You are on page 1of 4

ARTICLE IN PRESS

Phytomedicine 17 (2010) 19–22

Contents lists available at ScienceDirect

Phytomedicine
journal homepage: www.elsevier.de/phymed

Tea catechins’ affinity for human cannabinoid receptors


G. Korte a,n, A. Dreiseitel a, P. Schreier b, A. Oehme b, S. Locher b, S. Geiger c, J. Heilmann c, P.G. Sand a
a
Department of Psychiatry, University of Regensburg, Franz-Josef-Strauss-Allee 11, H4 R97 93053 Regensburg, Germany
b
Chair of Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
c
Chair of Pharmaceutical Biology, Institute of Pharmacy, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany

a r t i c l e in fo abstract

Among the many known health benefits of tea catechins count anti-inflammatory and neuroprotective
Keywords: activities, as well as effects on the regulation of food intake. Here we address cannabimimetic
Camellia sinensis bioactivity of catechin derivatives occurring in tea leaves as a possible cellular effector of these
Catechins functionalities. Competitive radioligand binding assays using recombinant human cannabinoid
Epigallocatechin-3-O-gallate receptors expressed in Chem-1 and CHO cells identified (–)-epigallocatechin-3-O-gallate, EGCG
Cannabinoid receptor (Ki = 33.6 mM), (–)-epigallocatechin, EGC (Ki =35.7 mM), and (–)-epicatechin-3-O-gallate, ECG (Ki =47.3
Neuroprotection mM) as ligands with moderate affinity for type 1 cannabinoid receptors, CB1. Binding to CB2 was weaker
with inhibition constants exceeding 50 mM for EGC and ECG. The epimers (+)-catechin and (–)-
epicatechin exhibited negligible affinities for both CB1 and CB2. It can be concluded that central nervous
cannabinoid receptors may be targeted by selected tea catechins but signaling via peripheral type
receptors is less likely to play a major role in vivo.
& 2009 Elsevier GmbH. All rights reserved.

Introduction rafts (Patra et al. 2008) and plasma membrane binding sites
(Bastianetto et al. 2009) have been implicated as molecular
Catechins are the most abundant polyphenols in the leaves of targets. We therefore hypothesized that catechin pharmacology
tea (Camellia sinensis (L.) O. KUNZE) but are also found in many could involve further sites of action and tested cannabimimetic
fruits and in some legumes, e.g. in cocoa beans (Arts et al. 2000). activities.
While the average daily catechin intake in Western diets has Two human cannabinoid receptors (CB) have so far been
been estimated at 50 mg (Arts et al. 2001), ‘‘Mediterranean’’ type identified. Of these, CB1 is expressed primarily in the central
diets contain twice as much catechins (Auger et al. 2004) nervous system (CNS) (Matsuda et al. 1990) where it acts through
and have become known for their health-promoting effects lipid rafts (Bari et al. 2005). CB1 plays a pivotal role in
(Ruidavets et al. 2000). These include a reduced incidence of neuroprotection (Galve-Roperh et al. 2008) and food intake
stroke (Keli et al. 1996; Tanabe et al. 2008), the slowing of age- (Richard et al. 2009). CB2, in contrast, is expressed predominantly
related cognitive decline (Kuriyama et al. 2006), and protection outside the CNS (Munro et al. 1993) and is most prevalent in cells
against obesity (Kao et al. 2000; Murase et al. 2006). Animal of the immune system (Lynn and Herkenham 1994). CB ligands
studies suggest an additional role of catechins or catechin comprise natural analgesics (Walker and Hohmann 2005) of
oligomers in nociception (Rylski et al. 1979; DalBó et al. 2005; which cannabinoids have been extensively investigated (Woelkart
Dias et al. 2007; Tang et al. 2007) and in imparting resistance to et al. 2008). The present study examines in vitro affinities of (–)-
stress (Abbas and Wink 2009). At the cellular level, catechins epigallocatechin-3-O-gallate (EGCG), (–)-epicatechin-3-O-gallate
induce anti-inflammatory (Tedeschi et al. 2002) and antioxidant (ECG), (–)-epigallocatechin EGC, (–)-epicatechin, and ( + )-catechin
activities (Higdon and Frei 2003) but there is some disagreement (Fig. 1) for human CB1 and CB2.
regarding the signaling pathways that mediate the above
functionalities (Cooper et al. 2005). Thus GABAergic, glutamater-
gic, monoamine and NO systems (Adachi et al. 2006; Chou et al. Materials and methods
2007; Rocha et al. 2007; Kim et al. 2007) have previously been
proposed as catechin effectors, among others. Only recently, lipid Membrane preparations of recombinant human cannabinoid
receptors 1 (Chem-1 cells) and 2 (CHO cells) were purchased from
Millipore (Schwalbach, Germany), catechin derivatives were
n
Corresponding author. Tel.: +49 941 944 8955; fax: + 49 941 944 8956. obtained from Extrasynthese (Genay, France), [3H]-CP55940 was
E-mail address: gabriele.korte@klinik.uni-regensburg.de (G. Korte). purchased from PerkinElmer (Boston, MA, USA) and unlabeled

