.; Moriello, A.S.; De Chiaro, M.; Piscitelli, F.; de Novellis, V.; Maione, S.; Di Marzo, V. N-palmitoyl-vanillamide (palvanil) is a non-pungent analogue of capsaicin with stronger desensitizing capability against the TRPV1 receptor and anti-hyperalgesic activity. Pharmacol. Res. 2011, 63, 294–299. [Google Scholar] [CrossRef] [PubMed]
  43. Malfitano, A.M.; Sosa, S.; Laezza, C.; De Bortoli, M.; Tubaro, A.; Bifulco, M. Rimonabant reduces keratinocyte viability by induction of apoptosis and exerts topical anti-inflammatory activity in mice. Br. J. Pharmacol. 2011, 162, 84–93. [Google Scholar] [CrossRef]
  44. Mecs, L.; Tuboly, G.; Toth, K.; Nagy, E.; Nyari, T.; Benedek, G.; Horvath, G. Peripheral antinociceptive effect of 2-arachidonoyl-glycerol and its interaction with endomorphin-1 in arthritic rat ankle joints. Clin. Exp. Pharmacol. Physiol. 2010, 37, 544–550. [Google Scholar] [CrossRef] [PubMed]
  45. Pulvirenti, N.; Nasca, M.R.; Micali, G. Topical adelmidrol 2% emulsion, a novel aliamide, in the treatment of mild atopic dermatitis in pediatric subjects: A pilot study. Acta Dermatovenerol. Croat. 2007, 15, 80–83. [Google Scholar] [PubMed]
  46. Petrosino, S.; Puigdemont, A.; Della Valle, M.F.; Fusco, M.; Verde, R.; Allarà, M.; Aveta, T.; Orlando, P.; Di Marzo, V. Adelmidrol increases the endogenous concentrations of palmitoylethanolamide in canine keratinocytes and down-regulates an inflammatory reaction in an in vitro model of contact allergic dermatitis. Vet. J. 2016, 207, 85–91. [Google Scholar] [CrossRef] [PubMed]
  47. Oka, S.; Wakui, J.; Gokoh, M.; Kishimoto, S.; Sugiura, T. Suppression by WIN55212-2, a cannabinoid receptor agonist, of inflammatory reactions in mouse ear: Interference with the actions of an endogenous ligand, 2-arachidonoylglycerol. Eur. J. Pharmacol. 2006, 538, 154–162. [Google Scholar] [CrossRef]
  48. Bieberich, E.; Kawaguchi, T.; Yu, R.K. N-Acylated serinol is a novel ceramide mimic inducing apoptosis in neuroblastoma cells. J. Biol. Chem. 2000, 275, 177–181. [Google Scholar] [CrossRef] [PubMed]
  49. Meng, Q.; Buchanan, B.; Zuccolo, J.; Poulin, M.M.; Gabriele, J.; Baranowski, D.C. A reliable and validated LC-MS/MS method for the simultaneous quantification of 4 cannabinoids in 40 consumer products. PLoS ONE 2018, 13, e0196396. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of phytocannabinoids and endocannabinoids.
Figure 1. Chemical structure of phytocannabinoids and endocannabinoids.
Cosmetics 06 00033 g001
Figure 2. Effects of cannabinoid receptor 1/cannabinoid receptor 2 (CBR1/CBR2) on skin barrier functions.
Figure 2. Effects of cannabinoid receptor 1/cannabinoid receptor 2 (CBR1/CBR2) on skin barrier functions.
Cosmetics 06 00033 g002
Figure 3. Characterization of N-palmitoyl serinol. (A) Activation of CB1R by tested compounds were measured using cAMP Hunter™ eXpress CNR1 (CB1) CHO-K1 GPCR Assay (Eurofins DiscoverX Corporation, Fremont, CA) according to the manufacturer’s recommendation. Results analysis was performed using GraphPad Prism, GraphPad Software, La Jolla, CA. (B) The structural property of N-Palmitoyl serinol-containing topical formulation was observed under cross-polarized microscope and the lateral organization of formulation was analyzed using wide-angle X-Ray Diffraction (WAXS). Optical anisotropy representing the concentric lamellar structure of emulsion particles, was observed under cross-polarized microscope, which is also observed from the lipids extracted from human stratum corneum. In order to analyze the lateral packing of formulation, the sample was put into a capillary (Mark-Rohrchem aus Glas no, length 80 mm, auBen 0.7 mm, Article no.4007807, Hilgenberg, Germany) and analyzed with D8 Discover XRD for WXRD. The Cu-Kα wavelength was set at 40 kV and 40 mA. The collimater was fixed at 0.3 mm and measured at 150 s per step. The wavelength from the sample was measured by transmission through a 30 cm helium gas tube. Distinct peaks at spacing of 0.41 nm and 0.37 nm represents existence of hexagonal and orthorhombic lateral packings respectively, within the emulsion particles. (C) Clinical efficacy of test formulation was evaluated in normal healthy human volunteers (N = 7). The control product was commercially available hexagonal gel-structured emulsion. While the change of trans-epidermal water loss (TEWL) was not changed in a significant way, an increase in skin hydration, as assessed by skin surface capacitance, was observed only in the test formulation-applied site.
