8
Review Article Endocannabinoid signaling and its regulation by nutrients Tiziana Bisogno 1,2 * Mauro Maccarrone 2,3 * 1 Endocannabinoid Research Group, Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli, Italy 2 Center of Integrated Research, Campus Bio-Medico University of Rome, 00128 Rome, Italy 3 European Center of Brain Research/Santa Lucia Foundation, 00143 Rome, Italy Abstract Diet plays a central role in maintaining health throughout life and a controlled food intake is associated to a reduced risk of certain diseases. A proper diet should include vitamins, miner- als, carbohydrates, proteins, and fats that have to be optimally balanced in order to exert their physiological functions. The endogenous ligands of type-1 and type-2 cannabinoid recep- tors, N-arachidonoyl-ethanolamine and 2-arachidonoylglycerol, are arachidonic acid (AA) derivatives whose levels are regu- lated by the activity of metabolic enzymes, as well as by AA availability. Since the only sources of AA in mammals are diet and the enzymatic production in the liver from shorter-chain essential fatty acids like linoleic acid, it is realistic to hypothe- size that endocannabinoid levels might be modulated by fatty acid composition of food. Therefore, in this review we summa- rize literature data indicating that endocannabinoid levels, and hence their activity at cannabinoid receptors, might be modu- lated by food composition. We focused our attention on die- tary fatty acid content, and on type and esterified form of fatty acids in the different diets. V C 2014 BioFactors, 00(00):000–000, 2014 Keywords: diet; endocannabinoid system; fatty acids 1. Introduction New scientific knowledge regarding the role that diet may play in preventing disease is rapidly emerging, as evidence contin- ues to reveal components within food that not only promote general health and well-being, but can also reduce the risk of illness. Health food, functional food, dietary supplements, and probiotics are now terms well-known not only to the scientific community but also to the consumers. It has been well- established that diet plays a central role in maintaining health throughout life. In the Western world, diet has significantly changed in recent years and obesity, diabetes, cardiovascular disease, hypertension, dyslipidemia, and cancer are only some of the pathological conditions that might benefit from a con- trolled food intake. People in developed countries tend to con- sume increasingly palatable food that, together with a diet rich in sugars, promotes excess body weight [1]. The endocannabinoid system (ECS) is a ubiquitous lipid signaling system in which different proteins control or modu- late several physiological processes and impact a huge variety of human diseases. These include, among others, neurological and neuropsychiatric diseases [2,3], obesity and metabolic Abbreviations: AA, arachidonic acid; AEA, N-arachidonoyl-ethanolamine; 2-AG, 2-arachidonoylglycerol; ALA, a-linolenic acid; CB 1 and CB 2 , cannabi- noid receptors type 1 and type 2; CLA, conjugated linoleic acid; DHA, docosahexaenoic acid; DHEA, N-docosahexaenoyl-ethanolamine; eCBs, endocannabinoids; ECS, endocannabinoid system; EPA, eicosapentaenoic acid; EPEA, N-eicosapentaenoyl-ethanolamine; FO, fish oil; KO, krill oil; LA, linolenic acid; NAE, N-acyl-ethanolamine; NAPE, N-arachidonoyl-phosphati- dylethanolamine; OA, oleic acid; OEA, N-oleoylethanolamine PA, palmitic acid, PEA, N-palmitoylethanolamine; PL, phospholipids; PLC, phospholi- pase C; PLD, phospholipase D; PUFA, polyunsaturated fatty acid; TAGs, triacylglycerols. V C 2014 International Union of Biochemistry and Molecular Biology Volume 00, Number 00, Month/Month 2014, Pages 00–00 *Address for correspondence: Dr. Tiziana Bisogno, Endocannabinoid Research Group, Institute of Biomolecular Chemistry, National Research Council, Via C. Flegrei 34, 80078 Pozzuoli, Italy. Tel.: 139-081 8675093; Fax: 139-081 8041770; E-mail: [email protected] or Prof. Mauro Maccar- rone, Center of Integrated Research, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy. Tel.: 139-06 225419169; Fax: 139-06 22541456; E-mail: [email protected]. Received 8 April 2014; accepted 8 April 2014 DOI 10.1002/biof.1167 Published online 00 Month 2014 in Wiley Online Library (wileyonlinelibrary.com) BioFactors 1

Endocannabinoid signaling and its regulation by nutrients

  • Upload
    mauro

  • View
    213

  • Download
    1

Embed Size (px)

Citation preview

Review Article

Endocannabinoid signaling and its regulation

by nutrients

Tiziana Bisogno1,2*

Mauro Maccarrone2,3*

1Endocannabinoid Research Group, Institute of Biomolecular Chemistry,National Research Council, 80078 Pozzuoli, Italy2Center of Integrated Research, Campus Bio-Medico University of Rome,00128 Rome, Italy3European Center of Brain Research/Santa Lucia Foundation, 00143 Rome,Italy

Abstract

Diet plays a central role in maintaining health throughout life

and a controlled food intake is associated to a reduced risk of

certain diseases. A proper diet should include vitamins, miner-

als, carbohydrates, proteins, and fats that have to be optimally

balanced in order to exert their physiological functions. The

endogenous ligands of type-1 and type-2 cannabinoid recep-

tors, N-arachidonoyl-ethanolamine and 2-arachidonoylglycerol,

are arachidonic acid (AA) derivatives whose levels are regu-

lated by the activity of metabolic enzymes, as well as by AA

availability. Since the only sources of AA in mammals are diet

and the enzymatic production in the liver from shorter-chain

essential fatty acids like linoleic acid, it is realistic to hypothe-

size that endocannabinoid levels might be modulated by fatty

acid composition of food. Therefore, in this review we summa-

rize literature data indicating that endocannabinoid levels, and

hence their activity at cannabinoid receptors, might be modu-

lated by food composition. We focused our attention on die-

tary fatty acid content, and on type and esterified form of fatty

acids in the different diets. VC 2014 BioFactors, 00(00):000–000,

2014

Keywords: diet; endocannabinoid system; fatty acids

1. IntroductionNew scientific knowledge regarding the role that diet may playin preventing disease is rapidly emerging, as evidence contin-ues to reveal components within food that not only promotegeneral health and well-being, but can also reduce the risk ofillness. Health food, functional food, dietary supplements, andprobiotics are now terms well-known not only to the scientificcommunity but also to the consumers. It has been well-established that diet plays a central role in maintaining healththroughout life. In the Western world, diet has significantlychanged in recent years and obesity, diabetes, cardiovasculardisease, hypertension, dyslipidemia, and cancer are only someof the pathological conditions that might benefit from a con-trolled food intake. People in developed countries tend to con-sume increasingly palatable food that, together with a diet richin sugars, promotes excess body weight [1].

