Journal Home > Volume 11 , Issue 3

(–)-Epicatechin-3-gallate (ECG), a bioactive polyphenolic compound, has contributed a lot to the health benefits of green tea. Great attention has been focused on (–)-epigallocatechin-3-gallate (EGCG), but limited research has been performed towards ECG. Like EGCG, ECG also possesses various pharmacological and physiological properties, such as mediation of antioxidant activities, anti-inflammation response, regulation of cell proliferation and apoptosis, as well as anticancer properties during angiogenesis, invasion and metastasis stages. Nontoxic ECG has various molecular targets within the cells, including CYP enzymes, phase Ⅱ detoxification and antioxidant enzymes, as well as pro-inflammatory mediators. The antineoplastic mechanism contains inhibition of phase 1 CYP enzymes, induction of phase Ⅱ detoxification and antioxidant enzymes, high anti-inflammatory efficacy, arrest of cell cycle progression, regulation of apoptosis, as well as mediation of metastasis processes. In particular, the gallate moiety of ECG is critical for mediating inhibitory effects towards cancer cells. Besides regulation of intracellular signaling pathways, ECG also inhibits RNase A and matrix metalloproteinase enzymatic activity via chelating metals (copper and zinc) in cancer cells. This review has summarized recent studies on pharmacological properties of ECG, and discussed corresponding mechanism on modulation of cellular signaling events by ECG, hoping to broaden its multiple usage.


menu
Abstract
Full text
Outline
About this article

Health promoting activities and corresponding mechanism of (–)-epicatechin-3-gallate

Show Author's information Zhiliang Lia,bChangchun FengaHongjin DongaWeibin JinaWenying ZhangbJianfeng Zhana( )Shuzhen Wanga( )
College of Life Science, Huanggang Normal University; Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization; Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains; Hubei Zhongke Research Institute of Industrial Technology, Huanggang 438000, China
College of Agriculture, Yangtze University, Jingzhou 434000, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Abstract

(–)-Epicatechin-3-gallate (ECG), a bioactive polyphenolic compound, has contributed a lot to the health benefits of green tea. Great attention has been focused on (–)-epigallocatechin-3-gallate (EGCG), but limited research has been performed towards ECG. Like EGCG, ECG also possesses various pharmacological and physiological properties, such as mediation of antioxidant activities, anti-inflammation response, regulation of cell proliferation and apoptosis, as well as anticancer properties during angiogenesis, invasion and metastasis stages. Nontoxic ECG has various molecular targets within the cells, including CYP enzymes, phase Ⅱ detoxification and antioxidant enzymes, as well as pro-inflammatory mediators. The antineoplastic mechanism contains inhibition of phase 1 CYP enzymes, induction of phase Ⅱ detoxification and antioxidant enzymes, high anti-inflammatory efficacy, arrest of cell cycle progression, regulation of apoptosis, as well as mediation of metastasis processes. In particular, the gallate moiety of ECG is critical for mediating inhibitory effects towards cancer cells. Besides regulation of intracellular signaling pathways, ECG also inhibits RNase A and matrix metalloproteinase enzymatic activity via chelating metals (copper and zinc) in cancer cells. This review has summarized recent studies on pharmacological properties of ECG, and discussed corresponding mechanism on modulation of cellular signaling events by ECG, hoping to broaden its multiple usage.

Keywords: (–)-Epicatechin-3-gallate, Polyphenolic compounds, Pharmacological properties, Anticarcinogenic agent, Natural antioxidants, Antimicrobial properties

References(111)

[1]

W.C. Reygaert, Green tea catechins: their use in treating and preventing infectious diseases, BioMed Res. Int. 2018 (2018) 9105261. https://doi.org/10.1155/2018/9105261.

[2]

X. Zhang, J. Li, Y. Li, et al., Anti-melanogenic effects of epigallocatechin-3-gallate (EGCG), epicatechin-3-gallate (ECG) and gallocatechin-3-gallate (GCG) via down-regulation of cAMP/CREB/MITF signaling pathway in B16F10 melanoma cells, Fitoterapia 145 (2020) 104634. https://doi.org/10.1016/j.fitote.2020.104634.

[3]

M.H. Ravindranath, V. Ramasamy, S. Moon, et al., Differential growth suppression of human melanoma cells by tea (Camellia sinensis) epicatechins (ECG, EGC and EGCG), Complement Alternat. Med. 6 (2009) 523. https://doi.org/10.1093/ecam/nem140.