0944-7113/$ - see front matter & 2009 Elsevier GmbH. All rights reserved.
doi:10.1016/j.phymed.2009.10.001
ARTICLE IN PRESS
20 G. Korte et al. / Phytomedicine 17 (2010) 19–22

Fig. 1. Chemical structures of catechin derivatives under study.

CP55940 was purchased from Sigma-Aldrich (Schnelldorf, 2.5 mM for ( + )-catechin and (–)-epicatechin with regard to CB1.
Germany). CB1 (Kd =29.4 nM) and CB2 (Kd = 12.0 nM) saturation Receptor affinities for CB1 were generally stronger than the
characteristics were determined with CP55940 concentrations of respective affinities for CB2 (Table 1). While CB1 inhibition
1–12.5 nM (six concentration steps), and 1–20 nM (five concen- constants below 50 mM were achieved by EGCG, EGC and ECG,
tration steps), respectively, using 10 mg (CB1) or 2.5 mg (CB2) of only EGCG exhibited a similar Ki for CB2. CB2 half-maximal
synaptosomes. For radioligand receptor assays, 100 ml of mem- inhibition could not be determined for ( +)-catechin and (–)-
brane preparations were added to 80 ml of assay buffer (50 mM epicatechin as values of these flavonoids fell outside the upper
Hepes, 0.5% BSA, pH 7.4), 10 ml of test compounds dissolved in detection limit of the displacement assay. The present data
DMSO and 10 ml of [3H]-CP55940, followed by an incubation of suggest that selected tea catechins feature moderate affinity for
90 min at room temperature in microtiter plates (final concentra- central type CB whereas binding to peripheral type CB is less
tions per well: DMSO 5% (v/v), ethanol 0.28% (v/v), radioligand prominent (Fig. 2). Thus cannabimimetic activities may contribute
12.5 nM or 15 nM, CB1/2 synaptosomes 10 mg). Specific binding to CNS effects of green tea extracts including the mitigation of
for each compound at each concentration was defined as total pain (Singal et al. 2005; Sattayasai et al. 2008), complementing a
binding minus binding in the presence of unlabeled CP55940 COX-2 inhibitory role (Kaur et al. 2005) and known opioid
(10 mM), and was determined for each concentration of [3H]- receptor functionalities of catechins (Capasso et al. 1998; Katavic
CP55940. Reactions were terminated by filtration through a GF/C et al. 2007). In addition, CB1 ligands EGCG, EGC and ECG in green
Filtermat A (PerkinElmer, Boston, MA, USA) using a 96-well tea may modulate food intake (Auvichayapat et al. 2008; Bose et
Inotech cell harvester (Dietikon, Switzerland) and by subsequent al. 2008). However, a cautionary note is warranted in that the
washing of filters with distilled water. Prior to filtration, filter magnitude of anti-obesity effects is not undisputed (Diepvens et
mats were first soaked in buffer containing 0.33% polyethylenei- al. 2005) and both anti-nociceptive and anti-obesity activities
mine for 1 h, and were washed with 50 mM Hepes (0.5% BSA, pH imply agonistic modulation of CB1 which is unproven by our
7.4). After filtration, filters were dried at 60 1C for 60 min and were experiments. Observational studies on neuroprotection by green
then transferred to scintillation vials. 4 ml of Rotiszint eco plus tea polyphenols (Kakuda 2002; Pan et al. 2003) lend some support
scintillation cocktail (Roth, Karlsruhe, Germany) were added to to putative CB1 agonistic effects but will require further
each vial and radioactivity was determined in a WinSpectral 1414 confirmation, e.g. by monitoring intracellular Ca2 + levels or
liquid scintillation counter (PerkinElmer Wallac GmbH, Freiburg, steady state [32P]-GTPase activity. As for anti-allergic properties
Germany). All experiments were performed in triplicate using of catechin-enriched preparations (Maeda-Yamamoto et al. 2007),
seven concentration steps per compound investigated. Mean these are less likely to result from binding to CB in view of mostly
values 7SD were obtained for each step and served for normal- poor CB2 Ki values.
ization and curve-fitting. Competitive radioligand displacement With respect to both CB1 and CB2, negligible bioactivities were
was evaluated by nonlinear regression analysis (GraphPad Prism noted for ungallated catechins. In previous reports, the galloyl
V2.01, GraphPad Software, LaJolla, CA, USA). Ki values were moiety has been associated with catechins’ radical scavenging
derived from the equation by Cheng and Prusoff (1973): qualities (Nanjo et al. 1996; Kinjo et al. 2002; Wolfe and Liu 2008),
EC50 with inhibition of COX-2 (Hou et al. 2007), protein tyrosine
Ki ¼ phosphatase (Okamoto et al. 2003), pancreatic lipase (Ikeda et al.
1 þ ½ligand
Kd 2005) and fatty acid synthase (Wang et al. 2003), but not with
enhanced GABAergic neurotransmission (Adachi et al. 2007). The
30 , 40 , 50 -trihydroxyl substitution in the catechin B-ring, in turn,
Results and discussion which has been implicated in antioxidant, apoptosis-inducing and
beta secretase-inhibiting activity of catechins (Furuno et al. 2002;
Dose-dependent binding to CB1 and CB2 was noted for all Saeki et al. 2000; Jeon et al. 2003) did not appear to contribute to
compounds under study. Overall, Ki values differed by several CB binding. More detailed structure-activity investigations are
orders of magnitude ranging from 33.6 mM for EGCG to over invited to address the interplay of galloyl and pyrogallol moieties,
ARTICLE IN PRESS
G. Korte et al. / Phytomedicine 17 (2010) 19–22 21