The endocannabinoid system (ECS) is a ubiquitous lipidsignaling system in which different proteins control or modu-late several physiological processes and impact a huge varietyof human diseases. These include, among others, neurologicaland neuropsychiatric diseases [2,3], obesity and metabolic

Abbreviations: AA, arachidonic acid; AEA, N-arachidonoyl-ethanolamine;2-AG, 2-arachidonoylglycerol; ALA, a-linolenic acid; CB1 and CB2, cannabi-noid receptors type 1 and type 2; CLA, conjugated linoleic acid; DHA,docosahexaenoic acid; DHEA, N-docosahexaenoyl-ethanolamine; eCBs,endocannabinoids; ECS, endocannabinoid system; EPA, eicosapentaenoicacid; EPEA, N-eicosapentaenoyl-ethanolamine; FO, fish oil; KO, krill oil; LA,linolenic acid; NAE, N-acyl-ethanolamine; NAPE, N-arachidonoyl-phosphati-dylethanolamine; OA, oleic acid; OEA, N-oleoylethanolamine PA, palmiticacid, PEA, N-palmitoylethanolamine; PL, phospholipids; PLC, phospholi-pase C; PLD, phospholipase D; PUFA, polyunsaturated fatty acid; TAGs,triacylglycerols.

VC 2014 International Union of Biochemistry and Molecular BiologyVolume 00, Number 00, Month/Month 2014, Pages 00–00

*Address for correspondence: Dr. Tiziana Bisogno, EndocannabinoidResearch Group, Institute of Biomolecular Chemistry, National ResearchCouncil, Via C. Flegrei 34, 80078 Pozzuoli, Italy. Tel.: 139-081 8675093;Fax: 139-081 8041770; E-mail: [email protected] or Prof. Mauro Maccar-rone, Center of Integrated Research, Campus Bio-Medico University ofRome, Via �Alvaro del Portillo 21, 00128 Rome, Italy. Tel.: 139-06225419169; Fax: 139-06 22541456; E-mail: [email protected] 8 April 2014; accepted 8 April 2014DOI 10.1002/biof.1167Published online 00 Month 2014 in Wiley Online Library(wileyonlinelibrary.com)

BioFactors 1

defects [4], as well as cardiovascular disorders [5], cancer [6],and gastrointestinal pathologies [7]. The ECS comprises endog-enous lipid transmitters (known as endocannabinoids, eCBs),their G-protein-coupled receptors (i.e., type 1 (CB1) and type 2(CB2) cannabinoid receptors), and the proteins responsible foreCB biosynthesis, inactivation, transport, and accumulation[8,9]. The main eCBs (Fig. 1), N-arachidonoyl-ethanolamine(anandamide, AEA) and 2-arachidonoylglycerol (2-AG), areproduced by an important n26 dietary polyunsaturated fattyacid (PUFA), that is arachidonic acid (AA), which is also thebiosynthetic precursor of a plethora of other chemical media-tors, overall known as “eicosanoids.” Thus, it is possible topredict that modifications of dietary intake may modulate eCBlevels, and then regulate several physiological functions medi-ated by their signaling. The ECS is implicated in both homeo-static, appetite-triggered, and hedonic (i.e., desire to consumehighly palatable foods that often occurs during periods of rela-tive energy abundance) food intakes [10]. Therefore, under-standing how direct manipulation of nutrients alters ECS activ-ity is a relevant research area, with a potential exploitation fora nutritional approach to treat human diseases. In this review,we summarize the main components of the ECS and discusshow expression and/or activity of its distinct elements might bemodulated by diet.

2. The Endocannabinoid SystemThat Cannabis preparations can stimulate appetite in humans,a phenomenon known as “munchies,” has been known forhundreds of years, but only the identification of the ECS pro-vided the rationale to the phenomenon and suggested a possi-ble interplay between food composition and ECS modulation[11]. Two G-protein-coupled receptors (GPCRs) with high affin-ity and specificity for D9-tetrahydrocannabinol (THC) havebeen cloned to date, CB1 and CB2 [12,13], that are bound andfunctionally activated by several endogenous ligands, termedeCBs. The main members of this group of lipids, AEA [14], and2-AG [15,16], are derived from AA that is hydrolyzed frommembrane phospholipids (PLs) (Fig. 2). Indeed, the direct PLprecursor of AEA is N-arachidonoyl-phosphatidylethanolamine(NAPE), which originates from the trans-acylase-catalyzedtransfer of AA from the sn21 position of PLs to the nitrogenatom of phosphatidylethanolamine [17,18]. NAPE can be con-verted into AEA in a one-step hydrolysis reaction, catalyzed bythe NAPE-specific phospholipase D (NAPE-PLD) [19]. Yet,transgenic mice where the nape-pld gene had been ablated

(NAPE-PLD “knockout,” NAPE-PLD-KO) do not exhibit reducedlevels of AEA in most tissues [20], suggesting that AEA can beformed from NAPE also via other biosynthetic pathways,including: (i) formation of phospho-AEA catalyzed by a phos-pholipase C (PLC) and subsequent action of a protein tyrosinephosphatase N22 [21], that hydrolyzes phospho-AEA to AEA;(ii) phosphodiesterase-mediated hydrolysis of glycerophospho-AEA, which in turn is produced via sequential cleavage of thetwo sn21 and 2-acyl groups of NAPE, catalyzed by a=b-hydrolase 4 (Abdh4) [22]; and (iii) formation of a 2-lyso-NAPEvia a soluble form of phospholipase A2 [23], followed by lyso-PLD-mediated hydrolysis of 2-lyso-NAPEs.

Moreover, by the same biosynthetic pathway responsiblefor AEA formation, other N-acyl-ethanolamines (NAEs), suchas N-palmitoylethanolamine (PEA) or N-oleoylethanolamine(OEA) are formed from palmitic acid (PA) or oleic acid (OA),respectively, esterified at sn21 position of PLs [18]. PEA andOEA do not activate CB receptors and exert their biologicalactivity by interacting with other targets. In particular, theanti-inflammatory mediator PEA acts by activating the nuclearperoxisome proliferator-activated receptor-a (PPAR-a) [24], orby enhancing AEA actions at CB1, transient receptor potentialvanilloid type-1 (TRPV1) channels, or PPAR-c [25]. The ano-rexigenic and neuroprotective agent OEA acts by activatingPPAR-a [26], TRPV1 channels [27], and GPR119 [28]. WhileAEA and other NAEs derive from fatty acids esterified on thesn21 position of PLs, in most cases 2-AG is produced from thehydrolysis of diacylglycerols (DAGs) containing arachidonate inthe 2 position, catalyzed by a DAG lipase that is selective forthe sn21 position [29]. DAGs, in turn, can be produced fromthe hydrolysis of either phosphoinositides (PI), catalyzed by aPI-selective phospholipase C (PI-PLC) [30], or phosphatidic acid(PA) [31], as shown in Fig. 3. The enzymatic inactivation ofAEA and 2-AG occurs through the hydrolysis of their amideand ester bonds, releasing AA and ethanolamine or glycerol,respectively, that are rapidly incorporated into membrane PLs[32,33]. Fatty acid amide hydrolase (FAAH) [34] has been iden-tified as the enzyme mainly responsible for AEA, and to aminor extent for 2-AG, hydrolysis [35]. Additionally, the mono-acylglycerol lipase (MAGL) is the enzyme that most specificallycontrols the levels of 2-AG [36,37]. Even though the latterenzyme is responsible for �85% of the total brain 2-AG degra-dation, two more enzymes, a/b-hydrolase-6 (ABHD6) and a/b-hydrolase-12 (ABHD12), with different subcellular distributionscompared to MAGL, have been suggested to control, togetherwith FAAH, the hydrolysis of distinct pools of 2-AG in the nerv-ous system [38]. From the observations reported above, it canbe anticipated that AEA is a minor component among NAEsand 2-AG is the most abundant among its congeners, reflectingthe relatively little and high amounts of AA esterified to thesn21 or sn22 position of PLs, respectively [39]. Therefore, it isrealistic to hypothesize that FA composition in the diet mightmodulate the amounts of FAs esterified on PLs and, as a con-sequence, the tissue concentrations of eCBs that in turndepend on essential FAs availability. Therefore, in the next

Chemical structures of the main eCBs.FIG 1

BioFactors

2 Endocannabinoid Signaling and Nutrients

sections, we summarize the available in vitro and in vivo stud-ies, as well as the clinical data, that support this hypothesis.

3. Effect of Dietary Fatty Acids onEndocannabinoid LevelsFatty acids found naturally in the diet are classified accord-ing to the number of carbon atoms in their side chains, andare thus short (<8), medium (8–12), long (14–18), or verylong (�20) chain fatty acids. The double-bonds in natural

unsaturated fatty acids are generally in the cis configuration.The two major families of PUFAs are the n26 (x-6) and then23 (x-3) fatty acids, they are named after the position ofthe first double bond from the x- or n-terminal methyl end ofthe molecule. Linoleic acid (LA) and a-linolenic acid (ALA)are the two traditionally recognized essential fatty acids,which are the 18 carbon atom precursors of the n26 andn23 families, respectively. They include the n26 AA, and then23 eicosapentaenoic acid (EPA) and docosahexaenoic acid(DHA).

Major biosynthetic pathways for anandamide.FIG 2

3

3.1. In Vitro StudiesThe hypothesis that the availability of biosynthetic precursors,rather than the activity of the biosynthetic enzymes, might beresponsible for changes in eCB levels was investigated in vitro inmouse adipocyte cell line, 3T3F442A, supplemented with differ-ent free FAs, like LA, ALA, DHA, AA, OA, and PA [40]. PUFAschanged the amount of AA on the sn22 position of PLs and con-sequently affected the levels of 2-AG. In particular, DHA reducedthe amount of AA esterified on both sn22 and sn21 position ofPLs, but not of triacylglycerols (TAGs), and decreased 2-AG lev-els; instead, AA increased 2-AG levels and the amounts of AAesterified on both TAGs and PLs at the glycerol sn22, but notsn21, carbon [40]. Moreover, while other free FAs had no statis-tically significant effect on 2-AG levels, both PA and OA enhancedthe content of PEA and OEA [40]. In MC3T3-E1 osteoblast-likecells, EPA was reported to modulate distinct elements of theECS, as it reduced NAPE-PLD and CB2 mRNA expression thatincreased during osteoblast maturation, unfortunately the effectof EPA on eCB levels was not addressed in that study [41].Recently, evidence for in vitro formation of n23 (x-3) NAEs, syn-thesized from their corresponding PUFAs has been reported, butthe potential interaction of x-3 NAEs with the ECS remains to beinvestigated. 3T3-L1 adipocytes are indeed able to convert DHA

and EPA to their NAE derivatives N-docosahexaenoyl-ethanola-mine (DHEA) and N-eicosapentaenoyl-ethanolamine (EPEA),respectively, that exert anti-inflammatory properties by decreas-ing Lipopolysaccharide (LPS)-induced Interleukin (IL)-6 andMonocyte chemoattractant protein (MCP)-1 levels in adipocytes[42]. In line with this formation of DHEA and of its putative directbiosynthetic precursor, N-docosahexaenoyl-PE, as well as that ofdidocosahexaenoyl-PC and -PE, has been reported in bovine ret-ina expressing high levels of DHA [43]. DHEA and EPEA werealso endogenously produced following x-3 FAs treatments in dif-ferent human prostate and breast cancer cell lines [44,45] butcell supplementation of DHA or EPA apparently did not signifi-cantly affect 2-AG and AEA levels [44]. Possibly relevant to theabove studies, at least to some extent, is the finding that theavailability of the precursor FAs, supplemented to the cell, ratherthan its incorporation into membrane PLs might be responsiblefor the x-3 NAE formation and for the lack of effect on the n26(x-6) NAE and glycerol levels.

3.2. Animal StudiesThe consumption of n–3 (x-3) and n26 (x-6) essential FAs inWestern diets has changed markedly during the 20th century,with an increase in LA availability from �3% to �8% of energysupply [46]. Since humans cannot synthesize AA de novo, theonly AA source apart from diet is the enzymatic elongationand desaturation of LA precursor [47], hence is also the pre-cursor of eCBs. The hypothesis that LA, might be a diet modu-lator of eCB levels was investigated in mice supplied for 14weeks with various diets, differing in LA content from 1 per-cent of energy (1 en%) to 8 en% [48]. Dietary LA increased tis-sue AA, subsequently elevating 2-AG and AEA levels and pro-moting accumulation of body fat. The adipogenic effect of LAcould be prevented by adding 1 en% EPA and DHA to an 8en% LA diet, able to reduce the AA-PL pool, and to normalizeeCBs tone [48]. In addition, an isocaloric increase of dietaryLA from 1 to 8 en% in both low fat diet (LFD, 12.5 en%) andmedium fat diets (MFD, 35 en%) for 16 weeks elevated livereCB levels and increased the risk to develop obesity [49].Moreover, in a recent study Atlantic salmons (Salmo salar L.)were fed with a high level of LA (8 en%) from soyabean oil(SO) for 6 months, and salmon fillet was used for 16 weeks toproduce food for mice. The replacement of fish oil (FO) withSO in the diet caused an increase in LA, AA, and eCB levels insalmon liver, that subsequently elevated eCB levels in mice fedthe SO salmon diet compared with mice fed the FO salmondiet [49]. The elevated eCB activity in mice was associatedwith increasing weight gain and body weight, supporting therole of dietary LA as a potential key factor in controlling theactivity of the ECS. The presence of LA or conjugated linoleicacid (CLA) in the diet was found to correlate with modulationof eCB levels also in the brain. Indeed, in mice fed with dietscontaining 3% LA or 3% CLA for 4 weeks, the alteration of eCBlevels seemed to be FA-specific, as CLA- but not LA-enricheddiet increased 2-AG but not AEA content selectively in the cer-ebral cortex [50]. Moreover, newborn piglets fed with milk

Major biosynthetic pathways for 2-AG.FIG 3

BioFactors

4 Endocannabinoid Signaling and Nutrients

formulations enriched in AA and DHA during the first monthof life showed significantly modified concentrations of the cor-responding NAEs in different brain regions, and such altera-tions were accompanied by changes in the distribution ofPUFAs in PLs [51]. A lipidomic analysis of acute 2-week DHAdietary supplementation showed a significant elevation ofDHA, EPA, 2-eicosapentaenoylglycerol (EPG), and DHEA inmurine plasma and brain, with a concomitant reduction ofAEA levels in the brain [52]. Since FA composition in dietmight modulate eCB levels in the brain, and CB1 might medi-ate emotional responses [53], it is reasonable to postulate thatdiets enriched or deprived with either n23 or n26 PUFAsmight produce effects on mood. In particular, maternal dietaryfat was reported to modulate AA incorporation into PLs andeCB production, with potential consequences on hypothalamic-pituitary-adrenal axis modulation during stress in developingpups [54]. Pregnant rats were fed a 5% (controls) or 30% highfat (HF) diet rich in either n26 (HF-n26) or n23 (HF-n23) fatduring the last week of gestation and lactation. On post-natalday 10 of lactation, milk from dams belonging to the HF-n23group displayed a reduced n26/n23 fat ratio compared withcontrol and HF milk, reflecting the composition of maternaldiet. Hypothalamic and hippocampal levels of AA-PLs andeCBs were found to be diet-specific, with positive correlationsin both the hippocampus and hypothalamus for 2-AG, and anegative correlation for AEA in the hypothalamus. Moreover,stress-induced Adreno Cortico Tropic Hormone (ACTH) secre-tion increased upon pre-treatment with CB1/CB2 antagonistsonly in control pumps [54]. More recently, in order to mimiclifelong n26/n23 imbalance of essential PUFAs, mice havebeen fed with n23 deficient diet throughout gestation and lac-tation; after weaning, the offspring received the same dietthroughout the rest of their life [55]. Reduced n23 FA levels inthe diet modified FA composition, and markedly diminishedthe synaptic and behavioral functions of CB1 receptor in twobrain regions that have been implicated in emotional behaviorand mood disorders (i.e., prefrontal cortex and accumbens),without significant alteration of eCB levels in the brain [55].Moreover, a low dietary level of DHA has been associated withincreased risk of developing neuropsychiatric diseases,because nutritional n23 deficiency throughout life causes ananxiety/depressive-like behavior in mice [56]. In particular,mice fed a n23-deficient diet exhibited altered CB1/CB2 signal-ing pathways in prefrontal cortex and hypothalamus, asrevealed by the ability of the CB1/CB2 agonist WIN55,212-2 toactivate MAPK pathway [56].

Does dietary form of fatty acids impact on eCB levels?

Different studies support the hypothesis that the choice of thedietary form of PUFAs may affect their lipid incorporationand/or nutritional activity, overall suggesting that PL-boundEPA and DHA have distinct effects compared to TAG-boundEPA and DHA [57]. In particular, n23 PUFAs are reported tobe mainly bound to PLs in fish products, while in fatty fishsuch as salmon they are bound to PLs and TAGs in a 40:60

ratio; when available as supplement like in FO, n23 PUFAsare almost exclusively bound to TAGs [58]. A relatively newsource rich in n23 PUFAs in the form of PLs (mainly PC)rather than TAGs is krill oil (KO), extracted from Antarctickrill (Euphausia superba) [59]. The proportion of PLs in thetotal lipids of krill has been reported to vary between 30% and60%, and the constitutive presence of a lipid-soluble antioxi-dant like astaxanthin appears to preserve KO from oxidation[60]. Recently, the effects on ectopic fat and inflammation ofeither FO or KO, balanced for EPA and DHA content, wereinvestigated in Zucker rats, a model of obesity and relatedmetabolic dysfunction [61], and were compared with a controldiet devoid of EPA and DHA and with similar contents of OA,LA, and ALA. Rats fed with KO for 4 weeks had significantlylower liver TAGs and reduced peritoneal macrophageresponse to inflammatory stimulus, compared with controlrats. Moreover, a lower concentration of AEA and 2-AG wasfound in the visceral adipose tissue, and of AEA only in theliver and heart [61]. These effects were associated with lowerlevels of AA in membrane PLs, yet not with higher activity ofeCB-degrading enzymes, suggesting that the beneficial effectsof KO might be ascribed to changes in membrane FA composi-tion rather than changes in eCB catabolism [61]. This hypothe-sis was confirmed by measuring eCB levels in different periph-eral tissues, including inguinal and epididymal adipose tissue,liver, gastrocnemius muscle, kidneys, and heart from mice fedfor 8 weeks with HFD diets with different KO content [62]. KOdose-dependently reduced AEA and/or 2-AG levels in almostall the tissues analyzed, by reducing the availability of the bio-synthetic precursors of these compounds. The higher efficacyof n23 PUFAs administered as PLs rather than TAGs was alsoreported by Rossmeisl et al. (2012) [63]. In dietary obese mice,DHA/EPA administered as PLs prevented glucose intoleranceand obesity better than TAGs, and only the PL form reducedplasma insulin and adipocyte hypertrophy, being also moreeffective in reducing hepatic steatosis and low-grade inflam-mation of white adipose tissue (WAT). These beneficial effectswere correlated with reduced 2-AG levels in WAT itself.

4. Human StudiesUnlike in vitro and in vivo animal studies, only a few datahave been reported in humans on the potential modulation ofthe ECS by n23 PUFAs. The effect of 4 weeks dietary treat-ment with KO, menhaden oil (MO), or olive oil (OO) in over-weight or obese individuals has been recently reported [64–66]. KO significantly decreased 2-AG but not AEA plasma lev-els in obese subjects, while MO or OO treatments did not mod-ify eCB levels in either overweight or obese individuals [64].Moreover, as previously observed in mice [62], in overweight/obese subjects the inhibitory effect of KO on eCB levels corre-lated with a reduction of the n26/n23 PUFA ratio in plasmaPLs [64]. In addition, dietary supplementation to obese men ofDHA and EPA for 24 weeks, in the form of krill powder, wasreported to be able to reduce high TAGs content and plasma

5

AEA levels [65]. The reduction of triglyceridemia and eCB lev-els was associated with a decreased waist/hip and visceral fat/skeletal muscle mass ratio [65]. Moreover, data reported abovesuggested that treatments with krill formulations may producedifferent effects on plasma eCB levels depending on differentcohorts of subjects, duration of treatments (4 vs. 24 weeks),and dosage (2 g/day vs. 4 g/day). Furthermore, the use of asheep cheese naturally enriched in ALA, CLA, and vaccenicacid (VA), has been more recently associated to a modulationof eCB profiles in mildly hypercholesterolaemic subjects [66]. A3-week treatment of 42 adult volunteers with control orenriched cheese significantly decreased AEA plasma levels,and after 90 g/per day of enriched cheese, leptin and AEA con-centrations appeared to be strongly correlated [66].

5. Do Spices and Food Plants Modulatethe ECS?The lipid nature of eCB calls for attention on whether and howother dietary fats might modulate eCB levels and their signal cas-cades, and indeed in the last few years several ingredients ofspice or food plant elements (Fig. 4) have emerged for their abil-ity to interact to different extents with ECS. The sesquiterpene(E)-caryophyllene [(E)-BCP] is a major plant volatile aromatic ter-pene found in large amounts in the essential oils of many differ-ent spice and food plants, such as oregano (Origanum vulgareL.), cinnamon (Cinnamomum spp.) and black pepper (Piper nig-rum L.) [67–69]. Because of its weak aromatic taste, (E)-BCP iscommercially used as a food additive. It has been shown that (E)-BCP selectively targets CB2 and produces analgesic and anti-inflammatory effects in vivo [70,71]. Falcarinol [(3R,9Z)21,9-hep-

tadecadiene-4,6-diyn-3-ol] containing vegetables like carrot (Dau-cus carota L.), celery (Apium graveolens L.), fennel (Foeniculumvulgare Mill.), and parsnip (Pastinaca sativa L.) induce pro-allergic effects in skin and exhibit affinity to both human CB1

and CB2 [72]. Several isoprenylated analogs of the naturallyoccurring plant stilbenoid trans-resveratrol, such as arachidin-1[trans-4-(3-methyl-1-butenyl)23,5,30,40-tetrahydroxystilbene] andarachidin-3 [trans-4-(3-methyl-1-butenyl)23,5,40-trihydroxystil-bene], bind to both CB1 and CB2 with low affinity (in the lMrange) [73]. Plant natural products have been also suggested toexert cannabimimetic effects without direct interaction with CB1/CB2 receptors. In particular, genistein and daidzein, found infood sources such as lupin (Lupinus albus, L.), fava (Vicia faba,L.), beans and soybeans (Glycine max L.), inhibit FAAH in a lowmM range [74,75]. Biochanin A, a phytoestrogen present in soy-beans, chickpea (Cicer arietinum), and other legumes, inhibitsFAAH both in vitro and in vivo, and has only modest effects onCB1 and CB2 receptors [76]. Although it is likely that these com-pounds might exert beneficial effects in humans, data on theirestimated daily intake and therapeutic efficacy are still missing.

6. ConclusionsAvailable data summarized in this review highlight the sensitiv-ity of eCB levels to FA composition of the diet. It seems evidentthat the presence in the diet of certain PUFAs has as first conse-quence the alteration of the availability of the eCB biosyntheticprecursors, thus resulting in an alteration of the eCB levels. Thewell-established involvement of the ECS in several central andperipheral human pathologies suggests that any dietary manipu-lation of eCB levels needs to be time- and space-specific, inorder to warrant a correct ECS function and therapeutic exploi-tation. For instance, while obesity and dyslipidemia might bene-fit from a reduction of eCB levels in particular brain areas (i.e.,hypothalamus) or peripheral organs (i.e., adipose tissue or gas-trointestinal tract), the same reduction in other brain regions(i.e., hippocampus or hypothalamus) might support developmentof neuropsychiatric diseases and mood disorders. Further stud-ies are necessary to assess a nutritional approach as alternativetherapy to treat metabolic or emotional diseases. Diet, depend-ing on its FA composition, might reduce (much alike CB1/CB2

antagonists and/or inhibitors of eCB biosynthetic enzymes) orenhance (much alike CB1/CB2 agonists and/or inhibitors of eCBhydrolytic enzymes) the biological activity of eCBs, thus finelytuning its therapeutic potential. On a final note, to fully appreci-ate the relevance of the therapeutic benefit derived from a die-tary modulation of eCB levels, it should be necessary also toevaluate the effect of different diets on the expression and func-tional activity of eCB binding receptors and metabolic enzymes.

AcknowledgementThis investigation was partly supported by Ministero dell’Istru-zione, dell’Universit�a e della Ricerca (PRIN 2010–2011 project)to TB and MM.

Chemical structures of spice and food plant ingre-

dients that interact with the ECS.FIG 4

BioFactors

6 Endocannabinoid Signaling and Nutrients

References

[1] Erlanson-Albertsson, C. (2005) How palatable food disrupts appetite regula-

tion. Basic Clin. Pharmacol. Toxicol. 97, 61–73.

[2] Bisogno, T. and Di Marzo, V. (2007) Short- and long-term plasticity of the

endocannabinoid system in neuropsychiatric and neurological disorders.

Pharmacol. Res. 56, 428–442.

[3] Maccarrone, M., Battista, N., and Centonze, D. (2007) The endocannabinoid

pathway in Huntington’s disease: a comparison with other neurodegenera-

tive diseases. Prog. Neurobiol. 81, 349–379.