[4]

T.D. Way, H.Y. Lin, D.H. Kuo, et al., Pu-erh tea attenuates hyperlipogenesis and induces hepatoma cells growth arrest through activating AMP-activated protein kinase (AMPK) in human HepG2 cells, J. Agric. Food Chem. 57 (2009) 5257. https://doi.org/10.1021/jf900730e.

[5]

A. Zinellu, S. Sotgia, B. Scanu, et al., Human serum albumin increases the stability of green tea catechins in aqueous physiological conditions, PLoS One 10 (2015) e0134690. https://doi.org/10.1371/journal.pone.0134690.

[6]

L. Piwowarczyk, M. Stawny, D.T. Mlynarczyk, et al., Role of curcumin and (–)-epigallocatechin-3-O-gallate in bladder cancer treatment: a review, Cancers 12 (2020) 1801. https://doi.org/10.3390/cancers12071801.

[7]

M.H. Pan, Y.S. Chiou, Y.J. Wang, et al., Multistage carcinogenesis process as molecular targets in cancer chemoprevention by epicatechin-3-gallate, Food Funct. 2 (2011) 101. https://doi.org/10.1039/c0fo00174k.

[8]

A. Sourabh, S.S. Kanwar, R.G. Sud, et al., Influence of phenolic compounds of Kangra tea [Camellia sinensis (L) O Kuntze] on bacterial pathogens and indigenous bacterial probiotics of Western Himalayas, Braz. J. Microbiol. 44 (2013) 709-715. https://doi.org/10.1590/S1517-83822013000300007.

[9]

J.B. Vaidyanathan, T. Walle, Cellular uptake and efflux of the tea flavonoid (–)-epicatechin-3-gallate in the human intestinal cell line Caco-2, J. Pharmacol. Exp. Ther. 307 (2003) 745. https://doi.org/10.1124/jpet.103.054296.

[10]

Y. Kawai, H. Tanaka, K. Murota, et al., (−)-Epicatechin gallate accumulates in foamy macrophages in human atherosclerotic aorta: implication in the anti-atherosclerotic actions of tea catechins, Biochem. Biophys. Res. Commun. 374 (2008) 527. https://doi.org/10.1016/j.bbrc.2008.07.086.

[11]

M. Friedman, C.E. Levin, S.U. Lee, et al., Stability of green tea catechins in commercial tea leaves during storage for 6 Months, J. Food Sci. 74 (2009) H47. https://doi.org/10.1111/j.1750-3841.2008.01033.x.

[12]

J. Kochman, K. Jakubczyk, J. Antoniewicz, et al., Health benefits and chemical composition of matcha green tea: a review, Molecules 26 (2021) 85. https://doi.org/10.3390/molecules26010085.

[13]

A.N. Tony Kong, E. Owuor, R. Yu, et al., Induction of xenobiotic enzymes by the map kinase pathway and the antioxidant or electrophile response element (ARE/EpRE), Drug Metab. Rev. 33 (2001) 255-271. https://doi.org/10.1081/DMR-120000652.

[14]

J.A. Joyce, J.W. Pollard, Microenvironmental regulation of metastasis, Nat. Rev. Cancer 9 (2009) 239. https://doi.org/10.1038/nrc2618.

[15]

F. Mohd, O. Mohammad, C. Sandesh, et al., Mobilization of nuclear copper by green tea polyphenol epicatechin-3-gallate and subsequent prooxidant breakage of cellular DNA: implications for cancer chemotherapy, Int. J. Mol. Sci. 18 (2017) 34. https://doi.org/10.3390/ijms18010034.

[16]

M.A. Morse, G.D. Stoner, Cancer chemoprevention: principles and prospects, Carcinogenesis 14 (1993) 1737-1746. https://doi.org/10.1093/carcin/14.9.1737.

[17]

X. Tan, D. Hu, S. Li, et al., Differences of four catechins in cell cycle arrest and induction of apoptosis in LoVo cells, Cancer Lett. 158 (2000) 1-6. https://doi.org/10.1016/S0304-3835(00)00445-6.

[18]

L. Chen, X. Yang, H. Jiao, Tea catechins protect against lead-induced cytotoxicity, lipid peroxidation, and membrane fluidity in HepG2 cells, Toxicol. Sci. 69 (2002) 149-156. https://doi.org/10.1093/toxsci/69.1.149.

[19]

T.M. Elattar, A.S. Virji, Effect of tea polyphenols on growth of oral squamous carcinoma cells in vitro, Anticancer Res. 20 (2000) 3459.

[20]

G.Y. Yang, J. Liao, K. Kim, et al., Inhibition of growth and induction of apoptosis in human cancer cell lines by tea polyphenols, Carcinogenesis 19 (1998) 611-616. https://doi.org/10.1093/carcin/19.4.611.