Figure 3. Characterization of N-palmitoyl serinol. (A) Activation of CB1R by tested compounds were measured using cAMP Hunter™ eXpress CNR1 (CB1) CHO-K1 GPCR Assay (Eurofins DiscoverX Corporation, Fremont, CA) according to the manufacturer’s recommendation. Results analysis was performed using GraphPad Prism, GraphPad Software, La Jolla, CA. (B) The structural property of N-Palmitoyl serinol-containing topical formulation was observed under cross-polarized microscope and the lateral organization of formulation was analyzed using wide-angle X-Ray Diffraction (WAXS). Optical anisotropy representing the concentric lamellar structure of emulsion particles, was observed under cross-polarized microscope, which is also observed from the lipids extracted from human stratum corneum. In order to analyze the lateral packing of formulation, the sample was put into a capillary (Mark-Rohrchem aus Glas no, length 80 mm, auBen 0.7 mm, Article no.4007807, Hilgenberg, Germany) and analyzed with D8 Discover XRD for WXRD. The Cu-Kα wavelength was set at 40 kV and 40 mA. The collimater was fixed at 0.3 mm and measured at 150 s per step. The wavelength from the sample was measured by transmission through a 30 cm helium gas tube. Distinct peaks at spacing of 0.41 nm and 0.37 nm represents existence of hexagonal and orthorhombic lateral packings respectively, within the emulsion particles. (C) Clinical efficacy of test formulation was evaluated in normal healthy human volunteers (N = 7). The control product was commercially available hexagonal gel-structured emulsion. While the change of trans-epidermal water loss (TEWL) was not changed in a significant way, an increase in skin hydration, as assessed by skin surface capacitance, was observed only in the test formulation-applied site.
Cosmetics 06 00033 g003
Table 1. Summary of studies about the effects of topical cannabinoid modulating compound (abbreviations: CB1R, CB2R; cannabinoid receptor 1 and cannabinoid receptor 2 respectively; FAAH: fatty acid amide hydrolase; AE: atopic eczema; IL-6: Interleukin-6; SCORAD: SCORing of Atopic Dermatitis; SC: Stratum corneum; MCP-1: monocyte chemoattractant protein-1; AEA: Arachinonoyl ethanolamine; TPA: 12-O-tetradecanoylphorbol-13-acetate; THC: Δ9-Tetrahydrocannabinol; 2-AG: 2-arachidonoyl glycerol; IGA: investigator’s global assessment).
Table 1. Summary of studies about the effects of topical cannabinoid modulating compound (abbreviations: CB1R, CB2R; cannabinoid receptor 1 and cannabinoid receptor 2 respectively; FAAH: fatty acid amide hydrolase; AE: atopic eczema; IL-6: Interleukin-6; SCORAD: SCORing of Atopic Dermatitis; SC: Stratum corneum; MCP-1: monocyte chemoattractant protein-1; AEA: Arachinonoyl ethanolamine; TPA: 12-O-tetradecanoylphorbol-13-acetate; THC: Δ9-Tetrahydrocannabinol; 2-AG: 2-arachidonoyl glycerol; IGA: investigator’s global assessment).
Compounds(Proposed) Mode of ActionStudy ModelEfficacy ObservedReference
Echinacea species-derived alkylamideCB2R agonist
Partial inhibition of FAAH
In vitro studyReduction of pro-inflammatory cytokines (IL-6 and IL-8)[37]
Clinical study on AEReduction of local SCORADindex
Increases in SC intercellular lipids
JWH015Selective CB2R agonistIn vitro studyReduction of pro-inflammatory cytokines (IL-6 and MCP-1)[38]
AEA mimic compoundSelective CB1R agonistIn vitro studyReduction of mast cell activation[39]
Oxazolone induced AE modelSuppression of mast cells infiltration into skin
AEA mimic compoundSelective CB1R agonistSkin barrier recovery after acute disruptionAccelerated recovery of skin barrier function[40]
TPA-induced acute inflammation modelReduction of oedema
Oxazolone induced AE modelImprovement in skin barrier function
THCCBR agonistDNFB-mediated allergic contact dermatitis modelReduction of keratinocyte-derived pro-inflammatory mediators[41]
N-palmitoyl-vanillamide (palvanil)Potential CBR agonist
Potential FAAH inhibitor
In vitro studyLower pungency and stronger anti-hyperalgesic activity[42]
Eye-wiping assayanti-hyperalgesic activity
Rimonabant (SR141716)CB1R antagonistCroton oil-induced ear dermatitis modelReduction of oedema and leukocyte infiltration[43]
2-AGCBR agonistCarrageenan-induced joint inflammation modelAntinociceptive effects[44]
AdelmidrolCBR agonistClinical study on AEImprovement in IGA score[45]
AdelmidrolIncreasing intracellular PEA concentration (Entourage effect)In vitro studyInhibition of the release of the pro-inflammatory chemokine MCP-2[46]
WIN55212-2CBR agonistTPA-induced acute inflammation modelReduction of oederma and leukocyte infiltration[47]
SR144528CB2R antagonistTPA-induced acute inflammation modelReduction of oederma and leukocyte infiltration

Share and Cite

MDPI and ACS Style

Jeong, S.; Kim, M.S.; Lee, S.H.; Park, B.D. Epidermal Endocannabinoid System (EES) and its Cosmetic Application. Cosmetics 2019, 6, 33. https://doi.org/10.3390/cosmetics6020033

AMA Style

Jeong S, Kim MS, Lee SH, Park BD. Epidermal Endocannabinoid System (EES) and its Cosmetic Application. Cosmetics. 2019; 6(2):33. https://doi.org/10.3390/cosmetics6020033

Chicago/Turabian Style

Jeong, Sekyoo, Min Seok Kim, Sin Hee Lee, and Byeong Deog Park. 2019. "Epidermal Endocannabinoid System (EES) and its Cosmetic Application" Cosmetics 6, no. 2: 33. https://doi.org/10.3390/cosmetics6020033

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop