Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Feb 1.
Published in final edited form as: J Nutr Biochem. 2016 May 27;40:1–13. doi: 10.1016/j.jnutbio.2016.05.005

Therapeutic properties of green tea against environmental insults

Lixia Chen 1, Huanbiao Mo 2, Ling Zhao 3, Weimin Gao 4, Shu Wang 5, Meghan M Cromie 4, Chuanwen Lu 1, Jia-Sheng Wang 6, Chwan-Li Shen 7,*
PMCID: PMC5124528  NIHMSID: NIHMS822017  PMID: 27723473

Abstract

Pesticides, smoke, mycotoxins, polychlorinated biphenyls, and arsenic are the most common environmental toxins and toxicants to humans. These toxins and toxicants may impact on human health at the molecular (DNA, RNA, or protein), organelle (mitochondria, lysosome, or membranes), cellular (growth inhibition or cell death), tissue, organ, and systemic levels. Formation of reactive radicals, lipid peroxidation, inflammation, genotoxicity, hepatotoxicity, embryotoxicity, neurological alterations, apoptosis, and carcinogenic events are some of the mechanisms mediating the toxic effects of the environmental toxins and toxicants. Green tea, the non-oxidized and non-fermented form of tea that contains several polyphenols, including green tea catechins, exhibits protective effects against these environmental toxins and toxicants in preclinical studies and to a much-limited extent, in clinical trials. The protective effects are collectively mediated by antioxidant, anti-inflammatory, anti-mutagenic, hepato- and neuroprotective, and anti-carcinogenic activities. In addition, green tea modulates signaling pathway including NFκB and ERK pathways, preserves mitochondrial membrane potential, inhibits caspase-3 activity, down-regulates pro-apoptotic proteins, and induces the phase II detoxifying pathway. The bioavailability and metabolism of green tea and its protective effects against environmental insults induced by pesticides, smoke, mycotoxins, polychlorinated biphenyls, and arsenic are reviewed in this paper. Future studies with emphasis on clinical trials should identify biomarkers of green tea intake, examine the mechanisms of action of green tea polyphenols, and investigate potential interactions of green tea with other toxicant-modulating dietary factors.

Keywords: green tea, pesticides, cigarette smoke, PCB, mycotoxin, heavy metal

Introduction

The biological, physical, and chemical environmental toxins/toxicants are introduced into the body via different routes. Food additives, contaminants, water pollutants, and drugs can be orally ingested. Airborne toxicants, particles, and tobacco smoke (active or passive) are inhaled, whereas cosmetic chemicals are absorbed through dermal contact [1]. Adverse health effects of these environmental toxicants on human bodies are determined by dose, route of exposure, toxicokinetic and toxicodynamic balance, and individual susceptibility.

Acute toxic effects can be attributed to exposure to large quantities of a toxicant, whereas chronic adverse health effects can be caused by prolonged exposures to small quantities of a specific toxicant, which can ultimately result in bioaccumulation [27]. Exposure to toxicants can promote the formation of reactive radical (oxygen or nitrogen) species, which are inflammatory molecules inflicting oxidative stress upon cells. Depending on the molecular targets, toxicants may impact human health at the molecular (DNA, RNA, or protein), organelle (mitochondria, lysosome, or membranes), cellular (growth inhibition or cell death), tissue, organ, and overall systemic levels [27]. Pesticides, smoke, mycotoxins, endocrine-disrupting chemicals (e.g., polychlorinated biphenyl), and heavy metals (e.g. arsenic) have been listed as the most common toxins/toxicants to humans.

Pesticides enter the body through inhalation of aerosols, dusts, and vapor, ingestion of food additives, and direct contact. Pesticides can damage vital organs, with the liver being the most susceptible due to its role in transforming, metabolizing and eliminating chemicals from the body [8]. Studies have found that many pesticides are potential hepatotoxicants. For example, chlorfenviphos, demeton-S-methyl (DSM), methiocarb, permethrin, chlorpyriphos, triazophos, and pirimicarb cause structural and functional changes in mammalian and avian hepatocytes [8]. Moreover, some neurotoxic pesticides have been associated with diseases characterized by neural damage or failure, such as Parkinson’s disease (PD) [9].

Smoke has long been recognized as a potent environmental toxicant and human health threat because it contains numerous chemical carcinogens and poisonous gases such as carbon monoxide, hydrogen cyanide, nitrogen and sulfur oxides, halogens, and organic acids [10]. The potential pathophysiological consequence associated with exposure to these poisonous gases may include the formation of carboxyhemoglobin, cyanide poisoning (cyanide blood), organic acid/ethanol intoxication [10], and enzymatic and morphologic alterations [11, 12]. Studies have found that inhalation of environmental smoke, including cigarette smoke, increases the risk of lung cancer, respiratory diseases, liver lesions, and liver cancer [11, 13] as well as the severity of liver damage in hepatitis patients [14].

Aflatoxins, mainly produced by Aspergillusflavus and A. parasiticus, are a subcategory of mycotoxins that, much like alcohol, often possess hepatotoxic properties [3]. Aflatoxin B1 (AFB1) exposure has been shown to cause acute, sub-acute, and chronic liver failure [14]. Furthermore, AFB1 is recognized as a potent carcinogen and mutagen [15]; the extent of aflatoxin contamination across regions of the United States has been correlated with incidences of hepatocellular carcinoma [15].

Many environmental contaminants act as endocrine disrupting chemicals (EDCs), capable of mimicking or blocking the action of hormones by binding to or interfering with their receptors. A subset of EDCs is known to affect metabolic processes if exposure occurs during early development, leading to obesity, type 2 diabetes mellitus and the metabolic syndrome. These chemicals are called “obesogens”. One class of the common obesogens is polychlorinated biphenyls (PCBs). PCBs are a major class of highly persistent organic pollutants (PCPs), widely used as synthetic chemical mixtures in industrial settings until it was banned in the United States and other developed countries beginning in 1970s. However, due to its resistance to degradation and bioaccumulation nature, the environmental and health impacts of PCBs are still of concerns. Epidemiological evidence now implicates exposure to PCBs in an increased risk of developing diabetes, hypertension, and obesity, all of which are clinically relevant to the onset and progression of cardiovascular disease. It is also suggested that PCBs exert their cardiovascular toxicity via additional mechanisms, including induction of chronic oxidative stress, inflammation, and endocrine disruption [16].

Exposure to inorganic and organic arsenic compounds in the environment remains a major public health problem, affecting hundreds of millions of people worldwide. Arsenic compounds affect almost every organ in the body, with health effects ranging from skin lesions and cancer to diabetes and lung disease [17, 18]. However, substantial knowledge gaps remain, particularly regarding the mechanisms by which arsenic induces such diverse health effects. Reactive oxygen species (ROS) generation is known to play a fundamental role in the arsenic-associated toxicity and carcinogenesis [19, 20].

Due to the inevitable human exposure to the aforementioned common environmental toxicants, there is a need for effective approaches to reduce or even eliminate their harmful impacts. Complementary and alternative approaches, such as dietary antioxidants or functional foods, could provide a safer way of protection or prevention than currently available options.

Tea, the dried leaves of the Camellia sinensis species of theaceae family, is a popular beverage with an annual production of three billion kilograms worldwide [21]. Green tea is a non-oxidized and non-fermented product that is made by drying fresh leaves (roasting) at high temperatures to inactivate the oxidizing enzymes. Green tea contains several tea polyphenols – primarily green tea catechins (GTCs) – that accounts for 30–40% of the extractable solids of dried green tea leaves [21]. Tea catechins include (−) epigallocatechingallate (EGCG), (−) epicatechingallate (ECG), (−) epicatechin (EC), and (−) epigallocatechin (EGC) [21], among which EGCG is the most abundant and bioactive and the most studied. GTCs are known to increase the amount of anti-oxidative enzymes in the blood, and function as antioxidants to scavenge ROS such as superoxide, hydrogen peroxide (H2O2), and hydroxyl radicals [22, 23]. In the past decades, GTCs have demonstrated the ability to quench free radicals generated by oxidative environmental toxicants [24] and consequently, reduce toxicant-mediated cytological damage, mutation-mediated DNA damage, cancer, and apoptosis. This review will discuss the potential benefits of GTCs in the attenuation of the side effects and toxicity associated with common environmental toxicants including pesticides, smoke, mycotoxins, PCBs, and arsenic in in vitro and in vivo studies.

Bioavailability and metabolism of green tea catechins

The bioavailability of oral GTCs is generally less than 0.2% in humans and research animals [2528]. Blood concentrations of GTCs peak at approximately 0.5 μM two to four hours after oral consumption of two cups of green tea [27]. The absolute oral bioavailability of EGCG is about 0.1% following the intake of 10 mg of green tea extract per kg body weight in humans and research animals [26, 28].

GTCs are metabolized in vivo through various metabolic transformations including methylation, glucuronidation, sulfation, oxidative degradation, and ring-fission metabolism [2933]. The liver and intestine are generally considered to be the main organs to metabolize GTC. One third of GTCs in mesenteric plasma are in the form of glucuronide conjugates of catechin and 3′-O-methyl catechin (3′OMC), suggesting that glucuronidation and methylation occur in the intestinal tract [34]. The absorbed GTC and associated metabolites are first delivered to the liver where high levels of UDP-glucuronyltransferase [35, 36], sulfotransferase [37, 38], and catechol-O-methyltransferase (COMT) [39], among other enzymes, further metabolize GTC. After exiting the liver, GTCs and their metabolites are released into circulation system and distributed to different organs and tissues. Although GTCs have many metabolites in the human body, the biological activity of those metabolites remains unknown.

Green tea modulates pesticide-related damage or disease

Massive application of pesticides worldwide has conferred immense agricultural advances that in turn have led to improved nutrition and health. Most pesticides work via inhibition of pest growth and development or direct toxicity. Though researchers initially believed pesticides were harmless to living organisms, including humans, the advancement of technology has revealed many toxic effects, such as hematologic and immunological abnormalities, genotoxicity, embryo toxicity, neurological alterations, and hepatic dysfunction [40]. The hepatotoxicity of pesticides is related to metabolism through cytochrome P450 (CYP450) enzymes. The nephrotoxic effects include the formation of calculi, renal dysfunction, renal tubular acidosis, crystal uria, and hematuria. Additional adverse effects induced by pesticides, such ascyromazine, include high blood pressure, reduced body weight, and epithelial hyperplasia [40]. Most damage to membranes and tissues caused by pesticides is attributed to oxidative stress mediated by ROS such as hydroxyl radicals and H2O2 [40].

Table 1 lists the in vitro and in vivo studies showing the protective effects of green tea against different pesticides [9, 4151]. Green tea extracts and polyphenols diminish pesticide-induced inhibition of cellular proliferation [9, 41, 42, 47, 49] and apoptosis [9, 41, 50], modulates intracellular signal transduction pathways [9], and elicits protective effect in a variety of neural cells [9, 45, 46]. For example, pretreatment with 500 μM L-theanine, a green tea ingredient, in SH-SY5Y neuroblastoma cells for 1 h significantly attenuated rotenone-induced loss of cell viability [9]. In PC12 cells, a commonly used cell line that retains the features of dopaminergic neurons, EGCG at low concentrations (1, 5, and 10 μM) significantly decreased paraquat-induced cell death, whereas higher concentrations of EGCG (50, 100, 200 μM) did not show any protective effect against paraquat damage [41]. In transformed RGC-5 retinal ganglion cells, EGCG (10–100 μM) and EC (10–75 μM) attenuated the rotenone-induced loss of cell viability [42]. In mouse hepatocytes and normal human epidermal keratinocytes (NHEK), 0.05–50 μg/mL green tea antioxidant (GTA) prevented the killing of hepatocytes by paraquat (1–10 mM) [47]. Tai et al. reported that EGCG protects SH-SY5Y from dichlorodiphenyl-trichloroethane (DDT)-induced cell death in a time-dependent manner [49].

Table 1.

Green tea and pesticide-related injury

First author, yr [ref] Study design Preparation of extract
or GTP Used
Treatments Effects of green tea
Cellular studies
Cho, 2008 [9] SH-SY5Y cells (neuroblastoma) Not available Rotenone
Rotenone+ L-theanine
↑ Cell proliferation
↓ DNA fragmentation and apoptosis
↓ Heme oxygenase-1 up-regulation
↑ ERK1/2 phosphorylation
↑ GDNF and BDNF production
Hou, 2008 [41] PC12 cells
(pheochromocytoma cells)
Pure EGCG Paraquat
Paraquat+EGCG
↓ Apoptosis via maintaining mitochondrial membrane potential
↓ Caspase-3 activity
↓ expression of SMAC in cytosol
Kamalden, 2012 [42] RGC-5 cells
(retinal ganglion cells)
Pure EGCG and EC Rotenone
Rotenone+EGCG
Rotenone+EC
↓ Rotenone-induced toxicity via JNK and p38 pathways
↓ Lipid peroxidation
Ruch, 1989 [47] Mouse hepatocytes
Human keratinocytes (NHEK cells)
GTE extracted with methanol Paraquat
Paraquat+GTE
↓ Apoptosis
↓ Inhibition of intercellular communication
Tai, 2010 [49] SHSY-5Y cells Pure EGCG DDT
DDT+EGCG
↓ DDT-induced toxicity
Tanaka, 2000 [50] CNL cell
(chronic neutrophilic leukemia)
Purchased from Kurita industry Company, Tokyo, Japan Paraquat
Paraquat+EGCG
Paraquat+catechin
Paraquat+EGCG+catechin
↓ Cell cycle rate
↓ ROS
↓ Paraquat-induced genotoxicity
Moldzio, 2010 [45] Mesencephalic cultures

Organotypic striatal cultures
Pure EGCG Rotenone
EGCG
Rotenone+EGCG
↓ Nitric oxide production
↓ Rotenone-induced toxicity
Pan, 2003 [46] Embryonic mesencephalic cells Green tea polyphenols Dopamine

Dopamine+GTP
↓ Dopamine uptake

↓ MPP-induced dopamine neuron injury
Animal studies
Pan, 2003 [46] Male C57BL/6 mice Green tea polyphenols Control
GTP
↓ Dopamine uptake in striatal synaptosomes
↓ MPP-induced dopamine neuron injury
Kim, 2006 [43] Rats Green tea leaves extracted with ethanol, concentrated, diluted with water, and extracted with ethyl acetate Sham
Paraquat
Paraquat+GTE
↓ Paraquat-induced pulmonary fibrosis
MDA, ET-1, and prepro-ET-1 mRNA
expression
↓ Catalase activity
Sai, 2000 [48] Male B6C3F1 mice
5-week-old
2% w/v green tea leaves were brewed with boiled water for 30 mins Control
PCP
GT infusion
PCP+green tea infusion
PCP-induced hepatocarcinogenesis
↑ Cell proliferation
↑ Production of GJIC and connexin32
Umemura, 2003 [51] Male B6C3F1 mice
5-week-old
2% w/v green tea leaves were brewed with boiled water for 30 mins Drinking water
DEN
DEN+GT infusion
DEN+PCP
DEN+PCP+GTE
↓ DEN-induced hepatocarcinogenesis
Drinking water
PCP
GT
PCP+GTE
↓ PCP-induced 8-oxodG levels in liver
↓ PCP-induced ALT activity in serum
Korany, 2011 [44] Adult male Wistar rats Film coated tablets contains 30% polyphenol which produced by Arab Co. Control
Fenitrothion
Fenitrothion+GTE
↓ Fenitrothion-induced toxicity in rat parotid
gland
↑ Caspase-3 cleaved activity
Preservation of normal architecture
Condensation of the chromatin

ALT, alanine aminotransferase; BDNF, brain-derived neurotrophic factor; DDT, dichlorodiphenyl-trichloroethane; DEN, diethylnitrosamine; EC, epicatechin; EGCG, (−)-epigallocatechingallate; ERK, extracellular signal-regulated kinase; ET-1, endothelin-1; GDNF, glial cell line-derived eurotrophic factor; GJIC, gap junction intercellular communication; GTE, green tea extract; GTP, green tea polyphenols; JNK, c-Jun N-terminal kinases; MDA, malondialdehyde; MPP, 1-methyl-4-phenylpyridinium; 8-oxodG, 8-oxodeoxyguanosine; PCP, pentachlorophenol; ROS, reactive oxygen species; SMAC, second mitochondria-derived activator of caspases. ↑ increase; ↓ decrease.

Green tea bioactive compounds (i.e., L-theanine or EGCG) have also demonstrated the ability to inhibit pesticide-induced apoptosis [9, 41, 50]. For instance, L-theanine (500 μM) inhibited pesticide-induced apoptosis in SH-SY5Y cells as shown by the marked attenuation of caspase-3 activities [9, 41], chromatin condensation, and nuclear fragmentation [9, 41]. EGCG (1, 5, 10 μM) was found to markedly reduce pesticide-induced DNA fragmentation [41]. The potential mechanisms include the maintenance of the mitochondrial membrane potential, inhibition of caspase-3 activity, and the down-regulation of the expression of the pro-apoptotic protein, Smac, in the cytosol [41]. EGCG and catechins at ≥ 10.0 μM both reduced the effect of paraquat on the cell cycle rate [50].

Furthermore, both L-theanine and EGCG have been shown to protect against pesticide-mediated neuronal damage [9, 45, 46]. Glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) act as potent and specific neurotrophic factors for midbrain dopaminergic neurons [52]. Cho et al. reported that L-theanine attenuated rotenone-mediated down-regulation of GDNF and BDNF in dopaminergic neurons [9]. Moldzio et al. also reported that EGCG significantly blocked rotenone-mediated increase in propidium iodide uptake, a measure of cell death, and 4-amino-5-methylamino-2′-7′-difluoroescein diacetate (DAF-FM) fluorescence intensity in organotypic striatal cultures from mice [45]. EGCG provides some protection for striatal slices by counteracting the rotenone-induced nitric oxide production. Pan et al. first reported that green tea polyphenols (GTP) exert a partial inhibitory effect on the uptake of 3H-dopamine (3H-DA) and 3H-1-methyl-4-phenylpyridinium (MPP+) by DA transporters (DAT) and provide partial neuro-protection from MPP+-induced injury in DAergic neurons, probably via their ability to block the DAT-dependent uptake of neurotoxin MPP+ [46].

Other studies examined the anti-oxidative activity of GTA, L-theanine, and EGCG in protection against pesticides. GTA prevents H2O2-induced cytotoxicity in cultured mouse hepatocytes (B6C3F1) and NHEK cells in a concentration-dependent manner [47]. Kamalden et al. found that green tea attenuates lipid peroxidation and rotenone toxicity in RGC-5 cells [42]. Heme oxygenase-1, a cellular stress protein expressed in brain and other tissues, responds to oxidative challenge [53]. L-theanine suppressed the production of ROS and the subsequent expression of heme oxygenase-1 induced by PD-related neurotoxins in SH-SY5Y cells [9]. L-theanine treatment also blocked the rotenone-induced oxidative stress and down-regulation of extra-cellular signal-regulated kinase 1/2 (ERK 1/2) phosphorylation [9]. Tanaka et al. reported that EGCG and green tea decreased the frequency of sister-chromatid exchanges (SCE) induced by paraquat, a generator of ROS, suggesting that green tea and polyphenol-containing foods may protect against ROS-induced genotoxicity [50].

Apart from in vitro testing, animal studies further confirmed the beneficial effects of green tea extracts (GTE, a mixtures of green tea polyphenols) against pesticide-induced toxicities including oxidative stress [40, 43, 46] in the liver [40], lung [43], and the nervous system (Table 1). Administration of the pesticides cyromazine and chlorpyrifos to rats led to significant elevations of transaminases and lactate dehydrogenase in the serum and a decrease in liver function. There were rises in catalase, hepatic SOD, and lipid peroxidation, and a decrease in glutathione (GSH), indicating increased oxidative stress [40]. Histopathological evaluation revealed that green tea supplementation mitigated mild hepatocyte necrosis and liver degeneration. Additionally, green tea normalized catalase, SOD, and liver function in the rats. Kim et al. showed that GTE significantly decreased pulmonary fibrosis induced by oxidative stress attributed toparaquat exposure in rats. GTE acted putatively by suppressing oxidative stress and endothelin-1 expression [43].

Lastly, green tea extract has been shown to suppress liver carcinogenesis induced by the pesticide pentachlorophenol (PCP) [48, 51]. In general, green tea extract contains, in descending order of abundance, EGCG, EGC, EC, ECG, and catechin. Umemura et al. found that a 2% green tea infusion prevented diethylnitrosamine (DEN)-induced and PCP-promoted hepatocellular tumors, and arrested the progression of cholangiocellular tumors, plausibly by counteracting carcinogen-mediated increases in serum alanine transaminase (ALT) activity and 8-oxodeoxyguanosine (8-oxodG) levels in the liver [51]. In a similar study, Sai et al. found that green tea acted as an anti-promoter against PCP-induced mouse hepatocarcinogenesis via its ability to prevent down-regulation of gap junction protein 1c (GJIC) [48]. Korany et al. [44] also showed that green tea pretreatment significantly reduced histopathological alterations induced by the pesticide fenitrothion. These studies, though limited in number, further elucidate the possible role of green tea in the inhibition of pesticide-induced carcinogenesis.

In summary, preclinical studies showed that the protective effects of green tea against pesticide toxicity are, in part, due to its antioxidant and free radical scavenging activity. Most prominently, green tea extracts and polyphenols have been shown to decrease the toxicity of rotenone [42, 45], DDT [49], and paraquat [41, 47]. Future investigations should further examine these findings in preclinical and hopefully clinical studies since the latter is a noticeable research gap in the current literature. The role of green tea as a prophylactic agent prior to pesticide exposure may also warrant examination.

Green tea modulates smoke related diseases or damage

Smoking has been linked to malignant transformation or abnormal cell proliferation [54], and tobacco smoke is regarded as one of the leading causes of lung cancer [41, 47, 55]. Carcinogenic materials present in smoke produce DNA adducts and a mutagenic and carcinogenic response [56]. Toxins in cigarette smoke might also initiate and exacerbate tissue injury [23]. Another smoking related ailment is Chronic Obstructive Pulmonary Disease (COPD). Oxidative stress caused by a high concentration of free radicals and other oxidants in cigarette smoke can lead to inflammation, direct damage to epithelial cells, inactivation of anti-protease, and lipid peroxidation [57].

The protective activity of green tea and its constituents, including GTE, GTP, EGCG, ECG, EGC, EC, and catechins, on smoke-induced damage has been shown in cell culture, animal, and human studies (Table 2) [22, 51, 5783]. Cellular studies have shown that EGCG and GTE protect against smoke-induced cellular proliferation [62, 71, 77], DNA damage [66, 80, 83], oxidative stress [22, 62, 71], and tumorigenesis [56, 71] through cell signaling pathways [71, 77]. More specifically, Syed et al. reported that EGCG pretreatment (20–80 μM) of normal human bronchial epithelial cells (NHBE) resulted in significant inhibition of cigarette smoke condensate (CSC)-induced cell proliferation [77]. Pre-, co-, and post-incubation of primary human osteoblasts with sub-toxic concentrations of GTE (0, 50, 100, and 200 μg/ml) or catechins (0, 50, 100, and 200 μM) dose-dependently reduced cigarette smoke medium (CSM)-mediated cellular damage and concomitantly increased the viability of human osteoblasts [62].

Table 2.

Green tea and smoke-related injury

First author, yr [ref] Study design Preparation of Extract
or GTP Used
Treatments Results
Cellular studies
Misra, 2003 [22] Guinea pig tissue microsomal
suspension
1g green tea added to 10 mL of boiling water and brewed for 5 min. Control
Smoke
Smoke+tea infusion
↓ Smoke-induced oxidative damage of protein
Holzer, 2012 [62] Primary human osteoblasts GTE (Sunphenon, Japan) Smoke
Smoke+GTE
Smoke+catechins
↑ Viability
↓ ROS formation
Rathore, 2012 [71] MCF10A cells MCF7 cells
(human breast epithelial cells)
Pure EC, ECG, EGC, ECGC (Sigma-Aldrich Ltd., St. Louis, MO) NNK+B[a]P
NNK+B[a]P+EC
NNK+B[a]P+ECG
NNK+B[a]P+EGC
NNK+B[a]P+ECGC
↓ NNK+B[a]P-induced carcinogenesis
↓ NNK+B[a]P-induced ROS production
↓ Phosphorylation of ERK1/2
↓ DNA damage
Syed, 2007 [77] Normal human bronchial epithelial cells EGCG (>98% pure) (Mitsui Norin Co., Ltd Shizuoka, Japan) Smoke
Smoke+EGCG
↓ Smoke-induced cell proliferation
↓ Smoke-induced activation of NF-κB,
↓ NF-κB-regulated proteins cyclin D1, MMP-9, IL-8, iNOS
↓ Smoke-induced phosphorylation of ERK1/2, JNK, p38 MAPK
Leanderson, 1997 [66] A549 cells
(human lung adinocarcinoma cells)
Green tea extracted in 75°C water, 80% ethanol, chloroform, and ethyl acetate Control
Smoke+H2O2+Iron
Smoke+H2O2+Iron+EGCG
↓ Smoke- and H2O2-induced toxicity and DNA strand breaks
↓ Production of lipid peroxidation
Weitberg, 1999 [80] A549 cells Pure EGCG (Sigma-Aldrich Ltd., St. Louis, MO) Control
NNK
NNK+EGCG
↓ NKK-induced tumor promotion
↓ NKK-induced single-strand DNA breaks
Zhou, 2000 [83] Human peripheral blood lymphocytes 20 g of dry green tea leaves extracted in 400 mL of distilled water at 80°C, 90°C, and 100°C for 10, 30, and 60 min, respectively ERP/ETS
ERP/ETS+GTE
↓ Smoke-induced mutations
Takahashi, 2004 [78] MDCK cells
(Mardin-Darby canine kidney cells)
Pure EGCG (Sigma-Aldrich Ltd., St. Louis, MO) Control
DMN
EGCG
DMN+EGCG
↑ Production of GJIC and connexin43
↑ Phosphorylation of connexin43
Khoi, 2013 [64] Human endothelial ECV304 cells Pure EGCG (Sigma-Aldrich Ltd., St. Louis, MO) Nicotine
Nicotine+EGCG
↓ Nicotine-induced ROS production
↓ Nicotine-induced cell invasion and MMP-9 activity
↑ Nicotine-induced NF-κB and AP-1 activation
Animal studies
Umemur, 2003 [51] Male B6C3F1 mice 5-week-old 2% w/v green tea leaves brewed with boiled water for 30 mins Drinking water
DEN
DEN+Green tea infusion
DEN+PCP
DEN+PCP+Green tea
↓ DEN-induced hepatocellular tumors
Arrest the progression of cholangiocellular tumors
Chan, 2009 [57] Sprague-Dawley rats 60 g dried Lung Chen tea leaves brewed in 600 mL hot water (not boiling) for 30 minutes Sham
Smoke
Sham+tea
Smoke+tea
↓ Serum 8-isoprostane level
↓ Lung SOD activity
↓ Catalase activity
Fiala, 2005 [60] Male Hartley guinea pigs
3-week-old
EGCG (~94% pure) Sham
Smoke
Smoke+p-XSC
Smoke+EGCG
↔ Smoke-induced lung cancer
Witschi, 1998 [81] Male and female strain A/J mice Boiling deionized water poured over 62.5 g green tea leaves and left to stand for 15 min. Sham
Smoke
Smoke+GTE
↔ Smoke-induced lung tumor multiplicity
Lu, 2006 [68] Female A/J mice
4–6 weeks old
N=11–24
Polyphenon E containing 65% EGCG, 7% ECG, 3% EGC, 9% EC, 3% GCG Control
NNK
NNK+Polyphenon E
↓ Cell proliferation of adenomas
↑ Cell apoptosis of adenomas
↓ c-Jun and ERK1/2 phosphorylation
Lubet, 2007[69] Female Fisher-344 rats and Sprague-Dawley rats
28-day-old
Polyphenon E containing 64.3% EGCG, 3.1% EGC, 9.1% EC, 8.1% ECG, other polyphenols OH-BBN
OH-BBN+Polyphenon E

MNU
MNU+Polyphenon E
↓ OH-BBN-induced urinary bladder tumors

↓ MMN-induced mammary cancer
Sato, 2003 [75] Male Wistar rats
7-week-old
Powdered green tea leaves BBN
BBN+Green tea
↓ BBN-induced urinary bladder tumors
Shimizu, 2011 [76] Male db/db mice Pure EGCG (Mitsui Norin Co. Ltd., Tokyo, Japan) Control
EGCG
DEN
DEN+EGCG
↓ Tumor incidence and multiplicity
↓ Serum levels of insulin, IGF-1, IGF-2
↓ p-IGF-1R protein
↓ Phospholylation of ERK and Akt
↓ Serum levels of free fatty acids
Maliakal, 2011 [70] Female Wistar rats Boiled tap water added to tea powder (2% w/v) with intermittent stirring
for 10 min and filtered
NMBA
NMBD+GTE
↓ Tumour multiplication, size, volume
↔ Tumour incidence
Chan, 2012 [59] Sprague-Dawley rats 60 g dried Lung Chen tea leaves brewed in 600 mL hot water (not boiling) for 30 minutes Sham
Smoke
Sham+tea
Smoke+tea
↓ Lung lipid peroxidation marker
↓ Smoke-induced serum MDA
↓ Neutrophil elastase concentration
↓ Smoke-induced MMP-12
Kaneko, 2003 [63] Female Syrian golden hamsters
6-week-old
Not available Control
BOP
BOP+Catechin
↓ BOP-induced formation of 8-oxodG in the pancreas
Abe, 2008[58] Male
BrlHan:WIST@Jcl (GALAS) rats
5-week-old
Green tea extract product Sunphenon BG (Taiyo Kagaku Company, Limited, Mie, Japan) DMBDD
DMBDD+GTC
↓ DMBDD-induced liver and stomach cancers
Kim, 2009[65] Male albino rats Green tea leaves
extracted with 80% ethanol
Control
DMN
Hepatic fibrosis control
Hepatic fibrosis+GTE
↓ DMN-induced hydroxyproline
↓ DMN-induced MDA
Yang, 2009[82] Female Wistar rats Decaffeinated catechins containing EGCG, ECG, GCG, EC, EGC, GC,
catechin
Non-cooking-oil-fumes
Non-cooking-oil-fumes+catechins
Cooking-oil-fumes (COF)
Cooking-oil-fumes+catechins
COF-enhanced ROS level
COF-enhanced lung dityrosine
COF-enhanced 4-HNE Level
↑ COF-decreased Bcl-2 and HSP70 Expression
Roy, 2010[72] Male swiss albino mice GTP (Indfrag limited, Bangalore, India) Control
GTP
BTP
DEN
DEN+GTP
DEN-induced COX-2 expression
DEN-induced activation of NFκB and Akt
Sagara, 2010[73] Female C3H/He mice GTP (Tokyo Kasei Industries, Tokyo, Japan) Control
BBN
GTP
BBN+GTP
↓ Frequency of invasive tumors
↓ Tumor volume and microvessel density of intratumoral and stromal region
Saiwichai, 2010[74] Rats Dried methanol extract dissolved in 500 mL of water at 50°C and washed with hexane, chloroform and ethyl acetate; residue dissolved in 50°C water and freeze dried to obtain mixture of EGCG, ECG, EGC, EC Sham
Smoke
Smoke+GTE
↓ Serum HMGB1 level
Human studies
Lee, 1997 [67] Questionnaire based study, Male Not available Non-smokers
Smokers
Smokers+Green tea
↑ SCE rates in smokers group
↔ Frequency of SCE in smokers
Vermeer, 1999 [79] Healthy female volunteers Lyophilized green tea solids dissolved in boiling water (0.5 g in 100 mL); four cups (2 g) of tea per day in test week 4, and eight cups (4 g) of tea per day in test week 5. Fish meal rich in amines
Fish meal+Green tea
↓ Nitrosation
Hakim, 2008 [61] RCT
Smokers (N=133)
4 cups (960 mL)/d of decaffeinated green tea product consisting of EGCG, EGC, EC, ECG, total catechins, gallic acid, total flavanol glycosides. Water
Green tea
Urinary 8-OHdG

4-HNE, 4-hydroxy-2-nonenal; 8-OHdG, 8-hydroxydeoxyguanosine; BBN, N-butyl-N-(4-hydroxybutyl)-nitrosamine; BOP, N-Nitrosobis(2-oxopropyl)amine; COX-2, cyclooxygenase-2; DEN, diethylnitrosamine; DMBDD, N-nitrosodiethylamine (DEN), N-methylnitrosourea (MNU), N-butyl-N-(4-hydroxybutyl)nitrosamine (BBN), 1,2-dimethylhydrazine (DMH) and 2,2′-dihydroxy-di-n-propylnitrosamine (DHPN); DMN, dimethylnitrosamine; EC, epicatechin; ECG, epicatechin-3-gallate; EGC, epigallocatechin; EGCG, (−)-epigallocatechin gallate; ERK, extracellul arsignal-regulated kinase; ERP/ETS, extractable respirable particulate/environmental tobacco smoke; GJIC, gap junction intercellul arcommunication; GTE, green teae xtract; GTP, green tea polyphenols; HMGBl,high-mobilitygroupproteinB1 ; IGF, insulin-like growth factor; IL-8, interleukin-8; iNOS, inducible nitric oxide synthase; JNK, c-Jun N-terminal kinases; MAPK, mitogen-activated protein kinases; MDA, malondialdehyde; MMP, matrixmetalloproteinases; MNU, methylnitrosourea; NF-κB, nuclearfactor-κB; NNK,4-(N-methyl-N-n-trosamino)-l-(3-pyridyl)-l-butanone; PCP, pentachlorophenolin; RCT, randomized controlled trial; ROS, reactive oxygen species; SCE, sister-chromatidex change; SOD, superoxide dismutase. ↑ increase; ↓ decrease; ↔ no difference

Aside from inhibiting smoke-induced cellular proliferation in normal cells, green tea polyphenols, such as GTP, EGCG, ECG, EGC, and EC, also reduced smoke-induced DNA damage [66, 71, 80]. Pretreatment with 20–100μg/mL GTP for 2 h resulted in a dose-dependent decrease in smoke-mediated DNA damage in A549 cells, and GTP concentration was inversely correlated with the extent of DNA damage observed [66]. In addition, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced single-strand DNA breaks were prevented by EGCG in A549 cells [80]. Rathore et al. reported that co-treatment with 10 μg/mL EC, ECG, EGC, and EGCG for 24 h reduced the NNK- and benzo[a]pyrene (B[a]P)-induced DNA damage in both human MCF10A breast epithelial cells and human MCF7breast cancer cells [71]. Zhou’s study demonstrated the strong anti-clastogenic effect of 0.025–0.075 mg/mL tea extract against extractable-respirable particulate in environmental tobacco smoke (ERP–ETS) in human peripheral blood lymphocytes [83]. The study also found that temperature and extraction soaking time impacted the potency of the tea extract. For example, increased soaking time (from 10, 30 to 60 min) at 100°C reduced the inhibitory activity of tea extract, whereas the same extended extraction period at lower temperatures increased the tea extract activity. Tea extract prepared by soaking at 80°C for 60 min produced the highest anti-clastogenic effect [83]. Further studies could help to confirm the precise mechanisms responsible for the inhibition of DNA damage.

Studies have found that GTE has a protective effect against smoke-induced oxidation [22, 62, 71]. Protein oxidation induced by cigarette smoke in guinea pig lung microsomes was inhibited by 93% upon exposure to the green tea infusion [22]. Holzer et al. reported that osteoblasts pretreated or co-incubated with 0–200 μg/mL GTE or 0–200 μM catechins for 4 h prior to a 15-min exposure to smoke reduced ROS formation dose-dependently [62]. The GTE used for the Hozer’s study (Sunphenon® 90LB, Taiyo Kagaku, Japan) was obtained from the leaf of traceable green tea (Camelliasinensis) and consisted of >80% polyphenols, of which >80% are catechins, >40% EGCG, and <1% caffeine.

Probable signal transduction pathways involved in EGCG’s protective effect against smoke induced injuries include nuclear factor-κB (NF-κB) [77], phosphoinositide 3-kinase (PI3K) signaling [77], extracellular signal-regulated kinase (ERK)1/2 pathway [71], and H2AX pathway [71]. EGCG treatment of NHBE cells suppressed smoke-induced phosphorylation of NF-κB/p65, p38, ERK1/2, and JNK1/2 MAPK in a dose-dependent manner [77]. EGCG not only inhibited DNA-binding of NF-κB, but also resulted in a significant decrease in smoke-induced NF-κB promoter activity [77]. The mechanism of NF-κB inactivation by EGCG involved IKKα phosphorylation and degradation of IkBα [77]. Rathore et al. reported that co-treatment with 10 μg/mL EC, ECG, EGC, and EGCG for 24 h reduced the NNK- and B[a]P-induced ERK1/2 and H2AX phosphorylation in both MCF10A cells and MCF7 cells [71].

In vitro studies have also shown that green tea possesses, in addition to altering signal transduction pathways, activity to block smoking-associated tumorigenesis [56, 71]. For instance, EC, EGC, EGCG and with the highest potency, ECG, have been reported to block ROS elevation and ERK pathway activation, and effectively suppress NNK- and B[a]P-induced cellular carcinogenesis [71]. Despite these promising findings, contradictory data exist on whether GTE inhibits mutagenicity [54]. As a potential mechanism for the chemopreventive activity of EGCG in renal epithelial cells, pretreatment with EGCG for 12 h preserved the level of gap junction intercellular communication (GJIC) protein, also known as connexin 43, in Madin-Darby canine kidney epithelial cells (MDCK) exposed to dimethylnitrosamine (DMN) [78].

Animal studies confirmed the anti-oxidative ability of EC, EGC, ECG, and EGCG in a variety of lung injury models. Chan et al. [57, 59] treated Sprague-Dawley rats with 2% Lung Chen Tea, a Chinese green tea, in the presence and absence of smoke for 56 days. Lung Chen Tea (2%) with the largest amount of EGCG was found to alter oxidative stress in the serum and lungs in the smoke-exposed group and protect against smoke-induced lung damage as determined by histological and morphometric analyses [57]. Concomitantly, 2% Lung Chen Tea may also help to prevent the smoke-induced up-regulation of matrix metalloproteinase-12 (MMP12) activity, leading to a reduction in ROS [59]. Yang et al. reported that catechins reduced oxidative stress by reducing the Bax/Bcl-2 ratio and preserving HSP70 chaperone protein expression in the lung of study animals [57].

Most animal studies – with a few exceptions [60, 81] – further confirmed the beneficial effects of EGCG or GTE on smoke related disease or damage. Administration of GTE, GTP, or EGCG during initiation, promotion, or progression inhibited NNK-induced lung tumorigenesis in rats, mice, and hamsters [84]. Lu’s study showed that the administration of tea polyphenols inhibited the progression of existing lung adenoma to adenocarcinoma [68]. In addition, GTP been shown to have protective effects against bladder tumors [69, 73, 75], hepatocellular tumors [51, 76], and esophageal tumors [70]. These studies provided additional evidence that GTP can possibly be used in conjunction with other therapeutics.

Several clinical studies examined the effects of green tea on smoke-related DNA damage and gene mutations (Table 2) [61, 67, 79]. Lee et al. found that drinking green tea inhibited DNA mutations in blood samples from smokers and non-smokers [67]. Another study with 30 male and 90 female smokers found that consuming 4 cups or 960 mL of tea was associated with a significant decrease in urinary 8-hydroxydeoxyguanosine (8-OHdG), an indicator of DNA damage, as determined by ELISA [61]. In addition, green tea has also been shown to reduce the formation of the endogenous carcinogen, N-nitrosodimethylamine (NDMA) [79]. Despite the promising results of these clinical studies, additional data are necessary to not only understand the specific mechanisms involved, but also to fully explore the therapeutic potential of GTC.

In summary, polyphenol-containing green tea may counteract environmental toxins by preventing extensive damage to DNA, proteins, and cells. GTP may reduce smoke-induced cancer risk. However, it is unclear if the action of GTP is specific for smoke-induced carcinogenesis. Besides cigarette smoke, other sources of inhaled pollution, for example, air pollutant, house dust, airborne fungi, allergic irritants and toxins, are also warranted more attention in future studies on how green tea may mitigate the potential risk of health. In addition, future studies may need to address the differential responses to green tea by individuals, and to compare the effects of various types of teas due to their diverse ingredients.

Green tea and mycotoxin-related disease or damage

Aflatoxin and fumonisins, low-molecular-weight secondary metabolites produced by filamentous fungi [85], are the most relevant mycotoxins associated with human diseases [86]. Aflatoxins, comprised of more than 20 fungal metabolites, were first isolated in dead turkey with turkey X disease, which was later reported to be associated with mold-contaminated peanuts [87]. The major naturally produced aflatoxins include B1, B2, G1 and G2 [88]. The letters B and G denote the blue and green fluorescence emitted upon exposure to UV light. The most toxic aflatoxin B1 (AFB1) is also a potent liver carcinogen widely used in carcinogenesis studies. Acute exposure to aflatoxins leads to acute aflatoxicosis characterized by acute hepatic injury, tissue edema, hemorrhage, and eventual death, whereas chronic exposure to aflatoxins gives rise to the development of malignancies [89]. A high level of aflatoxin exposure can promote hepatic cell necrosis. Fumonisins, first described in 1988, are produced by Fusarium verticillioides and F. proliferatum. Fumonisin B1 is the most abundant member in this family and has been associated with esophageal cancer [90], most prominently in areas with high incidence of esophageal cancer such as Transkei (South Africa), China, and northeast Italy [91].

The protective effects of green tea polyphenols, i.e. GTE and EGCG, against mycotoxin toxicity such as mutagenicity and DNA damage are summarized in Table 3 [92107]. Salmonella typhimurium strains such as TA100, TA98, and TA97 have been used to test the effect of green tea on AFB1-induced mutagenicity [92, 103, 107]. Wang reported that GTE, GTP, and EGCG inhibited the mutagenicity of each promutagen in a dose-dependent manner. At the highest dose of GTP (500 μg/plate), the mutagenicity was inhibited by more than 95% [107]. Hour et al. determined that oolong tea, black tea, and green tea had anti-mutagenic properties and prevented cancer development, while GTE (0.1–100 μg/plate) showed the greatest inhibitory effect in strain TA100. In Snijman’s study, 0.8μM EGCG was also shown to prevent AFB-induced mutagenicity [103]. Additionally, the anti-mutagenic properties of flavonoids were related to their lipophilicity or hydrophilicity, which ultimately influences methylation, hydroxylation and glycosylation. Wang et al. reported that green tea reduced DNA damage caused by AFB1 [107] and in a dose-dependent manner, the frequency of SCE, chromosomal aberrations, and 6-thioguanine (6-TG)-resistance in Chinese hamster lung fibroblasts (V79); 100 μg/mL GTP induced 75% inhibition of SCE [107]. In addition, Mo’s group reported that the tea extracts inhibit aflatoxin synthesis through the down-regulation of the transcription of aflR and aflS, two genes involved in the regulation of aflatoxin biosynthesis [100]. Green tea also inhibits gene forward mutation and reduces the frequencies of SCE and chromosomal aberrations. Animal studies confirmed the effect of green tea on AFB1-induced carcinogenesis [98, 102, 108, 109]. Marnewick et al.[98] found that green tea enhanced hepatic microsomal fractions and protected from AFB1-induced mutogenesis in Male Fischer rats.

Table 3.

Green tea and mycotoxin-related injury

First author, yr [ref] Study design Preparation of Extract
or GTP Used
Treatments Effects of green tea
Cellular studies
Hour, 1999[92] TA100, TA98, and TA97(Salmonella typhimurium strains) 12.5 g green tea leaves added to 500 mL boiling water and steeped for 15 min AFB1
AFB1+GTE AFB1+EGCG
↓ AFB1-induced mutagenesis
Mo, 2013 [100] Aspergillusflavus 10 g green tea soaked in 100 mL of boiling demi-water for 0.5 h AFB1
AFB1+Green tea
↓ Aflatoxin production by down-regulating the transcription of afIR and afIS
Snijman, 2007 [103] TA100 and TA98 Pure EGCG (Sigma-Aldrich, St. Louis, MO) AFB1
AFB1+EGCG
↓ AFB1-induced mutagenesis
Wang, 1989 [107] TA100 and TA98 100 g of green tea powder suspended in water (0.75 L, 75°C), filtered, and extracted with 80% ethanol (0.75 L, 50°C); aqueous and ethanol extracts combined, concentrated and extracted with chloroform; remaining aqueous phase extracted with ethyl acetate; residue dissolved in water and
freeze-dried to obtain GTP containing EC, EGC, ECG and EGCG.he oxidation of by atmospheric oxygen
AFB1AFB1+GTP ↓ AFBl-induced mutagenesis
↓ AFBl-induced frequency of SCE
↓ AFBl-induced chromosomal aberrations
Animal studies
Ito, 1989 [94] Male Wistar rats 20 g green tea added to 1 L of boiling water and steeped for 10 min AFB1
AFB1+GTE
↓ AFB1-induced chromosome aberration
Marnewick, 2004 [98] Male Fischer rats Boiled tap water added to tea leaves and stems (2 g/100 mL or 4 g/100 mL) AFB1
AFB1+Green tea
↓ Activation of AFB1-inducedmutagenesis
Marnewick, 2009 [99] Male Fischer rats Boiled tap water added to tea leaves and stems (2 g/100 mL or 4 g/100 mL) DMSO
DEN
DEN+FB1
DEN+FB1+Green tea
Oxygen radical absorbance capacity
Serum cholesterol
GSSG level and catalase activity
GSH:GSSG ratio
FB1-induced lipid peroxidation
Qin, 1997 [101] Male Fischer rats Instant GT powder (Thomas J. Lipton, Englewood Cliffs, NJ) AFB1
AFB1+Green tea
↓ AFB1-induced hepatocarcinogenesis by modulation of AFB1 metabolism
↓ AFB1-DNA binding
↓ AFB1-induced GST-P-positive hepatocytes
Qin, 2000 [102] Male Fischer rats Instant GT powder (Thomas J. Lipton, Englewood Cliffs, NJ) DMSO
DMSO+Green tea
DMSO+CCl4
AFB1
AFB1+CCl4 AFB1+CCl4 +Green tea
↓ Initiation and promotion of AFB1-induced hepatocarcinogenesis
↓ AFB1-induced GST-P-positive hepatic foci and area and volume
↑ GSH level
Tulayakul, 2007 [105] Female piglets Green tea extracts (Taiyo Kagaku Co., Ltd., Mie, Japan) AFB1
AFB1+GTE
↑ AFB1 detoxification
↓ P450 enzyme activity in liver
↑ GST activity in intestine
↑ Conversion of AFB1 to aflatoxicol in the liver
Human studies
Huang, 2004 [93] Randomized, double blinded and placebo-controlled phase IIa chemoprevention
Trial (design)
GTP consisted of EGCG, ECG, EGC, and EC (Shili Natural Product Company, Guilin, Guangxi) Placebo
Green tea (500 mg)
Green tea (1000 mg)
For 3 months
Green intake at 1000 mg daily for 3 months was safe
Luo, 2006 [95] Randomized, double blinded and placebo-controlled phase IIa chemoprevention GTP consisted of EGCG, ECG, EGC, and EC (Shili Natural Product Company, Guilin, Guangxi) Placebo
Green tea (500 mg)
Green tea (1000 mg)
For 3 month
↓ 8-OHdG levels
Luo, 2006 [96] Randomized, double blinded and placebo-controlled phase IIa chemoprevention
trial
GTP consisted of EGCG, ECG, EGC, and EC (Shili Natural Product Company, Guilin, Guangxi) Placebo
Green tea (500 mg)
Green tea group (1000 mg)
For 3 month
Metabolic profiling identified 106 metabolites, and 56 of them were chosen to construct discriminant functions.
Tang, 2008[104] Randomized, double blinded and placebo-controlled phase IIa chemoprevention
trial
GTP consisted of EGCG, ECG, EGC, and EC (Shili Natural Product Company, Guilin, Guangxi) Placebo
Green tea (500 mg)
Green tea(1000 mg)
For 3 month
AFB-AA levels
↓ AFM1
↑ AFB-NAC levels
↑ AFB-NAC:AFM1

8-OHdG, 8-hydroxydeoxyguanosine, AFB–AA, AFBl–albumin adducts; AFB–NAC, aflatoxin Bl–mercapturic acid; AFB1 aflatoxin B1; AFM1, aflatoxin M1; DEN, diethylnitrosamine; EGCG, (−)-epigallocatechin-3-gallate; FB, fumonisin B; GSH, glutathione; GSSG, oxidized glutathione; GST, glutathione S-transferase; GTE, green tea extract; GTP, green tea polyphenols; SCE, sister-chromatid exchange. ↑ increase; ↓ decrease; ↔ no difference

Consumption of green tea during initiation or promotion phases of AFB1-induced carcinogenesis inhibited the glutathione S-transferase positive hepatic foci in male Fischer rats [102]. Li’s team [109] demonstrated that green tea was effective in inhibiting the hepatocarcinogenic effects of pre-inoculated diethylnitrosamine in rats. Boer et al. found that green tea markedly reduced the mutagenic potency of AFB1 and other carcinogens in a transgenic rodent model [108], while Tulayakul et al. showed that the GTE increased AFB1 detoxification via conjugation of AFB1 to GSH in the intestinal tissues, but not the liver.

Dietary AFB1 has been identified as one of the major etiologic factors for primary liver cancer (mainly hepatocellular carcinoma) in the developing world, such as Southwest Asia and sub-Saharan Africa [106]. GTP has been shown as safe and effective chemopreventive agents in various AFB1-induced liver tumors. In a randomized, double-blinded, and placebo-controlled phase II chemoprevention trial in China, Wang’s team evaluated the efficacy of GTP intervention on urinary 8-OHdG in 124 high-risk individuals with aflatoxin exposure [106]. All participants tested positive for aflatoxin-albumin adducts and took either a placebo or a capsule containing 500 mg or 1,000 mg GTP daily for 3 months. Prior to the intervention and at the first and third months of the study, 24-hour urine samples were collected. The purity of GTP capsules was > 98.5% and the GTP consisted of a mixture of EGCG, EGC, ECG, EC, and catechin. Wang’s study demonstrated that (i) in GTP-treated groups, urinary EGC and EC levels displayed significant and dose-dependent increases, and (ii) at the end of 3-month intervention, urinary 8-OHdG concentrations decreased significantly in both GTP-treated groups [106], suggesting GTP is effective in diminishing oxidative DNA damage. A meta-analysis by Guyton et al. supported the preventive activity of green tea in liver cancer mediated by hepatotoxic contaminants including aflatoxins [110], but further studies are necessary to better understand the clinical applications of GTP upon mycotoxin exposure.

AFB1 metabolism and specific AFB1 biomarkers were studied to evaluate the efficacy of chemopreventive agents such as GTP, in order to determine if they could provide mechanistic information for human intervention trials. From the same clinical study by Wang’s research team, Tang et al. further elucidated that daily administration of GTP (500 mg) significantly (15%) reduced levels of AFB1-albumin adducts (AFB–AA) after 3 months, compared with levels of the placebo and baseline groups. Similar results were also observed in the 1000 mg GTP group at 1 and 3 months after the intervention. In addition to AFB–AA, urinary aflatoxin M1 (AFM1) is another biomarker for AFB1 exposure that correlates well with dietary intake of AFB1 and the risk of human hepatocellular carcinoma (HCC). A reduction in urinary levels of AFM1 at 3 months of intervention was also observed in Tang’s study [104]. Intervention with 500 and 1000 mg GTP significantly elevated urinary levels of AFB1-mercapturic acid (AFB–NAC), the major detoxifying metabolic product of AFB1-8,9-epoxide. The increase in the AFB1–NAC:AFM1 ratio in GTP-treated groups further demonstrated the GTP-mediated induction of the phase II detoxifying pathway in AFB1 metabolism. Future studies should identify the optimal doses of green tea for the prevention of aflatoxin damage, taking into account variation in individual aflatoxin levels as a confounding factor that is largely attributed to differences in food products and most important in regions with high aflatoxin exposure.

GTP has been considered as one of the major resources of chemopreventive agents against many types of cancers. Huang et al. reported that GTP have been shown to be relatively safe at target organs such as the liver [93]. In the same clinical trial, Wang and Luo reported that major GTP, especially EGCG, were detectable in plasma after 1- and 3-month GTP interventions; plasma ECG and EGCG and urinary EC and EGC may be used as reliable biomarkers to reflect the consumption of green tea or GTP supplementation at the population level [96, 106].

Green tea modulates toxicities associated with PCBs

Some coplanar PCBs, e.g., PCB 126, exert their vascular toxicity primarily via stimulation of the aryl hydrocarbon receptor (AhR) and subsequent uncoupling of cytochrome P450 1A1 (CYP1A1), leading to oxidative stress and inflammation [111].

ROS induce the activity of antioxidant enzymes, such as superoxide dismutase (SOD), catalase, glutathione reductase (GSR), glutathione transferases (GST), thioredoxins (Trx) and thioredoxin reductases (TrxR) [111]. The antioxidant enzymes work together either to quench ROS or to reactivate enzymes through a crosstalk of multiple regulatory pathways including the aryl hydrocarbon receptor (AhR) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcription factors [111]. Petriello et al. recently have demonstrated both AhR and Nrf2 signaling pathways are activated by PCBs [111].

Table 4 summaries the protective effects of green tea against the toxicity of PCBs [112116]. Anti-inflammatory food components, such as green tea EGCG, can work through both AhR- and Nrf2-mediated mechanisms to prevent PCB-induced inflammation. For example, EGCG can protect against the activation of vascular endothelial cells by coplanar PCBs [112, 115]. EGCG can suppress the expression of AhR-regulated CYP1A1 and induce Nrf2-regulated antioxidant enzymes, such as GST and NQO1, thus providing protection against PCB-induced inflammatory responses in cultured endothelial cells [112]. In addition, GTE has been shown to decrease oxidative stress in livers of mice exposed to PCB 126 via the induction of SOD1, GSR, NQO1 and GST [114].

Table 4.

Green tea and PCB-related injury

First author, yr [ref] Study design Preparation of Extract
or GTP Used
Treatments Effects of green tea
Cellular studies
Han, 2012[112] Primary vascular endothelial cells Pure EGCG (Cayman Chemical Co., Ann Arbor, MI) PCB
PCB+EGCG
↓ CYP1A1 mRNA and protein expression
↓ Superoxide production
↓ MCP-1 and VCAM-1
↑ Nrf2-controlled genes (GST and NQO1)
Ramadass, 2003[115] Endothelial cells Pure EGCG (Sigma-Aldrich, Ltd., St. Louis, MO) PCB 77 (3,3′, 4,4′-tetrachlorobiphenyl)
PCB 77+EGCG
↓ Oxidative stress↓ CYP1A1 activity
Weisburger, 1994[116] in vitro systems of Jägerstad Not available PCB
PCB+EGCG
↓ Formation of typical HCAs (MeIQx and PhIP)
Animal studies
Morita, 1997[113] Male rats Not available PCB
PCB+GTE
↓ Liver distribution of PCB
Newsome, 2014 [114] C57BL/6 mice GTE (Taiyo International Inc., Minneapolis, MN) PCB
PCB+GTE
↑ SOD1, GSR, NQO1 and GST
↓ Oxidative stress

EGCG, (−)-epigallocatechingallate; GTE, green tea extract; GSR, glutathione S-reductase; GST, glutathione S transferase; HCAs, Heterocyclic amines; MCP-1, monocyte chemotactic protein-1; MeIQx, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline; NQO1, NAD(P)H:quinoneoxidoreductase; Nrf2, nuclear factor erythroid 2 [NF-E2]-related factor 2; PCB, polychlorinated biphenyl; PhIP, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine ; SOD1, Superoxide Dismutase 1; VCAM-1, vascular cell adhesion protein-1. ↑ increase; ↓ decrease.

In summary, epidemiological studies have linked exposure to POPs, such as PCBs, with an increased risk of developing diabetes, hypertension, and obesity, all of which lead to onset and progression of CVD [16]. Emerging evidence now suggests that green tea with its anti-inflammatory and anti-oxidative properties can decrease vascular toxicity and provide protection against PCBs-induced vascular toxicities.

Green tea modulates arsenic-associated toxicities and cancer

The beneficial effects of green tea on arsenic-associated toxicities were summarized in Table 5 [117130]. Green tea and its principal polyphenol, (−)-EGCG, efficiently counteracted the cytotoxic effects of arsenic compounds in Chinese hamster lung fibroblast V69 cells [128]. This in vitro study is supported by subsequent in vivo studies [124]. Arsenic administration (3 mg/kg/day) for 14 days in rabbits resulted in significant oxidative stress, as revealed by reduction of whole blood GSH and elevation of thiobarbituric acid reactive substances (TBARS) and the index of nitrite/nitrate (NOx) levels. GTE administered to arsenic-treated rabbits for 14 days caused a significant elevation of the depleted GSH levels, which is likely attributed to the high polyphenol content of GTE [124]. Similarly, GT ameliorated arsenite (As III)-induced oxidative stress in Swiss albino mice by reducing the levels of lipid peroxides and protein carbonyl [130]. Co-administration of green tea extract also reduced arsenic (NaAsO2)-induced toxicity in liver, kidney and testicular and lipid peroxidation in experimental rats [122]. Interestingly, it has been reported that GTE significantly altered transport and uptake of As (III) across the Caco-2 cell model system, providing additional means of protection against arsenic exposure [119].

Table 5.

Green tea and arsenic-related injury

First author, yr [ref] Study design Preparation of Extract
or GTP Used
Treatments Effects of green tea
Cellular studies
Calatayud, 2011[119] Caco-2 cells GTE (Plantextrakt,
Germany)
As(III)
As(III)+GTE
↑ TEER
↓ As(III) Papp
Kim, 2015[120] BAE cells Pure EGCG As
As+EGCG
↑ Cell cytotoxicity↑ ROS formation
↑ Lipid peroxidation ↓ Catalase activity
Lee, 2011[121] HL60 cells Pure EGCG ATO
ATO+EGCG
↑ Mitochondria-induced apoptosis
↑ ROS formation to damage cell
Nakazato, 2005[123] Malignant B-cell lines Pure ECGC (WAKO Chemical Co. Tokyo, Japan) As2O3
As2O3+EGCG
↑ Apoptotic cell death
↑ ROS formation
Sinha, 2005[125] V79 cells Green tea leaves extracted with hot water and extracted with ethyl acetate NaAsO2
NaAsO2+GTE
↓ Chromatid breaks
↑ SOD and CAT
Sinha, 2007[126] Normal human lymphocytes Green tea leaves extracted with hot water and extracted with ethyl acetate As(III)
As(III)+GTE
↓ DNA damage and ROS formation
↓ ROS formation and lipid peroxidation
↑ CAT, SOD, and GPx
Sinha, 2003[128] V79 cells 2.5 g of dry tea brewed in 100 ml of boiled water for 5 min. GTE consisted of ECG, EGC, EGCG As
As+GTE
↓ Apoptosis
Sinha, 2005[129] V79 cells Green tea leaves extracted with hot water and extracted with ethyl acetate As
As+GTE
↑ Repair activity
Animal studies
Acharyya, 2014[117] Rats Leaf dust of green tea dried, crushed, and extracted by distilled water As
As+GTE
↓ Apoptotic hepatic degeneration
↓ DNA damage
↑ SOD1 protection
Acharyya, 2015[118] Rats Leaf dust of green tea dried, crushed, and extracted by distilled water NaAsO2
NaAsO2+GTE
↓ Intestinal tissue degeneration
↓ DNA-breakages
↓ Necrotic damage
Messarah, 2013[122] Rats 66 g dry leaves extracted per liter of tap water in drinking water NaAsO2
NaAsO2+GTE
↓ TBARS
↓ Oxidative stress
↑ Body weight
Raihan, 2009[124] Rabbits 3.75 g green tea boiled with arsenic-free fresh drinking (100 mL) for 10 min. As
As+GTE
↑ GSH
TBARS and NOx
Sinha, 2011[127] Swiss albino mice 0.5 g of dry tea leaves boiled with milli-Q water (5 mL) for 5 min. As(III)
As(III)+GTE
↓ 8 OHdG and OGG1
↑ DNA repair enzymes
Sinha, 2010[130] Swiss albino mice 0.5 g of dry tea leaves boiled with milli-Q water (5 mL) for 5 min. As(III)
As(III)+GTE
↓ Lipid peroxides
↓ Protein carbonyl
↑ CAT, SOD, GPx, GR, GST, and GSH

As, arsenic; As(III), arsenite; As2O3, arsenic trioxide; ATO, arsenic trioxide; BAE, bovine aortic endothelial cells; Caco-2 cells, human epithelial colorectal adenocarcinoma cells; CAT, catalase; EGCG, (−)-epigallocatechingallate; GPx, glutathione peroxidase; GR, glutathione reductase; GSH, glutathione; GST, glutathione-S-transferase; GTE, green tea extract; HL60, human promyelocytic leukemia cells; NaAsO2, sodium arsenite; NOx, nitrogen oxides; OGG1, 8-oxoguanine DNA glycosylase; 8 OHdG, 8-hydroxy-2′-deoxyguanosine; Papp, apparent permeability; SOD, superoxide dismutase; TBARS, thiobarbituric acid reactive substances; TEER, transepithelial resistance; V79, male Chinese hamster lung fibroblasts; ↑ increase; ↓ decrease.

Arsenic exposure is associated with DNA damage, changes in ploidy of cells, and nonrandom chromosome aberrations, leading to development of cancer. Both green tea and its polyphenols ameliorated arsenic-induced genotoxicity in the Chinese hamster lung fibroblast cells (V79) as determined by micronucleus assay [128]. In addition, tea extracts are effective in counteracting the sodium arsenite-induced clastogenicity (chromatid breaks, in particular) and inducing phase II detoxification enzymes, such as SOD and catalase, in the same V79 cells, suggesting that the antioxidant function of tea in reducing clastogenicity may be partly due to the induction of these enzymes [129]. Moreover, tea extracts reduced As III-induced DNA damage in human lymphocytes, as determined by comet assays. Tea also quenched excessive ROS production, reduced the elevated levels of lipid peroxidation, and increased the activities of catalase, SOD, and glutathione peroxidase. Furthermore, tea enhanced recovery from DNA damage, as confirmed by unscheduled DNA synthesis and pronounced expression of DNA repair enzyme poly (ADP-ribose) polymerase [126].

Tea extracts have been shown to reduce oxidative stress and induce DNA repair, all of which led to protection of arsenic-induced cancer. GTE reduced arsenic-induced formation of 8OHdG and induced arsenic-suppressed DNA repair enzymes, such as PARP1, DNA β-polymerase, XRCC1, DNA ligase III, DNA protein kinase (catalytic subunit), XRCC 4, DNA ligase IV, and DNA topoisomerase Iiβ, in Swiss albino mice [127]. GTE (≥10 mg/ml aqueous) restored arsenic-induced mutagenic DNA breaks and liver damages in rats via upregulation of cytosolic SOD [117]. Similarly, supplementation of tea extracts (10 mg/mL water) with NaAsO2 (0.6 ppm)/100 g b.w. for 28 days in rats protected against arsenic-induced oxidative damages to DNA and small intestinal tissues by upregulation of antioxidant systems. In addition, in situ incubation of rat intestinal loop with NaAsO2 alone (250 μM) or with aqueous GTE (250 mg/mL) for 24 h showed that small intestinal epithelial cells were significantly protected by tea extract against arsenic-associated necrotic/mutagenic damages, suggesting that GTE have a direct role in free radical scavenging; the latter is associated with protection against mutagenic DNA breakages and tissue necrosis induced by arsenic [118]. Arsenic is metabolized to monomethylated arsenic (MMA), and subsequently becomes dimethylated arsenic (DMA). It was also reported that the incidence of lung tumors induced by lung tumor initiators (4NQO) and dimethylarsinic acid (DMA(V)), and 8-oxodG, were suppressed by co-treatment with EGCG [131].

Conclusions and summary

Figure 1 summarizes the working mechanism of green tea in its protection against the five most common environmental toxins and toxicants. Green tea and its extracts reduce the environmental toxicant-induced oxidative stress and damages to DNA and cellular structures by down-regulation of HO-1, MMP12, NFκB, PI3K and ERK and up-regulation of antioxidant enzymes such as SOD and GSH, resulting in ROS quenching and lowering of oxidative products, including 8-OHdG. Green tea also suppresses toxicant-mediated carcinogenesis by preventing carcinogen-induced DNA damage, inhibiting cellular proliferation, inducing apoptosis and restoring connexin levels. On the other hand, green tea protects normal tissues and cells from toxicant-induced apoptosis by down-regulating caspases-3 and up-regulating SDO and GSH, thereby maintaining mitochondrial integrity. In addition, green tea attenuates signaling pathways such as NFκB and ERK pathways triggered by toxicants.

Figure 1.

Figure 1

Schematics representing the mechanisms of action for green tea in its protection against the five most common environmental toxins and toxicants. Green tea and its extracts suppress toxicant-induced cell proliferation and tumorigenesis by blocking carcinogen-mediated DNA damage. Green tea also quenches ROS and reduces oxidative stress by down-regulating MMP12, Bax/Bcl, HO-1, and NFκB, PI3K and ERK pathways and up-regulating SOD and GSH; the latter effects, coupled with down-regulation of caspases-3, offer additional protection of normal tissues from toxicant-induced apoptosis.

The emerging evidence shows the promising protective/detoxifying impacts of green tea on environmental toxins. The lack of clinical evidence contributes to the need for such future studies. For instance, the development of predictive biomarkers for green tea consumption in the human population will offer a better understanding of the interaction between green tea and endogenous and exogenous factors that affect its bioavailability, and help to establish the safe doses of green tea consumption. Further development of molecular markers for the biological effects of green tea will also help to elucidate its underlying mechanisms of action. The interaction of green tea ingredients and other dietary factors with similar toxicant-modulating activities may also warrant further investigation.

Acknowledgments

This study was supported by the National Center for Complementary and Integrative Health (NCCIH) of the National Institutes of Health, under grant U01AT006691 (C.L.S.). The contents of this manuscript are solely the responsibility of the authors and do not necessarily represent the official views of the NCCIH or the National Institutes of Health.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Toebak MJ, Gibbs S, Bruynzeel DP, Scheper RJ, Rustemeyer T. Dendritic cells: biology of the skin. Contact dermatitis. 2009;60:2–20. doi: 10.1111/j.1600-0536.2008.01443.x. [DOI] [PubMed] [Google Scholar]
  • 2.Cohen M. Environmental toxins and health–the health impact of pesticides. Australian family physician. 2007;36:1002–4. [PubMed] [Google Scholar]
  • 3.Hilgenkamp K. Environmental health:ecological perspectives. Jones & Bartlett Learning; 2006. [Google Scholar]
  • 4.Lanphear BP, Vorhees CV, Bellinger DC. Protecting children from environmental toxins. PLoS medicine. 2005;2:e61. doi: 10.1371/journal.pmed.0020061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Meadows-Oliver M. Environmental toxins. Journal of pediatric health care: official publication of National Association of Pediatric Nurse Associates & Practitioners. 2006;20:350–2. doi: 10.1016/j.pedhc.2006.06.005. [DOI] [PubMed] [Google Scholar]
  • 6.Pan-Montojo F, Schwarz M, Winkler C, Arnhold M, O’Sullivan GA, Pal A, et al. Environmental toxins trigger PD-like progression via increased alpha-synuclein release from enteric neurons in mice. Scientific reports. 2012;2:898. doi: 10.1038/srep00898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Stephenson J. CDC report on environmental toxins: some progress, some concerns. JAMA: the journal of the American Medical Association. 2003;289:1230, 3. doi: 10.1001/jama.289.10.1230. [DOI] [PubMed] [Google Scholar]
  • 8.Zimmerman HJ, Lewis JH. Chemical- and toxin-induced hepatotoxicity. Gastroenterology clinics of North America. 1995;24:1027–45. [PubMed] [Google Scholar]
  • 9.Cho HS, Kim S, Lee SY, Park JA, Kim SJ, Chun HS. Protective effect of the green tea component, L-theanine on environmental toxins-induced neuronal cell death. Neurotoxicology. 2008;29:656–62. doi: 10.1016/j.neuro.2008.03.004. [DOI] [PubMed] [Google Scholar]
  • 10.Alarie Y. Toxicity of fire smoke. Critical reviews in toxicology. 2002;32:259–89. doi: 10.1080/20024091064246. [DOI] [PubMed] [Google Scholar]
  • 11.Moszczynski P, Slowinski S, Lisiewicz J. [Enzyme deficiency in the lymphocytes of tobacco smokers] Przeglad lekarski. 1989;46:719–22. [PubMed] [Google Scholar]
  • 12.Patel BP, Rawal UM, Shah PM, Prajapati JA, Rawal RM, Dave TK, et al. Study of tobacco habits and alterations in enzymatic antioxidant system in oral cancer. Oncology. 2005;68:511–9. doi: 10.1159/000086995. [DOI] [PubMed] [Google Scholar]
  • 13.Bosetti C, Turati F, La Vecchia C. Hepatocellular carcinoma epidemiology. Best practice & research Clinical gastroenterology. 2014;28:753–70. doi: 10.1016/j.bpg.2014.08.007. [DOI] [PubMed] [Google Scholar]
  • 14.Bose T. Bitter correlationship between autoimmune hepatitis and smoking. Medical hypotheses. 2015;84:118–21. doi: 10.1016/j.mehy.2014.12.006. [DOI] [PubMed] [Google Scholar]
  • 15.Abbas HK. In: Aflatoxin and Food Safety. Abbas HK, editor. CRC Press; 2005. [Google Scholar]
  • 16.Perkins JT, Petriello MC, Newsome BJ, Hennig B. Polychlorinated biphenyls and links to cardiovascular disease. Environ Sci Pollut Res Int. 2016;23:2160–72. doi: 10.1007/s11356-015-4479-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Council CoIABoESaTDoEaLSNR. Critical Aspects of EPA’s IRIS Assessment of Inorganic Arsenic. 2014 [Google Scholar]
  • 18.Naujokas MF, Anderson B, Ahsan H, Aposhian HV, Graziano JH, Thompson C, et al. The broad scope of health effects from chronic arsenic exposure: update on a worldwide public health problem. Environ Health Perspect. 2013;121:295–302. doi: 10.1289/ehp.1205875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.De Vizcaya-Ruiz A, Barbier O, Ruiz-Ramos R, Cebrian ME. Biomarkers of oxidative stress and damage in human populations exposed to arsenic. Mutation research. 2009;674:85–92. doi: 10.1016/j.mrgentox.2008.09.020. [DOI] [PubMed] [Google Scholar]
  • 20.Shi H, Shi X, Liu KJ. Oxidative mechanism of arsenic toxicity and carcinogenesis. Mol Cell Biochem. 2004;255:67–78. doi: 10.1023/b:mcbi.0000007262.26044.e8. [DOI] [PubMed] [Google Scholar]
  • 21.Yang CS, Landau JM. Effects of tea consumption on nutrition and health. The Journal of nutrition. 2000;130:2409–12. doi: 10.1093/jn/130.10.2409. [DOI] [PubMed] [Google Scholar]
  • 22.Misra A, Chattopadhyay R, Banerjee S, Chattopadhyay DJ, Chatterjee IB. Black tea prevents cigarette smoke-induced oxidative damage of proteins in guinea pigs. The Journal of nutrition. 2003;133:2622–8. doi: 10.1093/jn/133.8.2622. [DOI] [PubMed] [Google Scholar]
  • 23.Sreekanth KS, Sabu MC, Varghese L, Manesh C, Kuttan G, Kuttan R. Antioxidant activity of Smoke Shield in-vitro and in-vivo. The Journal of pharmacy and pharmacology. 2003;55:847–53. doi: 10.1211/002235703765951474. [DOI] [PubMed] [Google Scholar]
  • 24.Hara Y. Green Tea: Health Benefits and Applications. CRC Press; 2001. [Google Scholar]
  • 25.Chen L, Lee MJ, Li H, Yang CS. Absorption, distribution, elimination of tea polyphenols in rats. Drug Metab Dispos. 1997;25:1045–50. [PubMed] [Google Scholar]
  • 26.Warden BA, Smith LS, Beecher GR, Balentine DA, Clevidence BA. Catechins are bioavailable in men and women drinking black tea throughout the day. The Journal of nutrition. 2001;131:1731–7. doi: 10.1093/jn/131.6.1731. [DOI] [PubMed] [Google Scholar]
  • 27.Lee MJ, Maliakal P, Chen L, Meng X, Bondoc FY, Prabhu S, et al. Pharmacokinetics of tea catechins after ingestion of green tea and (−)-epigallocatechin-3-gallate by humans: formation of different metabolites and individual variability. Cancer Epidemiol Biomarkers Prev. 2002;11:1025–32. [PubMed] [Google Scholar]
  • 28.Lambert JD, Yang CS. Mechanisms of cancer prevention by tea constituents. The Journal of nutrition. 2003;133:3262S–7S. doi: 10.1093/jn/133.10.3262S. [DOI] [PubMed] [Google Scholar]
  • 29.Dou QP. Molecular mechanisms of green tea polyphenols. Nutr Cancer. 2009;61:827–35. doi: 10.1080/01635580903285049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lu H, Meng X, Li C, Sang S, Patten C, Sheng S, et al. Glucuronides of tea catechins: enzymology of biosynthesis and biological activities. Drug Metab Dispos. 2003;31:452–61. doi: 10.1124/dmd.31.4.452. [DOI] [PubMed] [Google Scholar]
  • 31.Lu H, Meng X, Yang CS. Enzymology of methylation of tea catechins and inhibition of catechol-O-methyltransferase by (−)-epigallocatechin gallate. Drug Metab Dispos. 2003;31:572–9. doi: 10.1124/dmd.31.5.572. [DOI] [PubMed] [Google Scholar]
  • 32.Vaidyanathan JB, Walle T. Glucuronidation and sulfation of the tea flavonoid (−)-epicatechin by the human and rat enzymes. Drug Metab Dispos. 2002;30:897–903. doi: 10.1124/dmd.30.8.897. [DOI] [PubMed] [Google Scholar]
  • 33.Sang S, Lambert JD, Ho CT, Yang CS. The chemistry and biotransformation of tea constituents. Pharmacol Res. 2011;64:87–99. doi: 10.1016/j.phrs.2011.02.007. [DOI] [PubMed] [Google Scholar]
  • 34.Donovan JL, Crespy V, Manach C, Morand C, Besson C, Scalbert A, et al. Catechin is metabolized by both the small intestine and liver of rats. The Journal of nutrition. 2001;131:1753–7. doi: 10.1093/jn/131.6.1753. [DOI] [PubMed] [Google Scholar]
  • 35.Boutin JA, Thomassin J, Siest G, Cartier A. Heterogeneity of hepatic microsomal UDP-glucuronosyltransferase activities. Conjugations of phenolic and monoterpenoid aglycones in control and induced rats and guinea pigs. Biochem Pharmacol. 1985;34:2235–49. doi: 10.1016/0006-2952(85)90777-4. [DOI] [PubMed] [Google Scholar]
  • 36.Strassburg CP, Nguyen N, Manns MP, Tukey RH. UDP-glucuronosyltransferase activity in human liver and colon. Gastroenterology. 1999;116:149–60. doi: 10.1016/s0016-5085(99)70239-8. [DOI] [PubMed] [Google Scholar]
  • 37.Matsui M, Homma H. Biochemistry and molecular biology of drug-metabolizing sulfotransferase. Int J Biochem. 1994;26:1237–47. doi: 10.1016/0020-711x(94)90093-0. [DOI] [PubMed] [Google Scholar]
  • 38.Shali NA, Curtis CG, Powell GM, Roy AB. Sulphation of the flavonoids quercetin and catechin by rat liver. Xenobiotica. 1991;21:881–93. doi: 10.3109/00498259109039528. [DOI] [PubMed] [Google Scholar]
  • 39.Flohe L. Catechol-O-methyltransferase. Int Pharmacopsychiatry. 1974;9:52–60. doi: 10.1159/000468115. [DOI] [PubMed] [Google Scholar]
  • 40.HEIKAL TMA-T HMOSSA, ABDEL RASOUL MONAA, MAREI GEHANIKH. THE AMELIORATING EFFECTS OF GREEN TEA EXTRACT AGAINST CYROMAZINE AND CHLORPYRIFOS INDUCED LIVER TOXICITY IN MALE RATS. Asian Journal of Pharmaceutical & Clinical Research. 2013;6:48. [Google Scholar]
  • 41.Hou RR, Chen JZ, Chen H, Kang XG, Li MG, Wang BR. Neuroprotective effects of (−)-epigallocatechin-3-gallate (EGCG) on paraquat-induced apoptosis in PC12 cells. Cell biology international. 2008;32:22–30. doi: 10.1016/j.cellbi.2007.08.007. [DOI] [PubMed] [Google Scholar]
  • 42.Kamalden TA, Ji D, Osborne NN. Rotenone-induced death of RGC-5 cells is caspase independent, involves the JNK and p38 pathways and is attenuated by specific green tea flavonoids. Neurochemical research. 2012;37:1091–101. doi: 10.1007/s11064-012-0713-5. [DOI] [PubMed] [Google Scholar]
  • 43.Kim HR, Park BK, Oh YM, Lee YS, Lee DS, Kim HK, et al. Green tea extract inhibits paraquat-induced pulmonary fibrosis by suppression of oxidative stress and endothelin-l expression. Lung. 2006;184:287–95. doi: 10.1007/s00408-005-2592-x. [DOI] [PubMed] [Google Scholar]
  • 44.Korany NS, Ezzat BA. Prophylactic effect of green tea and Nigella sativa extracts against fenitrothion-induced toxicity in rat parotid gland. Archives of oral biology. 2011;56:1339–46. doi: 10.1016/j.archoralbio.2011.05.006. [DOI] [PubMed] [Google Scholar]