[21]

S. Okabe, M. Suganuma, M. Hayashi, et al., Mechanisms of growth inhibition of human lung cancer cell line, PC-9, by tea polyphenols, Jpn. J. Cancer Res. 88 (1997) 639-643. https://doi.org/10.1111/j.1349-7006.1997.tb00431.x.

[22]

Z.Y. Wang, M. Das, D.R. Bickers, et al., Interaction of epicatechins derived from green tea with rat hepatic cytochrome P-450, Drug Metab. Dispos. 16 (1988) 98.

[23]

V.E. Steele, G.J. Kelloff, D. Balentine, et al., Comparative chemopreventive mechanisms of green tea, black tea and selected polyphenol extracts measured by in vitro bioassays, Carcinogenesis 21 (2000) 63-67. https://doi.org/10.1093/carcin/21.1.63.

[24]

H.J. Kim, J.H. Ryu, C.H. Kim, Epicatechin gallate suppresses oxidative stress-induced MUC5AC overexpression by interaction with epidermal growth factor receptor, Am. J. Resp. Cell Mol. 43 (2010) 349-357. https://doi.org/10.1165/rcmb.2009-0205OC.

[25]

D.A. Mir, U. Mehraj, N.A. Wani, et al., Tumor microenvironment promotes breast cancer chemoresistance, Cancer Chemoth. Pharm. 87 (2021) 147-158. https://doi.org/10.1007/s00280-020-04222-w.

[26]

C. Santangelo, R. Vari, B. Scazzocchio, et al., Polyphenols, intracellular signalling and inflammation, Ann. Ist. Super. Sanita 43 (2007) 394-405.

[27]

J. Hong, T.J. Smith, C.T. Ho, et al., Effects of purified green and black tea polyphenols on cyclooxygenase- and lipoxygenase-dependent metabolism of arachidonic acid in human colon mucosa and colon tumor tissues, Biochem. Pharmacol. 62 (2001) 1175-1183. https://doi.org/10.1016/S0006-2952(01)00767-5.

[28]

C.C. Huang, J.Y. Fang, W.B. Wu, et al., Protective effects of (–)-epicatechin -3-gallate on UVA-induced damage in HaCaT keratinocytes, Arch. Dermatol. Res. 296 (2005) 473. https://doi.org/10.1007/s00403-005-0540-5.

[29]

M.A. el-Saadany, H.M. Rawel, J. Raila, Antioxidants modulate the IL-6 induced inhibition of negative acute-phase protein secretion in HepG2 cells, Cell Biochem. Funct. 26 (2008) 95-101. https://doi.org/10.1002/cbf.1405.

[30]

Y. Hosokawa, I. Hosokawa, K. Ozaki, et al., Catechins inhibit CCL20 production in IL-17A-stimulated human jingival fibroblasts, Cell. Physiol. Biochem. 24 (2009) 391-396. https://doi.org/10.1159/000257431.

[31]

M.T. Huang, C.T. Ho, Z.Y. Wang, et al., Inhibitory effect of topical application of a green tea polyphenol fraction on tumor initiation and promotion in mouse, Carcinogenesis 13 (1992) 947-954. https://doi.org/10.1093/carcin/13.6.947.

[32]

Y.S. Chiou, Q.R. Huang, C.T. Ho, Directly interact with keap1 and lps is involved in the anti-inflammatory mechanisms of (–)-epicatechin-3-gallate in lps- induced macrophages and endotoxemia, Free Radical Bio. Med. 94 (2016) 1-16. https://doi.org/10.1016/j.freeradbiomed.2016.02.010.

[33]

T. Nakanishi, K. Mukai, H. Yumoto, et al., Anti-inflammatory effect of catechin on cultured human dental pulp cells affected by bacteria-derived factors, Eur. J. Oral Sci. 118 (2010) 145-150. https://doi.org/10.1111/j.1600-0722.2010.00714.x.

[34]

T. Nakanishi, K. Mukai, Y. Hosokawa, et al., Catechins inhibit vascular endothelial growth factor production and cyclooxygenase-2 expression in human dental pulp cells, Int. Endod. J. 48 (2015) 277-282. https://doi.org/10.1111/iej.12312.

[35]

I. Collins, M.D. Garrett, Targeting the cell division cycle in cancer: CDK and cell cycle checkpoint kinase inhibitors, Curr. Opin. Pharmacol. 5 (2005) 366-373. https://doi.org/10.1016/j.coph.2005.04.009.

[36]

S. Manna, S. Mukherjee, A. Roy, et al., Tea polyphenols can restrict benzo[a]pyrene-induced lung carcinogenesis by altered expression of p53-associated genes and H-ras, c-myc and cyclin D1, J. Nutr. Biochem. 20 (2009) 337-349. https://doi.org/10.1016/j.jnutbio.2008.04.001.

[37]

Y.C. Lim, S.H. Lee, M.H. Song, et al., Growth inhibition and apoptosis by (−)-epicatechin gallate are mediated by cyclin D1 suppression in head and neck squamous carcinoma cells, Eur. J. Cancer 42 (2006) 3260-3266. https://doi.org/10.1016/j.ejca.2006.07.014.

[38]

N. Fujimoto, N. Sueoka, E. Sueoka, Lung cancer prevention with (–)-epigallocatechin gallate using monitoring by heterogeneous nuclear ribonucleoprotein B1, Int. J. Oncol. 20 (2002) 1233-1239. https://doi.org/10.1002/ijc.10400.

[39]

I. Cordero-Herrera, M.A. Martín, L. Bravo, et al., Epicatechin gallate induces cell death via p53 activation and stimulation of p38 and JNK in human colon cancer SW480 cells, Nutr. Cancer 65 (2013) 718-728. https://doi.org/10.1080/01635581.2013.795981.

[40]

M.G. Goodin, K.C. Fertuck, T.R. Zacharewski, et al., Estrogen receptor-mediated actions of polyphenolic catechins in vivo and in vitro, Toxicol. Sci. 69 (2002) 354-361. https://doi.org/10.1093/toxsci/69.2.354.

[41]

E. Shaulian, AP-1 — the Jun proteins: oncogenes or tumor suppressors in disguise? Cell. Signalling 22 (2010) 894-899. https://doi.org/10.1016/j.cellsig.2009.12.008.

[42]

A. Sachinidis, C. Seul, S. Seewald, Green tea compounds inhibit tyrosine phosphorylation of PDGF β-receptor and transformation of A172 human glioblastoma, FEBS Lett. 471 (2000) 51-55. https://doi.org/10.1016/S0014-5793(00)01360-0.

[43]

J. Jodoin, M. Demeule, R. Béliveau, Inhibition of the multidrug resistance P-glycoprotein activity by green tea polyphenols, Biochim. Biophys. Acta 1542 (2002) 149-159. https://doi.org/10.1016/S0167-4889(01)00175-6.

[44]

G. Liang, A. Tang, X. Lin, et al., Green tea catechins augment the antitumor activity of doxorubicin in an in vivo mouse model for chemoresistant liver cancer, Int. J. Oncol. 37 (2010) 111-123. https://doi.org/10.3892/ijo-00000659.

[45]

S. Kitagawa, T. Nabekura, S. Kamiyama, Inhibition of P-glycoprotein function by tea catechins in KB-C2 cells, J. Pharm. Pharmacol. 56 (2004) 1001-1005. https://doi.org/10.1211/0022357044003.

[46]

S. Elmore, Apoptosis: a review of programmed cell death, Toxicol. Pathol. 35 (2007) 495-516. https://doi.org/10.1080/01926230701320337.

[47]

G. Klein, Cancer, apoptosis, and nonimmune surveillance, Cell Death. Differ. 11 (2004) 13-17. https://doi.org/10.1038/sj.cdd.4401342.

[48]

S. Okabe, Y. Ochiai, M. Aida, et al., Mechanistic aspects of green tea as a cancer preventive: effect of components on human stomach cancer cell Lines, Jpn. J. Cancer Res. 90 (1999) 733-739. https://doi.org/10.1111/j.1349-7006.1999.tb00808.x.

[49]

M. Hessien, S. El-Gendy, T. Donia, et al., Growth inhibition of human non-small lung cancer cells H460 by green tea and ginger polyphenols, Anticancer Agents Med. Chem. 12 (2012) 383-390. https://doi.org/10.2174/187152012800228698.

[50]

S.J. Baek, J.S. Kim, F.R. Jackson, et al., Epicatechin gallate-induced expression of NAG-1 is associated with growth inhibition and apoptosis in colon cancer cells, Carcinogenesis 25 (2004) 2425-2432. https://doi.org/10.1093/carcin/bgh255.

[51]

L.Y. Chung, T.C. Cheung, S.K. Kong, et al., Induction of apoptosis by green tea catechins in human prostate cancer DU145 cells, Life Sci. 68 (2001) 1207-1214. https://doi.org/10.1016/S0024-3205(00)01020-1.

[52]

H. Babich, M.E. Krupka, H.A. Nissim, et al., Differential in vitro cytotoxicity of (−)-epicatechin gallate (ECG) to cancer and normal cells from the human oral cavity, Toxicol. in Vitro 19 (2005) 231-242. https://doi.org/10.1016/j.tiv.2004.09.001.

[53]

Y. Cai, H.Q. Hong, L. Zhao, et al., (−)-Epicatechin-3-gallate (a polyphenol from green tea) potentiates doxorubicin-induced apoptosis in H9C2 cardiomyocytes, Biotechnol. Lett. 37 (2015) 193943. https://doi.org/10.1007/s10529-015-1879-0.

[54]

M. Egeblad, Z. Werb, New functions for the matrix metalloproteinases in cancer progression, Nat. Rev. Cancer 2 (2002) 161-174. https://doi.org/10.1038/nrc745.

[55]

H. Oneda, M. Shiihara, K. Inouye, Inhibitory effects of green tea catechins on the activity of human matrix metalloproteinase 7 (matrilysin), J. Biochem. 133 (2003) 571-576. https://doi.org/10.1093/jb/mvg073.

[56]

M. Maeda-Yamamoto, H. Kawahara, N. Tahara, et al., Effects of tea polyphenols on the invasion and matrix metalloproteinases activities of human fibrosarcoma HT1080 cells, J. Agric. Food Chem. 47 (1999) 2350-2354. https://doi.org/10.1021/jf9811525.

[57]

M. Isemura, Y. Suzuki, K. Satoh, et al., Effects of catechins on the mouse lung carcinoma cell adhesion to the endothelial cells, Cell Biol. Int. 17 (1993) 559-564. https://doi.org/10.1006/cbir.1993.1099.

[58]

K. Ogata, N. Mukae, Y. Suzuki, et al., Effects of catechins on the mouse tumor cell adhesion to fibronectin, Planta Med. 61 (1995) 472-474. https://doi.org/10.1055/s-2006-958140.

[59]

E.B. Jasser, O. Min-Ho, A. Patrick, et al., Catechins prevent vascular smooth muscle cell invasion by inhibiting MT1-MMP activity and MMP-2 expression, Cardiovasc. Res. 67 (2005) 317-325. https://doi.org/10.1016/j.cardiores.2005.03.017.

[60]

D. Duhon, R.L.H. Bigelow, D.T. Coleman, et al., The polyphenol epigallocatechin-3-gallate affects lipid rafts to block activation of the c-Met receptor in prostate cancer cells, Mol. Carcinog. 49 (2010) 739-749. https://doi.org/10.1002/mc.20649.

[61]

S.F. Huang, C.T. Horng, Y.S. Hsieh, et al., Epicatechin-3-gallate reverses TGF-β1-induced epithelial-tomesenchymal transition and inhibits cell invasion and protease activities in human lung cancer cells, Food Chem. Toxicol. 94 (2016) 1-10. https://doi.org/10.1016/j.fct.2016.05.009.

[62]

C. Chen, R. Yu, E.D. Owuor, et al., Activation of antioxidant response element (ARE), mitogen-activated protein kinases (MAPKs) and caspases by major green tea polyphenol components during cell survival and death, Arch. Pharm. Res. 23 (2000) 605-612. https://doi.org/10.1007/BF02975249.

[63]

C. Murakami, Y. Hirakawa, H. Inui, et al., Effect of tea catechins on cellular lipid peroxidation and cytotoxicity in HepG2 cells, Biosci. Biotechnol. Biochem. 66 (2002) 1559-1562. https://doi.org/10.1271/bbb.66.1559.

[64]

C.H. Coyle, B.J. Philips, S.N. Morrisroe, Antioxidant effects of green tea and its polyphenols on bladder cells, Life Sci. 83 (2008) 12-18. https://doi.org/10.1016/j.lfs.2008.04.010.

[65]

I. Rodríguez-Ramiro, M. Á. Martín, S. Ramos, Comparative effects of dietary flavanols on antioxidant defences and their response to oxidant-induced stress on Caco2 cells, Eur. J. Nutr. 50 (2011) 313-322. https://doi.org/10.1007/s00394-010-0139-2.

[66]

S.S. Liao, R.A. Hiipakka, Selective-inhibition of steroid 5 α-reductase isozymes by tea epicatechin-3-gallate and epigallocatechin-3-gallate, Biochem. Biophys. Res. Commun. 214 (1995) 833-838. https://doi.org/10.1006/bbrc.1995.2362.

[67]

X. Wang, K.S. Song, Q.X. Guo, et al., The galloyl moiety of green tea catechins is the critical structural feature to inhibit fatty-acid synthase, Biochem. Pharmacol. 66 (2003) 2039-2047. https://doi.org/10.1016/S0006-2952(03)00585-9.