Table 1
Affinities of catechin derivatives and CP55940 for human cannabinoid receptors 1 and 2 as determined in competition binding assays on membrane preparations of
recombinant receptors. [3H]-CP55940 was used as radiolabeled ligand. All experiments were performed in triplicate.

Compound Ki (CB1) in lM 95% CI Ki (CB2) in lM 95% CI

(-)-epigallocatechin-3-O-gallate (EGCG) 33.6 17.2 - 65.7 42.8 17.9 - 101.0


(-)-epicatechin-3-O-gallate (ECG) 47.3 16.7 - 133.8 95.6 23.0 - 397.4
(-)-epigallocatechin (EGC) 35.7 3.5 - 368.2 361.3 0.5 - 24,000
( +)-catechin 42,500.0 n.d. n.d. n.d.
(-)-epicatechin 42,500.0 n.d. n.d. n.d.
CP55940 0.028 0.008 - .095 0.025 0.015 - .039

n.d.= not determined.

of ungallated flavan-3-ols (Warden et al. 2001; Henning et al.


CB1
2008), signal strength may be amplified in vivo by non-receptor
% specific [3H]-CP55940

100 related mechanisms, e.g. by inhibition of fatty acid amide


hydrolase or COX-2 (Jhaveri et al. 2008). Future studies will need
to address the impact of these confounders to allow for a balanced
binding

view of putative health benefits that may be mediated by CB.


50
CP55940
EGCG
EGC Acknowledgement
ECG
0
This work was supported by grant #0313848C from the
-10 -9 -8 -7 -6 -5 -4 -3 German Federal Ministry of Education, Science, Research and
log (drug) [M] Technology.

CB2
% specific [3H]-CP55940

100 References

Abbas, S., Wink, M., 2009. Epigallocatechin gallate from green tea (Camellia
binding

sinensis) increases lifespan and stress resistance in Caenorhabditis elegans.


50 Planta Med. 75, 216–221.
CP55940
Adachi, N., Tomonaga, S., Tachibana, T., Denbow, D.M., Furuse, M., 2006.
EGCG Epigallocatechin gallate attenuates acute stress responses through GABAergic
system in the brain. Eur. J. Pharmacol. 531, 171–175.
EGC
Adachi, N., Tomonaga, S., Suenaga, R., Denbow, D.M., Furuse, M., 2007. Galloyl
0 ECG group is not necessary for a sedative effect of catechin through GABAergic
system. Lett. Drug Des. Discovery 4, 163–167.
-10 -9 -8 -7 -6 -5 -4 -3 Arts, I.C., van de Putte, B., Hollman, P.C., 2000. Catechin contents of foods
commonly consumed in The Netherlands. 1. Fruits, vegetables, staple foods,
log (drug) [M] and processed foods. J. Agric. Food Chem. 48, 1746–1751.
Arts, I.C., Hollman, P.C., Feskens, E.J., Bueno de Mesquita, H.B., Kromhout, D., 2001.
Fig. 2. Displacement curves for selected CB1 and CB2 ligands in competition Catechin intake and associated dietary and lifestyle factors in a representative
binding assays using recombinant human receptors expressed in Chem-1 and CHO sample of Dutch men and women. Eur. J. Clin. Nutr. 55, 76–81.
cells. Auvichayapat, P., Prapochanung, M., Tunkamnerdthai, O., Sripanidkulchai, B.O.,
Auvichayapat, N., Thinkhamrop, B., Kunhasura, S., Wongpratoom, S., Sinawat,
S., Hongprapas, P., 2008. Effectiveness of green tea on weight reduction in
obese Thais: a randomized, controlled trial. Physiol. Behav. 93, 486–491.
as well as further studies on structural and stereochemical  A., Maccarrone, M., 2005. Lipid rafts
Bari, M., Battista, N., Fezza, F., Finazzi-Agro,
prerequisites for cannabinoid signaling. control signaling of type-1 cannabinoid receptors in neuronal cells.
Recent pharmacokinetic data suggest that green tea polyphe- Implications for anandamide-induced apoptosis. J. Biol. Chem. A280,
nols are readily absorbed in the small intestine and reach peak 12,212–12,220.
Bastianetto, S., Dumont, Y., Han, Y., Quirion, R., 2009. Comparative neuroprotective
plasma concentrations at 1.6 to 2.3 hours post ingestion (Stalmach properties of stilbene and catechin analogs: action via a plasma membrane
et al. 2009). Apart from indirect evidence using catechin-enriched receptor site? CNS Neurosci. Ther. 15, 76–83.
diets (e.g. Nagao et al. 2009), unfortunately, limited proof is Bose, M., Lambert, J.D., Ju, J., Reuhl, K.R., Shapses, S.A., Yang, C.S., 2008. The major
green tea polyphenol, (  )-epigallocatechin-3-gallate, inhibits obesity,
currently available from placebo-controlled trials of catechins to metabolic syndrome, and fatty liver disease in high-fat-fed mice. J. Nutr. 138,
put their putative bioactivities into perspective. Some have 1677–1683.
concluded to poor effects of catechins on sustained weight loss Capasso, A., Piacente, S., Pizza, C., Sorrentino, L., 1998. Flavonoids reduce morphine
withdrawal in-vitro. J. Pharm. Pharmacol. 50, 561–564.
(Hursel and Westerterp-Plantenga 2009) while others have Cheng, Y., Prusoff, W.H., 1973. Relationship between the inhibition constant (K1)
reported a favourable outcome with regard to plasma glucose, and the concentration of inhibitor which causes 50 per cent inhibition (IC50)
and compliance with diet in overweight subjects (Hill et al. 2007; of an enzymatic reaction. Biochem. Pharmacol. 22, 3099–3108.
Chou, C.W., Huang, W.J., Tien, L.T., Wang, S.J., 2007. Epigallocatechin gallate, the
Rondanelli et al. 2009). None of these early trials have monitored
most active polyphenolic catechin in green tea, presynaptically facilitates
plasma concentrations of ECG and EGCG to assess the uptake of Ca2 + -dependent glutamate release via activation of protein kinase C in rat
green tea polyphenols (Wang et al. 2008) and results of cerebral cortex. Synapse 61, 889–902.
Cooper, R., Morré, D.J., Morré, D.M., 2005. Medicinal benefits of green tea: part I.
investigations that have taken such measures into account are
Review of noncancer health benefits. J. Altern. Complement. Med. 11, 521–528.
still pending (Shen et al. 2009). DalBó, S., Jürgensen, S., Horst, H., Ruzza, A.A., Soethe, D.N., Santos, A.R., Pizzolatti,
In summary, the in vitro targeting of central, and to a lesser M.G., Ribeiro-do-Valle, R.M., 2005. Antinociceptive effect of proanthocyanidins
degree, of peripheral CB by EGCG, EGC and ECG adds to the from Croton celtidifolius bark. J. Pharm. Pharmacol. 57, 765–771.
Dias, K.S., Marques, M.S., Menezes, I.A., Santos, T.C., Silva, A.B., Estevam, C.S.,
complexity of tea catechins’ known multilevel functionalities. Sant’Ana, A.E., Pizza, C., Antoniolli, A.R., Marc- al, R.M., 2007. Antinociceptive
While bioavailability of gallated forms is generally lower than that activity of Maytenus rigida stem bark. Fitoterapia 78, 460–464.
ARTICLE IN PRESS
22 G. Korte et al. / Phytomedicine 17 (2010) 19–22

Diepvens, K., Kovacs, E.M., Nijs, I.M., Vogels, N., Westerterp-Plantenga, M.S., 2005. Murase, T., Haramizu, S., Shimotoyodome, A., Tokimitsu, I., 2006. Reduction of diet-
Effect of green tea on resting energy expenditure and substrate oxidation induced obesity by a combination of tea-catechin intake and regular
during weight loss in overweight females. Br. J. Nutr. 94, 1026–1034. swimming. Int. J. Obes. 30, 561–568.
Furuno, K., Akasako, T., Sugihara, N., 2002. The contribution of the pyrogallol Nagao, T., Meguro, S., Hase, T., Otsuka, K., Komikado, M., Tokimitsu, I., Yamamoto,
moiety to the superoxide radical scavenging activity of flavonoids. Biol. Pharm. T., Yamamoto, K., 2009. A catechin-rich beverage improves obesity and blood
Bull. 25, 19–23. glucose control in patients with type 2 diabetes. Obesity 17, 310–317.
Galve-Roperh, I., Aguado, T., Palazuelos, J., Guzmán, M., 2008. Mechanisms of Okamoto, M., Leung, K.P., Ansai, T., Sugimoto, A., Maeda, N., 2003. Inhibitory effects
control of neuron survival by the endocannabinoid system. Curr. Pharm. Des. of green tea catechins on protein tyrosine phosphatase in Prevotella
14, 2279–2288. intermedia. Oral Microbiol. Immunol. 18, 192–195.
Henning, S.M., Choo, J.J., Heber, D., 2008. Nongallated compared with Pan, T., Jankovic, J., Le, W., 2003. Potential therapeutic properties of green tea
gallated flavan-3-ols in green and black tea are more bioavailable. J. Nutr. polyphenols in Parkinson’s disease. Drugs Aging 20, 711–721.
138, 1529S–1534S. Patra, S.K., Rizzi, F., Silva, A., Rugina, D.O., Bettuzzi, S., 2008. Molecular targets of
Higdon, J.V., Frei, B., 2003. Tea catechins and polyphenols: health effects, (  )-epigallocatechin-3-gallate (EGCG): specificity and interaction with mem-
metabolism, and antioxidant functions. Crit. Rev. Food Sci. Nutr. 43, 89–143. brane lipid rafts. J. Physiol. Pharmacol. 59 (Suppl 9), 217–235.
Hill, A.M., Coates, A.M., Buckley, J.D., Ross, R., Thielecke, F., Howe, P.R., 2007. Can
Richard, D., Guesdon, B., Timofeeva, E., 2009. The brain endocannabinoid system in
EGCG reduce abdominal fat in obese subjects? J. Am. Coll. Nutr. 26, 396S–402S.
the regulation of energy balance. Best Pract. Res. Clin. Endocrinol. Metab. 23,
Hou, D.X., Masuzaki, S., Hashimoto, F., Uto, T., Tanigawa, S., Fujii, M., Sakata, Y.,
17–32.
2007. Green tea proanthocyanidins inhibit cyclooxygenase-2 expression in
Rocha, F.F., Lima-Landman, M.T., Souccar, C., Tanae, M.M., De Lima, T.C., Lapa, A.J.,
LPS-activated mouse macrophages: molecular mechanisms and structure–
2007. Antidepressant-like effect of Cecropia glaziouiSNETH and its constituents –
activity relationship. Arch. Biochem. Biophys. 460, 67–74.
in vivo and in vitro characterization of the underlying mechanism. Phytome-
Hursel, R., Westerterp-Plantenga, M.S., 2009. Green tea catechin plus caffeine
dicine 14, 396–402.
supplementation to a high-protein diet has no additional effect on body weight
maintenance after weight loss. Am. J. Clin. Nutr. 89, 822–830. Rondanelli, M., Opizzi, A., Solerte, S.B., Trotti, R., Klersy, C., Cazzola, R., 2009.
Ikeda, I., Tsuda, K., Suzuki, Y., Kobayashi, M., Unno, T., Tomoyori, H., Goto, H., Administration of a dietary supplement (N-oleyl-phosphatidylethanolamine
Kawata, Y., Imaizumi, K., Nozawa, A., Kakuda, T., 2005. Tea catechins with a and epigallocatechin-3-gallate formula) enhances compliance with diet in
galloyl moiety suppress postprandial hypertriacylglycerolemia by delaying healthy overweight subjects: a randomized controlled trial. Br. J. Nutr. 101,
lymphatic transport of dietary fat in rats. J. Nutr. 135, 155–159. 457–464.
Jeon, S.Y., Bae, K., Seong, Y.H., Song, K.S., 2003. Green tea catechins as a BACE1 Ruidavets, J., Teissedre, P., Ferrie res, J., Carando, S., Bougard, G., Cabanis, J., 2000.
(beta-secretase) inhibitor. Bioorg. Med. Chem. Lett. 13, 3905–3908. Catechin in the Mediterranean diet: vegetable, fruit or wine? Atherosclerosis
Jhaveri, M.D., Richardson, D., Robinson, I., Garle, M.J., Patel, A., Sun, Y., Sagar, D.R., 153, 107–117.
Bennett, A.J., Alexander, S.P., Kendall, D.A., Barrett, D.A., Chapman, V., 2008. Rylski, M., Duriasz-Rowińska, H., Rewerski, W., 1979. The analgesic action of some
Inhibition of fatty acid amide hydrolase and cyclooxygenase-2 increases levels flavonoids in the hot plate test. Acta Physiol. Pol. 30, 385–388.
of endocannabinoid related molecules and produces analgesia via peroxisome Saeki, K., Hayakawa, S., Isemura, M., Miyase, T., 2000. Importance of a pyrogallol-
proliferator-activated receptor-alpha in a model of inflammatory pain. type structure in catechin compounds for apoptosis-inducing activity.
Neuropharmacology 55, 85–93. Phytochemistry 53, 391–394.
Kakuda, T., 2002. Neuroprotective effects of the green tea components theanine Sattayasai, J., Tiamkao, S., Puapairoj, P., 2008. Biphasic effects of Morus alba leaves
and catechins. Biol. Pharm. Bull. 25, 1513–1518. green tea extract on mice in chronic forced swimming model. Phytother. Res.
Kaur, S., Anurag, A., Tirkey, N., Chopra, K., 2005. Reversal of LPS-induced central 22, 487–492.
and peripheral hyperalgesia by green tea extract. Phytother. Res. 19, 39–43. Shen, C.L., Chyu, M.C., Yeh, J.K., Felton, C.K., Xu, K.T., Pence, B.C., Wang, J.S., 2009.
Katavic, P.L., Lamb, K., Navarro, H., Prisinzano, T.E., 2007. Flavonoids as opioid Green tea polyphenols and Tai Chi for bone health: designing a placebo-
receptor ligands: identification and preliminary structure-activity relation- controlled randomized trial. BMC Musculoskelet. Disord. 10, 110.
ships. J. Nat. Prod. 70, 1278–1282. Singal, A., Anjaneyulu, M., Chopra, K., 2005. Modulatory role of green tea extract on
Kao, Y.H., Hiipakka, R.A., Liao, S., 2000. Modulation of obesity by a green tea antinociceptive effect of morphine in diabetic mice. J. Med. Food 8, 386–391.
catechin. Am. J. Clin. Nutr. 72, 1232–1234. Stalmach, A., Troufflard, S., Serafini, M., Crozier, A., 2009. Absorption, metabolism
Keli, S.O., Hertog, M.G., Feskens, E.J., Kromhout, D., 1996. Dietary flavonoids, and excretion of Choladi green tea flavan-3-ols by humans. Mol. Nutr. Food Res.
antioxidant vitamins, and incidence of stroke: the Zutphen study. Arch. Intern. 53 (Suppl. 1), 44–53.
Med. 156, 637–642. Tanabe, N., Suzuki, H., Aizawa, Y., Seki, N., 2008. Consumption of green and roasted
Kim, J.A., Formoso, G., Li, Y., Potenza, M.A., Marasciulo, F.L., Montagnani, M., Quon, teas and the risk of stroke incidence: results from the Tokamachi–Nakasato
M.J., 2007. Epigallocatechin gallate, a green tea polyphenol, mediates NO- cohort study in Japan. Int. J. Epidemiol. 37, 1030–1040.
dependent vasodilation using signaling pathways in vascular endothelium
Tang, L.Q., Wei, W., Wang, X.Y., 2007. Effects and mechanisms of catechin for
requiring reactive oxygen species and Fyn. J. Biol. Chem. 282, 13736–13745.
adjuvant arthritis in rats. Adv. Ther. 24, 679–690.
Kinjo, J., Nagao, T., Tanaka, T., Nonaka, G., Okawa, M., Nohara, T., Okabe, H., 2002.
Tedeschi, E., Suzuki, H., Menegazzi, M., 2002. Antiinflammatory action of EGCG, the
Activity-guided fractionation of green tea extract with antiproliferative activity
main component of green tea, through STAT-1 inhibition. Ann. N. Y. Acad. Sci.
against human stomach cancer cells. Biol. Pharm. Bull. 25, 1238–1240.
973, 435–437.
Kuriyama, S., Hozawa, A., Ohmori, K., Shimazu, T., Matsui, T., Ebihara, S., Awata, S.,
Walker, J.M., Hohmann, A.G., 2005. Cannabinoid mechanisms of pain suppression.
Nagatomi, R., Arai, H., Tsuji, I., 2006. Green tea consumption and cognitive
Handb. Exp. Pharmacol. 168, 509–554.
function: a cross-sectional study from the Tsurugaya Project 1. Am. J. Clin. Nutr.
83, 355–361. Wang, J.S., Luo, H., Wang, P., Tang, L., Yu, J., Huang, T., Cox, S., Gao, W., 2008.
Lynn, A.B., Herkenham, M., 1994. Localization of cannabinoid receptors and Validation of green tea polyphenol biomarkers in a phase II human
nonsaturable high-density cannabinoid binding sites in peripheral tissues of intervention trial. Food Chem. Toxicol. 46, 232–240.
the rat: implications for receptor-mediated immune modulation by cannabi- Wang, X., Song, K.S., Guo, Q.X., Tian, W.X., 2003. The galloyl moiety of green tea
noids. J. Pharmacol. Exp. Ther. 268, 1612–1623. catechins is the critical structural feature to inhibit fatty-acid synthase.
Maeda-Yamamoto, M., Ema, K., Shibuichi, I., 2007. In vitro and in vivo anti-allergic Biochem. Pharmacol. 66, 2039–2047.
effects of ‘benifuuki’ green tea containing O-methylated catechin and ginger Warden, B.A., Smith, L.S., Beecher, G.R., Balentine, D.A., Clevidence, B.A., 2001.
extract enhancement. Cytotechnology 55, 135–142. Catechins are bioavailable in men and women drinking black tea throughout
Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, A.C., Bonner, T.I., 1990. Structure the day. J. Nutr. 131, 1731–1737.
of a cannabinoid receptor and functional expression of the cloned cDNA. Woelkart, K., Salo-Ahen, O.M., Bauer, R., 2008. CB receptor ligands from plants.
Nature 346, 561–564. Curr. Top. Med. Chem. 8, 173–186.
Munro, S., Thomas, K.L., Abu-Shaar, M., 1993. Molecular characterization of a Wolfe, K.L., Liu, R.H., 2008. Structure-activity relationships of flavonoids in the
peripheral receptor for cannabinoids. Nature 365, 61–65. cellular antioxidant activity assay. J. Agric. Food Chem. 56, 8404–8411.

You might also like