[4] Engeli, S. (2012) Central and peripheral cannabinoid receptors as therapeutic

targets in the control of food intake and body weight. Handb. Exp. Pharma-

col. 209, 357–381.

[5] O’Sullivan, S. E., Kendall, P. J., and Kendall, D. A. (2012) Endocannabinoids

and the cardiovascular response to stress. J. Psychopharmacol. 26, 71–82.

[6] Grimaldi, C. and Capasso, A. (2011) The endocannabinoid system in the can-

cer therapy: an overview. Curr. Med. Chem. 18, 1575–1583.

[7] Schicho, R. and Storr, M. (2011) Alternative targets within the endocannabi-

noid system for future treatment of gastrointestinal diseases. Can. J. Gastro-

enterol. 25, 377–383.

[8] Maccarrone, M., Dainese, E., and Oddi, S. (2011) Intracellular trafficking of

anandamide: new concepts for signaling. Trends Biochem. Sci. 35, 601–608.

[9] Min, R., Di Marzo, V., and Mansvelder, H. D. (2010) DAG lipase involvement

in depolarization-induced suppression of inhibition: does endocannabinoid

biosynthesis always meet the demand? Neuroscientist 16, 608–613.

[10] Monteleone, P., Piscitelli, F., Scognamiglio, P., Monteleone, A. M.,

Canestrelli, B., et al. (2012) Hedonic eating is associated with increased

peripheral levels of ghrelin and the endocannabinoid 2-arachidonoyl-

glycerol in healthy humans: a pilot study. J. Clin. Endocrinol. Metab. 97,

E917–E924.

[11] Sharkey, K. A. and Pittman, Q. J. (2005) Central and peripheral signaling

mechanisms involved in endocannabinoid regulation of feeding: a perspec-

tive on the munchies. Sci. STKE. 2005, pe15.

[12] Matsuda, L. A., Lolait, S. J., Brownstein, M. J., Young, A. C., and Bonner, T.

I. (1990) Structure of a cannabinoid receptor and functional expression of

the cloned cDNA. Nature 346, 561–564.

[13] Munro, S., Thomas, K. L., and Abu-Shaar, M. (1993) Molecular characteriza-

tion of a peripheral receptor for cannabinoids. Nature 365, 61–65.

[14] Devane, W. A., Hanus, L., Breuer, A., Pertwee, R. G., Stevenson, L. A. et al.

(1992) Isolation and structure of a brain constituent that binds to the canna-

binoid receptor. Science 258, 1946–1949.

[15] Mechoulam, R., Ben-Shabat, S., Hanus, L., Ligumsky, M., Kaminski, N. E.,

et al. (1995) Identification of an endogenous 2-monoglyceride, present in

canine gut, that binds to cannabinoid receptors. Biochem. Pharmacol. 50,

83–90.

[16] Sugiura, T., Kondo, S., Sukagawa, A., Nakane, S. Shinoda, A., et al. (1995)

2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand

in brain. Biochem. Biophys. Res. Commun. 215, 89–97.

[17] Sugiura, T., Kondo, S., Sukagawa, A., Tonegawa, T., Natane, S., et al. (1996)

Transacylase-mediated and phosphodiesterase-mediated synthesis of N-

arachidonoylethanolamine, an endogenous cannabinoidreceptor ligand, in

rat brain microsomes. Comparison with synthesis from free arachidonic

acid and ethanolamine. Eur. J. Biochem. 240, 53–62.

[18] Schmid, H. H. (2000) Pathways and mechanisms of N-acylethanolamine bio-

synthesis: can anandamide be generated selectively? Chem. Phys. Lipids.

108, 71–87.

[19] Okamoto, Y., Morishita, J., Tsuboi, K., Tonai, T., and Ueda, N. (2004) Molec-

ular characterization of a phospholipase D generating anandamide and its

congeners. J. Biol. Chem. 279, 5298–5305.

[20] Leung, D., Saghatelian, A., Simon, G. M., and Cravatt, B. F. (2006) Inactiva-

tion of N-acyl-phosphatidylethanolamine phospholipase D reveals multiple

mechanisms for the biosynthesis of endocannabinoids. Biochemistry 45,

4720–4726.

[21] Liu, J., Wang, L., Harvey-White, J., Huang, B. X., Kim, H. Y., et al. (2008)

Multiple pathways involved in the biosynthesis of anandamide. Neurophar-

macology 54, 1–7.

[22] Simon, G. M. and Cravatt, B. F. (2006) Endocannabinoid biosynthesis pro-

ceeding through glycerophospho-N-acyl ethanolamine and a role for alpha/

beta-hydrolase 4 in this pathway. J. Biol. Chem. 281, 26465–26472.

[23] Sun, Y. X., Tsuboi, K., Okamoto, Y., Tonai, T., and Murakami M, et al. (2004)

Biosynthesis of anandamide and N-palmitoylethanolamine by sequential

actions of phospholipase A2 and lysophospholipase D. Biochem. J. 380,

749–756.

[24] LoVerme, J., La Rana, G., Russo, R., Calignano, A., and Piomelli, D. (2005)

The search for the palmitoylethanolamide receptor. Life Sci. 77, 1685–1698.

[25] Costa, B., Comelli, F., Bettoni, I., Colleoni, M., and Giagnoni, G. (2008) The

endogenous fatty acid amide, palmitoylethanolamide, has anti-allodynic

and anti-hyperalgesic effects in a murine model of neuropathic pain:

involvement of CB(1), TRPV1 and PPARgamma receptors and neurotrophic

factors. Pain 139, 541–550.

[26] Fu, J., Gaetani, S., Oveisi, F., Lo Verme, J., Serrano. A., et al. (2003) Oleyle-

thanolamide regulates feeding and body weight through activation of the

nuclear receptor PPAR-alpha. Nature 425, 90–93.

[27] Movahed, P., J€onsson, B. A., Birnir, B., Wingstrand, J. A., J�rgensen, T. D.

et al. (2005) Endogenous unsaturated C18 N-acylethanolamines are vanilloid

receptor (TRPV1) agonists. J. Biol. Chem. 280, 38496–38504.

[28] Hansen, H. S., Rosenkilde, M. M., Holst, J. J., and Schwartz, T. W. (2012)

GPR119 as a fat sensor. Trends Pharmacol. Sci. 33, 374–381.

[29] Bisogno, T., Howell, F., Williams, G., Minassi, A., Cascio, M. G., et al. (2003)

Cloning of the first sn1-DAG lipases points to the spatial and temporal regu-

lation of endocannabinoid signaling in the brain. J. Cell. Biol. 163, 463–468.