[68]

H. Ge, J. Liu, W. Zhao, et al., Mechanistic studies for tri-targeted inhibition of enzymes involved in cholesterol biosynthesis by green tea polyphenols, Org. Biomol. Chem. 12 (2014) 4941-4951. https://doi.org/10.1039/c4ob00589a.

[69]

D. Hayashi, S. Ueda, M. Yamanoue, Epigallocatechin-3-gallate activates diacylglycerol kinase alpha via a 67 kDa laminin receptor: a possibility of galloylated catechins as functional food to prevent and/or improve diabetic renal dysfunctions, J. Funct. Foods 15 (2015) 561-569. https://doi.org/10.1016/j.jff.2015.04.005.

[70]

Y. Yang, L. Qiao, X. Zhang, et al., Effect of methylated tea catechins from Chinese oolong tea on the proliferation and differentiation of 3T3-L1 preadipocyte. Fitoterapia 104 (2015) 45-49. https://doi.org/10.1016/j.fitote.2015.05.007.

[71]

W. Zhu, B. Zou, R. Nie, et al., A-type ECG and EGCG dimers disturb the structure of 3T3-L1 cell membrane and strongly inhibit its differentiation by targeting peroxisome proliferator-activated receptor gamma with miR-27 involved mechanism, J. Nutr. Biochem. 26 (2015) 1124-1135.

[72]

W. Zhu, L. Xiong, J. Peng, et al., Structure-dependent membrane-perturbing potency of four proanthocyanidin dimers on 3T3-l1 preadipocytes, J. Agr. Food Chem. 64 (2016) 7022-7032. https://doi.org/10.1021/acs.jafc.6b02671.

[73]

W. Zhu, X. Deng, J. Peng, et al., A-type ECG and EGCG dimers inhibit 3T3-L1 differentiation by binding to cholesterol in lipid rafts, J. Nutr. Biochem. 48 (2017) 62-73. https://doi.org/10.1016/j.jnutbio.2017.06.012.

[74]

H.M. Lo, C.F. Hung, Y.Y. Huang, et al., Tea polyphenols inhibit rat vascular smooth muscle cell adhesion and migration on collagen and laminin via interference with cell–ECM interaction, J. Biomed. Sci. 14 (2007) 637-645. https://doi.org/10.1007/s11373-007-9170-6.

[75]

A.Z. Wu, S.H. Loh, T.H. Cheng, et al., Antiarrhythmic effects of (−)-epicatechin-3-gallate, a novel sodium channel agonist in cultured neonatal rat ventricular myocyte, Biochem. Pharmacol. 85 (2013) 69-80. https://doi.org/10.1016/j.bcp.2012.10.003.

[76]

Y. Qi, C. Yang, Z. Jiang, et al., Epicatechin-3-gallate signaling and protection against cardiac ischemia/reperfusion injury, J. Pharmacol. Exp. Ther. 371 (2019) 663-674. https://doi.org/10.1124/jpet.119.260117.

[77]

S.D. Hettiarachchi, Y. Zhou, E. Seven, et al., Nanoparticle-mediated approaches for Alzheimer's disease pathogenesis, diagnosis, and therapeutics, J. Control. Release 314 (2019) 125-140. https://doi.org/10.1016/j.jconrel.2019.10.034.

[78]

X. Li, X. Du, J. Ni, Zn2+ aggravates tau aggregation and neurotoxicity, Int. J. Mol. Sci. 20 (2019) 487. https://doi.org/10.3390/ijms20030487.

[79]

T. Chen, Y. Yang, S. Zhu, et al., Inhibition of Aβ aggregates in Alzheimer's disease by epigallocatechin and epicatechin-3-gallate from green tea, Bioorg. Chem. 105 (2020) 104382. https://doi.org/10.1016/j.bioorg.2020.104382.

[80]

S. Perumal, R. Mahmud, S. Ismail, Mechanism of action of isolated caffeic acid and epicatechin 3-gallate from euphorbia hirta against pseudomonas aeruginosa, Pharmacogn. Mag. 13 (2017) S311-S315. https://doi.org/10.4103/pm.pm_309_15.

[81]

L. Chen, B. Yu, Y. Zhang, et al., Bioactivity-guided fractionation of an antidiarrheal Chinese herb Rhodiola kirilowii (Regel) maxim reveals (–)-epicatechin-3-gallate and (–)-epigallocatechin-3-gallate as inhibitors of cystic fibrosis transmembrane conductance regulator, PLoS One 10 (2015) e0119122. https://doi.org/10.1371/journal.pone.0119122.

[82]

J. Xu, Z. Xu, W. Zheng, et al., A review of the antiviral role of green tea catechins, Molecules 22 (2017) 1337. https://doi.org/10.3390/molecules22081337.

[83]

M. Aboulaila, N. Yokoyama, I. Igarashi, Inhibitory effects of (–)-Epigallocatechin-3-gallate from green tea on the growth of Babesia parasites, Parasitology 137 (2010) 785-791. https://doi.org/10.1017/S0031182009991594.

[84]

J.D.F. Inacio, L. Gervazoni, M.M. Canto-Cavalheiro, et al., The effect of (–)-epigallocatechin 3-O-gallate in vitro and in vivo in Leishmania braziliensis: involvement of reactive oxygen species as a mechanism of action, PLoS Neglect. Trop. D. 8 (2014) e3093. https://doi.org/10.1371/journal.pntd.0003093.

[85]

P. Thipubon, C. Uthaipibull, S. Kamchonwongpaisan, et al., Inhibitory effect of novel iron chelator, 1-(N-acetyl-6- aminohexyl)-3-hydroxy-2-methylpyridin-4-one (CM1) and green tea extract on growth of Plasmodium falciparum, Malaria J. 14 (2015) 382. https://doi.org/10.1186/s12936-015-0910-1.

[86]

M. Muthu, J. Gopal, S.X. Min, et al., Green tea versus traditional Korean teas: antibacterial/antifungal or both? Appl. Biochem. Biotech. 180 (2016) 780-790. https://doi.org/10.1007/s12010-016-2132-6.

[87]

Y. Ning, J. Ling, C.D. Wu, Synergistic effects of tea catechin epigallocatechin gallate and antimycotics against oral Candida species, Arch. Oral Biol. 60 (2015) 1565-1570. https://doi.org/10.1016/j.archoralbio.2015.07.001.

[88]

B.E. Roberts, M.L. Duennwald, H. Wang, et al., A synergistic small-molecule combination directly eradicates diverse prion strain structures, Nat. Chem. Biol. 5 (2010) 936-946. https://doi.org/10.1038/nchembio.246.

[89]

J. Ju, G. Lu, J.D. Lambert, et al., Inhibition of carcinogenesis by tea constituents, Cancer Biol. 17 (2007) 395-402. https://doi.org/10.1016/j.semcancer.2007.06.013.

[90]

M. Skrt, E. Benedik, C. Podlipnik, et al., Interactions of different polyphenols with bovine serum albumin using fluorescence quenching and molecular docking, Food Chem. 135 (2012) 2418-2424. https://doi.org/10.1016/j.foodchem.2012.06.114.

[91]

R.D.P. Stanzel, S. Lourenssen, D.G. Nair, et al., Mitogenic factors promoting intestinal smooth muscle cell proliferation, Am. J. Physiol. Cell Physiol. 299 (2010) C805-C817. https://doi.org/10.1152/ajpcell.00086.2010.

[92]

D. Trachootham, Y. Zhou, H. Zhang, et al., Selective killing of oncogenically transformed cells through a ROS-mediated mechanism by β-phenylethylisothiocyanate, Cancer Cell 10 (2006) 241–252. https://doi.org/10.1016/j.ccr.2006.08.009.

[93]

F. Ceciliani, A. Giordano, V. Spagnolo, The systemic reaction during inflammation: the acute-phase proteins, Protein Pept. Lett. 9 (2002) 211-223. https://doi.org/10.2174/0929866023408779.

[94]

T.E. Kim, K.H. Shin, J.E. Park, et al., Effect of green tea catechins on the pharmacokinetics of digoxin in humans. Drug Des. Dev. Ther. 12 (2018) 2139-2147. https://doi.org/10.2147/DDDT.S148257.

[95]

A. Araghizadeh, J. Kohanteb, M.M. Fani, Inhibitory activity of green tea (Camellia sinensis) extract on some clinically isolated cariogenic and periodontopathic bacteria, Med. Prin. Pract. 22 (2013) 368-372. https://doi.org/10.1159/000348299.

[96]

M.D.R. De Moraes, J.R.M. Carneiro, V.F. Passos, et al., Effect of green tea as a protective measure against dental erosion in coronary dentine, Braz. Oral Res. 30 (2016) S1806. https://doi.org/10.1590/1807-3107BOR-2016.vol30.0013.