[30] Stella, N., Schweitzer, P., and Piomelli, D. (1997) A second endogenous can-

nabinoid that modulates long-term potentiation. Nature 388, 773–778.

[31] Bisogno, T., Melck, D., De Petrocellis, L., and Di Marzo, V. (1999) Phosphati-

dic acid as the biosynthetic precursor of the endocannabinoid 2-

arachidonoylglycerol in intact mouse neuroblastoma cells stimulated with

ionomycin. J. Neurochem. 72, 2113–2119.

[32] Di Marzo, V., Fontana, A., Cadas, H., Schinelli, S., Cimino, G., et al. (1994)

Formation and inactivation of endogenous cannabinoid anandanide in cen-

tral neurons. Nature 372, 686–691.

[33] Di Marzo, V., Bisogno, T., Sugiura, T., Melck, D., and De Petrocellis, L.

(1998) The novel endogenous cannabinoid 2-arachidonoylglycerol is inacti-

vated by neuronal- and basophil-like cells: connections with anandamide.

Biochem. J. 331, 15–19.

[34] Cravatt, B. F., Giang, D. K., Mayfield, S. P., Boger, D. L., Lerner, R. A., et al.

(1996) Molecular characterization of an enzyme that degrades neuromodula-

tory fatty-acid amides. Nature 384, 83–87.

[35] Patricelli, M. P. and Cravatt, B. F. (1999) Fatty acid amide hydrolase competi-

tively degrades bioactive amides and esters through a nonconventional cat-

alytic mechanism Biochemistry 38, 14125–14130.

[36] Dinh, T. P., Carpenter, D., Leslie, F. M., Freund, T. F., Katona, I., et al. (2002)

Brain monoglyceride lipase participating in endocannabinoid inactivation.

Proc. Natl. Acad. Sci. USA 99, 10819–10824.

[37] Dinh, T. P., Kathuria, S., and Piomelli, D. (2004) RNA interference suggests a

primary role for monoacylglycerol lipase in the degradation of the endocan-

nabinoid 2-arachidonoylglycerol. Mol. Pharmacol. 66, 1260–1264.

[38] Blankman, J. L., Simon, G. M., and Cravatt, B. F. (2007) A comprehensive

profile of brain enzymes that hydrolyze the endocannabinoide 2-arachido-

noylglycerol. Chem. Biol. 14, 1347–1356.

[39] Lands, W. E. (2000) Stories about acyl chains. Biochim. Biophys. Acta 1483,

1–14.

[40] Matias, I., Carta, G., Murru, E., Petrosino, S., Banni, S., et al. (2008). Effect of

polyunsaturated fatty acids on endocannabinoid and N-acyl-ethanolamine

levels in mouse adipocytes. Biochim. Biophys. Acta 1781, 52–60.

[41] Hutchins, H. L., Li, Y., Hannon, K., and Watkins, B. A. (2011) Eicosapenta-

enoic acid decreases expression of anandamide synthesis enzyme and can-

nabinoid receptor 2 in osteoblast-like cells. J. Nutr. Biochem. 22, 195–200.

[42] Balvers, M. G., Verhoeckx, K. C., Plastina, P., Wortelboer, H. M., Meijerink,

J., et al. (2010) Docosahexaenoic acid and eicosapentaenoic acid are con-

verted by 3T3-L1 adipocytes to N-acyl-ethanolamines with anti-

inflammatory properties. Biochim. Biophys. Acta 1801, 1107–1114.

7

[43] Bisogno, T., Delton-Vandenbroucke, I., Milone, A., Lagarde, M., and Di

Marzo, V. (1999) Biosynthesis and inactivation of N-arachidonoylethanol-

amine (anandamide) and N-docosahexaenoylethanolamine in bovine retina.

Arch. Biochem. Biophys. 370, 300–307.

[44] Brown, I., Cascio, M. G., Wahle, K. W., Smoum, R., Mechoulam, R., et al.

(2010) Cannabinoid receptor-dependent and -independent anti-proliferative

effects of omega-3 ethanolamides in androgen receptor-positive and -nega-

tive prostate cancer cell lines. Carcinogenesis 31, 1584–1591.

[45] Brown, I., Wahle, K. W., Cascio, M. G., Smoum-Jaouni, R., Mechoulam, R.,

et al. (2011) Omega-3 N-acylethanolamines are endogenously synthesised

from omega-3 fatty acids in different human prostate and breast cancer cell

lines. Prostaglandins Leukot. Essent. Fatty Acids 85, 305–310.

[46] Blasbalg, T. L., Hibbeln, J. R., Ramsden, C. E., Majchrzak, S. F., and

Rawlings, R. R. (2011) Changes in consumption of omega-3 and omega-6

fatty acids in the United States during the 20th century. Am. J. Clin. Nutr.

93, 950–962.

[47] Fischer, S. (1989) Dietary polyunsaturated fatty acids and eicosanoid forma-

tion in humans. Adv. Lipid Res. 23, 169–198.

[48] Alvheim, A. R., Malde, M. K., Osei-Hyiaman, D., Lin, Y. H., Pawlosky, R. J.,

et al. (2012) Dietary linoleic acid elevates endogenous 2-AG and ananda-

mide and induces obesity. Obesity (Silver Spring) 20, 1984–1994.

[49] Alvheim, A. R., Torstensen, B. E., Lin, Y. H., Lillefosse, H. H., Lock, E. J., et al.

(2013) Dietary linoleic acid elevates endogenous 2-arachidonoylglycerol and

anandamide in Atlantic salmon (Salmo salar L.) and mice, and induces

weight gain and inflammation in mice. Br. J. Nutr. 109, 1508–1517.

[50] Tsuyama, S., Oikawa, D., Tsuji, Y., Akimoto, Y., Jikuya, H., et al. (2009) Die-

tary conjugated linoleic acid modifies the brain endocannabinoid system in

mice. Nutr. Neurosci. 12, 155–159.

[51] Berger, A., Crozier, G., Bisogno, T., Cavaliere, P., Innis, S., et al. (2001)

Anandamide and diet: inclusion of dietary arachidonate and docosahexae-

noate leads to increased brain levels of the corresponding N-acylethanol-

amines in piglets. Proc. Natl. Acad. Sci. USA 98, 6402–6406.

[52] Wood, J. T., Williams, J. S., Pandarinathan, L., Janero, D. R., Lammi-Keefe,

C. J., et al. (2010) Dietary docosahexaenoic acid supplementation alters

select physiological endocannabinoid-system metabolites in brain and

plasma. J. Lipid Res. 51, 1416–1423.