[97]

Y. Wang, S. Ma, Recent advances in inhibitors of bacterial fatty acid synthesis type II (FASII) system enzymes as potential antibacterial agents, Chemmedchem 8 (2013) 1589-1608. https://doi.org/10.1002/cmdc.201300209.

[98]

W.C. Reygaert, The antimicrobial possibilities of green tea, Front. Microbiol. 5 (2014) 434. https://doi.org/10.3389/fmicb.2014.00434.

[99]

M. Spina, M. Cuccioloni, M. Mozzicafreddo, et al., Mechanism of inhibition of wt-dihydrofolate reductase from E. coli by tea epigallocatechin-gallate, Proteins 72 (2008) 240-251. https://doi.org/10.1002/prot.21914.

[100]

N. Chinnam, P.K. Dadi, S.A. Sabri, et al., Dietary bioflavonoids inhibit Escherichia coli ATP synthase in a differential manner, Int. J. Biol. Macromol. 46 (2010) 478-486. https://doi.org/10.1016/j.ijbiomac.2010.03.009.

[101]

Q. Fu, Q. Li, X. Lin, et al., Antidiabetic effects of tea, Molecules 22 (2017) 849. https://doi.org/10.3390/molecules22050849.

[102]

W. Dai, C. Ruan, Y. Zhang, et al., Bioavailability enhancement of EGCG by structural modification and nano-delivery: a review, J. Funct. Foods 65 (2020) 103732. https://doi.org/10.1016/j.jff.2019.103732.

[103]

J. Wang, L. Tang, J.S. Wang, Biomarkers of dietary polyphenols in cancer studies: current evidence and beyond, Oxid. Med. Cell. Longev. 2015 (2015) 732302. https://doi.org/10.1155/2015/732302.

[104]

X. Jin, M. Liu, Z. Chen, et al., Separation and purification of epigallocatechin-3-gallate (EGCG) from green tea using combined macroporous resin and polyamide column chromatography, J. Chromatogr. B 1002 (2015) 113-122. https://doi.org/10.1016/j.jchromb.2015.07.055.

[105]

X. Wang, W. Dong, X. Zhang, et al., Antiviral mechanism of tea polyphenols against Porcine reproductive and respiratory syndrome virus, Pathogens 10 (2021) 202. https://doi.org/10.3390/pathogens10020202.

[106]

H. Tian, D. Xiang, C. Li, Tea polyphenols encapsulated in W/O/W emulsions with xanthan gum–locust bean gum mixture: evaluation of their stability and protection, Int. J. Biol. Macromol. 175 (2021) 40-48. https://doi.org/10.1016/j.ijbiomac.2021.01.161.

[107]
J. Shang, F. Zhong, S. Zhu, D. et al., Formation, structural characteristics and physiochemical properties of beeswax oleogel prepared with tea polyphenols loaded gelator, Food Funct. 12 (2021) 1662-1671. https://doi.org/10.1039/D0FO02772C.
[108]

W.R. Wang, P.L. Zhang, J. Ou, et al., Selective extraction of ECG from tea polyphenols by one step in centrifugal contactor separators: modeling and application, Ind. Eng. Chem. Res. 58 (2019) 2027-2035. https://doi.org/10.1021/acs.iecr.8b06042.

[109]

C. Chu, J. Deng, Y. Hou, et al., Application of PEG and EGCG modified collagen-base membrane to promote osteoblasts proliferation, Mater. Sci. Eng. C Mater. Biol. 76 (2017) 31-36. https://doi.org/10.1016/j.msec.2017.02.157.

[110]

C. Chu, Y. Wang, Y. Wang, et al., Evaluation of epigallocatechin-3-gallate (EGCG) modified collagen in guided bone regeneration (GBR) surgery and modulation of macrophage phenotype, Mater. Sci. Eng. C Mater. Biol. 99 (2019) 73-82. https://doi.org/10.1016/j.msec.2019.01.083.

[111]

C. Chu, L. Liu, Y. Wang, et al., Evaluation of epigallocatechin-3-gallate (EGCG)-modified scaffold determines macrophage recruitment, Mater. Sci. Eng. C Mater. Biol. 100 (2019) 505-513. https://doi.org/10.1016/j.msec.2019.03.007.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 31 January 2021
Revised: 01 April 2021
Accepted: 02 April 2021
Published: 04 February 2022
Issue date: May 2022

Copyright

© 2022 Beijing Academy of Food Sciences.

Acknowledgements

Acknowledgement

This research reported in this paper are funded by Hubei Science and Technology Plan Key Project (G2019ABA100), open fund of Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization (201932103), and fund from Assessment and Comprehensive Utilization of Characteristic Biological resources in Dabie Mountains (4022019006).

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return