[53] Lutz, B. (2009) Endocannabinoid signals in the control of emotion. Curr.

Opin. Pharmacol. 9, 46–52.

[54] D’Asti, E., Long, H., Tremblay-Mercier, J., Grajzer, M., Cunnane, S. C., et al.

(2010) Maternal dietary fat determines metabolic profile and the magnitude

of endocannabinoid inhibition of the stress response in neonatal rat off-

spring. Endocrinology 151, 1685–1694.

[55] Lafourcade, M., Larrieu, T., Mato, S., Duffaud, A., Sepers, M., et al. (2011)

Nutritional omega-3 deficiency abolishes endocannabinoid-mediated neuro-

nal functions. Nat. Neurosci. 14, 345–350.

[56] Larrieu, T., Madore, C., Joffre, C., and Lay�e, S. (2012) Nutritional n-3 polyun-

saturated fatty acids deficiency alters cannabinoid receptor signaling path-

way in the brain and associated anxiety-like behavior in mice. J. Physiol.

Biochem. 68, 671–681.

[57] Murru, E., Banni, S., and Carta, G. (2013) Nutritional properties of dietary

omega-3-enriched phospholipids. Biomed. Res. Int. 2013, 965417–965430.

[58] Polvi, S. M. and Ackman, R. G. (1992) Atlantic salmon (Salmo salar) muscle

lipids and their response to alternative dietary fatty acid sources. J. Agric.

Food Chem. 40, 1001–1007.

[59] Kolakowska, A., Kolakowska, E., and Szcygielski, M. (1994) Winter season

krill (Euphausia superba Dana). Nahrung 38, 128–134.

[60] Tou, J. C., Jaczynski, J., and Chen, Y. (2007) Krill for human consumption:

nutritional value and potential health benefits. Nutr. Rev. 65, 63–77.

[61] Batetta, B., Griinari, M., Carta, G., Murru, E., Ligresti, A., et al. (2009) Endo-

cannabinoids may mediate the ability of (n 2 3) fatty acids to reduce ectopic

fat and inflammatory mediators in obese Zucker rats. J. Nutr. 139, 1495–

1501.

[62] Piscitelli, F., Carta, G., Bisogno, T., Murru, E., Cordeddu, L., et al. (2011)

Effect of dietary krill oil supplementation on the endocannabinoidome of

metabolically relevant tissues from high-fat-fed mice. Nutr. Metab. (Lond).

8, 51–66.

[63] Rossmeisl, M., Jilkova, Z. M., Kuda, O., Jelenik, T., Medrikova, D., et al.

(2012) Metabolic effects of n 2 3 PUFA as phospholipids are superior to tri-

glycerides in mice fed a high-fat diet: possible role of endocannabinoids.

PLoS One. 7, e38834.

[64] Banni, S., Carta, G., Murru, E., Cordeddu, L., Giordano, E., et al. (2011) Krill

oil significantly decreases 2-arachidonoylglycerol plasma levels in obese

subjects. Nutr. Metab. (Lond) 8, 7–12.

[65] Berge, K., Piscitelli, F., Hoem, N., Silvestri, C., Meyer, I., et al., (2013) Chronic

treatment with krill powder reduces plasma triglyceride and anandamide

levels in mildly obese men. Lipids Health Dis. 12, 78–85.

[66] Pintus, S., Murru, E., Carta, G., Cordeddu, L., Batetta, B., et al., (2013) Sheep

cheese naturally enriched in a-linolenic, conjugated linoleic and vaccenic

acids improves the lipid profile and reduces anandamide in the plasma of

hypercholesterolaemic subjects. Br. J. Nutr. 109, 1453–1462.

[67] Orav, A., Stulova, I., Kailas, T., and Muurisepp M (2004) Effect of storage on

the essential oil composition of Piper nigrum L. fruits of different ripening

states. J. Agric. Food Chem. 52, 2582–2586.

[68] Jayaprakasha, G. K., Jagan Mohan Rao, L., and Sakariah, K. K. (2003) Vola-

tile constituents from Cinnamomum zeylanicum fruit stalks and their antiox-

idant activities. J. Agric. Food Chem. 51, 4344–4348.

[69] Mockute, D., Bernotiene, G., and Judzentiene, A. (2001) The essential oil of

Origanum vulgare L. ssp. vulgare growing wild in vilnius district (Lithuania).

Phytochemistry 57, 65–69.

[70] Gertsch, J., Leonti, M., Raduner, S., Racz, I., Chen, J. Z., et al. (2008) Beta-

caryophyllene is a dietary cannabinoid. Proc. Natl. Acad. Sci. USA 105,

9099–9104.

[71] Klauke, A. L., Racz, I., Pradier, B., Markert, A., Zimmer, A. M., et al. (2014)

The cannabinoid CB2 receptor-selective phytocannabinoid beta-

caryophyllene exerts analgesic effects in mouse models of inflammatory

and neuropathic pain. Eur. Neuropsychopharmacol., 24, 608–620.

[72] Leonti, M., Casu, L., Raduner, S., Cottiglia, F., Floris, C., et al., (2010) Falcari-

nol is a covalent cannabinoid CB1 receptor antagonist and induces pro-

allergic effects in skin. Biochem. Pharmacol. 79, 1815–1826.

[73] Brents, L. K., Medina-Bolivar, F., Seely, K. A, Nair, V., Bratton, S. M., et al.,

(2012) Natural prenylated resveratrol analogs arachidin-1 and 23 demon-

strate improved glucuronidation profiles and have affinity for cannabinoid

receptors. Xenobiotica 42, 139–156.

[74] Thors, L., Alajakku, K., and Fowler, C. J. (2007) The ‘specific’ tyrosine kinase

inhibitor genistein inhibits the enzymic hydrolysis of anandamide: implica-

tions for anandamide uptake. Br. J. Pharmacol. 150, 951–960.

[75] Thors, L., Belghiti, M., and Fowler, C. J. (2008) Inhibition of fatty acid amide

hydrolase by kaempferol and related naturally occurring flavonoids. Br. J.

Pharmacol. 155, 244–252.

[76] Thors, L., Burston, J. J., Alter, B. J., McKinney, M. K., Cravatt, B. F., et al.,

(2010) Biochanin A, a naturally occurring inhibitor of fatty acid amide hydro-

lase. Br. J. Pharmacol. 160, 549–560.

BioFactors

8 Endocannabinoid Signaling and Nutrients