  • 45.Moldzio R, Radad K, Krewenka C, Kranner B, Duvigneau JC, Wang Y, et al. Effects of epigallocatechin gallate on rotenone-injured murine brain cultures. Journal of neural transmission. 2010;117:5–12. doi: 10.1007/s00702-009-0284-z. [DOI] [PubMed] [Google Scholar]
  • 46.Pan T, Fei J, Zhou X, Jankovic J, Le W. Effects of green tea polyphenols on dopamine uptake and on MPP+ -induced dopamine neuron injury. Life sciences. 2003;72:1073–83. doi: 10.1016/s0024-3205(02)02347-0. [DOI] [PubMed] [Google Scholar]
  • 47.Ruch RJ, Cheng SJ, Klaunig JE. Prevention of cytotoxicity and inhibition of intercellular communication by antioxidant catechins isolated from Chinese green tea. Carcinogenesis. 1989;10:1003–8. doi: 10.1093/carcin/10.6.1003. [DOI] [PubMed] [Google Scholar]
  • 48.Sai K, Kanno J, Hasegawa R, Trosko JE, Inoue T. Prevention of the down-regulation of gap junctional intercellular communication by green tea in the liver of mice fed pentachlorophenol. Carcinogenesis. 2000;21:1671–6. doi: 10.1093/carcin/21.9.1671. [DOI] [PubMed] [Google Scholar]
  • 49.Tai KK, Truong DD. (−)-Epigallocatechin-3-gallate (EGCG), a green tea polyphenol, reduces dichlorodiphenyl-trichloroethane (DDT)-induced cell death in dopaminergic SHSY-5Y cells. Neuroscience letters. 2010;482:183–7. doi: 10.1016/j.neulet.2010.06.018. [DOI] [PubMed] [Google Scholar]
  • 50.Tanaka R. Protective effects of (−)-epigallocatechin gallate and (+)-catechin on paraquat-induced genotoxicity in cultured cells. The Journal of toxicological sciences. 2000;25:199–204. doi: 10.2131/jts.25.3_199. [DOI] [PubMed] [Google Scholar]
  • 51.Umemura T, Kai S, Hasegawa R, Kanki K, Kitamura Y, Nishikawa A, et al. Prevention of dual promoting effects of pentachlorophenol, an environmental pollutant, on diethylnitrosamine-induced hepato- and cholangiocarcinogenesis in mice by green tea infusion. Carcinogenesis. 2003;24:1105–9. doi: 10.1093/carcin/bgg053. [DOI] [PubMed] [Google Scholar]
  • 52.Siegel GJ, Chauhan NB. Neurotrophic factors in Alzheimer’s and Parkinson’s disease brain. Brain research Brain research reviews. 2000;33:199–227. doi: 10.1016/s0165-0173(00)00030-8. [DOI] [PubMed] [Google Scholar]
  • 53.Schipper HM, Liberman A, Stopa EG. Neural heme oxygenase-1 expression in idiopathic Parkinson’s disease. Experimental neurology. 1998;150:60–8. doi: 10.1006/exnr.1997.6752. [DOI] [PubMed] [Google Scholar]
  • 54.Bothe H, Gassmann K, Gotz C, Fritsche E, Abel J, Haarmann-Stemmann T. Epigallocatechin-3-gallate does not affect the activity of enzymes involved in metabolic activation and cellular excretion of benzo[a]pyrene in human colon carcinoma cells. Toxicology letters. 2011;203:258–64. doi: 10.1016/j.toxlet.2011.03.026. [DOI] [PubMed] [Google Scholar]
  • 55.Roomi MW, Roomi NW, Kalinovsky T, Rath M, Niedzwiecki A. Chemopreventive effect of a novel nutrient mixture on lung tumorigenesis induced by urethane in male A/J mice. Tumori. 2009;95:508–13. doi: 10.1177/030089160909500417. [DOI] [PubMed] [Google Scholar]
  • 56.Kuttan R, Kuttan G, Joseph S, Ajith TA, Mohan M, Srimal RC. Antimutagenicity of herbal detoxification formula Smoke Shield against environmental mutagens. Journal of experimental & clinical cancer research: CR. 2004;23:61–8. [PubMed] [Google Scholar]
  • 57.Chan KH, Ho SP, Yeung SC, So WH, Cho CH, Koo MW, et al. Chinese green tea ameliorates lung injury in cigarette smoke-exposed rats. Respiratory medicine. 2009;103:1746–54. doi: 10.1016/j.rmed.2009.04.027. [DOI] [PubMed] [Google Scholar]
  • 58.Abe M, Suzuki N, Yoshida M, Usuda K, Furukawa S, Juneja LR, et al. Possible carcinogenic risks of copper gluconate and their prevention by co-administered green tea catechins evaluated by a rat medium-term multi-organ carcinogenicity bioassay protocol. Food and chemical toxicology. 2008;46:1760–70. doi: 10.1016/j.fct.2008.01.024. [DOI] [PubMed] [Google Scholar]
  • 59.Chan KH, Chan SC, Yeung SC, Man RY, Ip MS, Mak JC. Inhibitory effect of Chinese green tea on cigarette smoke-induced up-regulation of airway neutrophil elastase and matrix metalloproteinase-12 via antioxidant activity. Free radical research. 2012;46:1123–9. doi: 10.3109/10715762.2012.692786. [DOI] [PubMed] [Google Scholar]
  • 60.Fiala ES, Sohn OS, Wang CX, Seibert E, Tsurutani J, Dennis PA, et al. Induction of preneoplastic lung lesions in guinea pigs by cigarette smoke inhalation and their exacerbation by high dietary levels of vitamins C and E. Carcinogenesis. 2005;26:605–12. doi: 10.1093/carcin/bgh341. [DOI] [PubMed] [Google Scholar]
  • 61.Hakim IA, Chow HH, Harris RB. Green tea consumption is associated with decreased DNA damage among GSTM1-positive smokers regardless of their hOGG1 genotype. The Journal of nutrition. 2008;138:1567S–71S. doi: 10.1093/jn/138.8.1567S. [DOI] [PubMed] [Google Scholar]
  • 62.Holzer N, Braun KF, Ehnert S, Egana JT, Schenck TL, Buchholz A, et al. Green tea protects human osteoblasts from cigarette smoke-induced injury: possible clinical implication. Langenbeck’s archives of surgery/Deutsche Gesellschaft fur Chirurgie. 2012;397:467–74. doi: 10.1007/s00423-011-0882-8. [DOI] [PubMed] [Google Scholar]
  • 63.Kaneko T, Tahara S, Takabayashi F. Protective Effect of Fluvastatin, an HMG-CoA Reductase Inhibitor, on the Formation of 8-Oxo-2′-deoxyguanosine in the Nuclear DNA of Hamster Pancreas after a Single Administration of N-Nitrosobis (2-oxopropyl) amine. Biological and Pharmaceutical Bulletin. 2003;26:1245–8. doi: 10.1248/bpb.26.1245. [DOI] [PubMed] [Google Scholar]
  • 64.Khoi PN, Park JS, Kim JH, Xia Y, Kim NH, Kim KK, et al. (−)-Epigallocatechin-3-gallate blocks nicotine-induced matrix metalloproteinase-9 expression and invasiveness via suppression of NF-κB and AP-1 in endothelial cells. International journal of oncology. 2013;43:868–76. doi: 10.3892/ijo.2013.2006. [DOI] [PubMed] [Google Scholar]
  • 65.Kim HK, Yang T-H, Cho H-Y. Antifibrotic effects of green tea on in vitro and in vivo models of liver fibrosis. World journal of gastroenterology: WJG. 2009;15:5200. doi: 10.3748/wjg.15.5200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Leanderson P, Faresjo AO, Tagesson C. Green tea polyphenols inhibit oxidant-induced DNA strand breakage in cultured lung cells. Free radical biology & medicine. 1997;23:235–42. doi: 10.1016/s0891-5849(96)00590-4. [DOI] [PubMed] [Google Scholar]
  • 67.Lee IP, Kim YH, Kang MH, Roberts C, Shim JS, Roh JK. Chemopreventive effect of green tea (Camellia sinensis) against cigarette smoke-induced mutations (SCE) in humans. Journal of cellular biochemistry Supplement. 1997;27:68–75. [PubMed] [Google Scholar]
  • 68.Lu G, Liao J, Yang G, Reuhl KR, Hao X, Yang CS. Inhibition of adenoma progression to adenocarcinoma in a 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-induced lung tumorigenesis model in A/J mice by tea polyphenols and caffeine. Cancer research. 2006;66:11494–501. doi: 10.1158/0008-5472.CAN-06-1497. [DOI] [PubMed] [Google Scholar]
  • 69.Lubet RA, Yang CS, Lee MJ, Hara Y, Kapetanovic IM, Crowell JA, et al. Preventive effects of polyphenon E on urinary bladder and mammary cancers in rats and correlations with serum and urine levels of tea polyphenols. Molecular cancer therapeutics. 2007;6:2022–8. doi: 10.1158/1535-7163.MCT-07-0058. [DOI] [PubMed] [Google Scholar]
  • 70.Maliakal P, Sankpal UT, Basha R, Maliakal C, Ledford A, Wanwimolruk S. Relevance of drug metabolizing enzyme activity modulation by tea polyphenols in the inhibition of esophageal tumorigenesis. Medicinal chemistry. 2011;7:480–7. doi: 10.2174/157340611796799096. [DOI] [PubMed] [Google Scholar]
  • 71.Rathore K, Wang HC. Green tea catechin extract in intervention of chronic breast cell carcinogenesis induced by environmental carcinogens. Molecular carcinogenesis. 2012;51:280–9. doi: 10.1002/mc.20844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Roy P, Nigam N, Singh M, George J, Srivastava S, Naqvi H, et al. Tea polyphenols inhibit cyclooxygenase-2 expression and block activation of nuclear factor-kappa B and Akt in diethylnitrosoamine induced lung tumors in Swiss mice. Investigational new drugs. 2010;28:466–71. doi: 10.1007/s10637-009-9274-0. [DOI] [PubMed] [Google Scholar]
  • 73.Sagara Y, Miyata Y, Nomata K, Hayashi T, Kanetake H. Green tea polyphenol suppresses tumor invasion and angiogenesis in N-butyl-(-4-hydroxybutyl) nitrosamine-induced bladder cancer. Cancer epidemiology. 2010;34:350–4. doi: 10.1016/j.canep.2010.03.001. [DOI] [PubMed] [Google Scholar]
  • 74.Saiwichai T, Sangalangkarn V, Kawahara K, Oyama Y, Chaichalotornkul S, Narkpinit S, et al. Green tea extract supplement inhibition of HMGB1 release in rats exposed to cigarette smoke. The Southeast Asian journal of tropical medicine and public health. 2010;41:250–8. [PubMed] [Google Scholar]
  • 75.Sato D, Matsushima M. Preventive effects of urinary bladder tumors induced by N-butyl-N-(4-hydroxybutyl)-nitrosamine in rat by green tea leaves. International journal of urology: official journal of the Japanese Urological Association. 2003;10:160–6. doi: 10.1046/j.1442-2042.2003.00587.x. [DOI] [PubMed] [Google Scholar]
  • 76.Shimizu M, Adachi S, Masuda M, Kozawa O, Moriwaki H. Cancer chemoprevention with green tea catechins by targeting receptor tyrosine kinases. Molecular nutrition & food research. 2011;55:832–43. doi: 10.1002/mnfr.201000622. [DOI] [PubMed] [Google Scholar]
  • 77.Syed DN, Afaq F, Kweon MH, Hadi N, Bhatia N, Spiegelman VS, et al. Green tea polyphenol EGCG suppresses cigarette smoke condensate-induced NF-kappaB activation in normal human bronchial epithelial cells. Oncogene. 2007;26:673–82. doi: 10.1038/sj.onc.1209829. [DOI] [PubMed] [Google Scholar]
  • 78.Takahashi H, Nomata K, Mori K, Matsuo M, Miyaguchi T, Noguchi M, et al. The preventive effect of green tea on the gap junction intercellular communication in renal epithelial cells treated with a renal carcinogen. Anticancer research. 2004;24:3757–62. [PubMed] [Google Scholar]
  • 79.Vermeer IT, Moonen EJ, Dallinga JW, Kleinjans JC, van Maanen JM. Effect of ascorbic acid and green tea on endogenous formation of N-nitrosodimethylamine and N-nitrosopiperidine in humans. Mutation research. 1999;428:353–61. doi: 10.1016/s1383-5742(99)00061-7. [DOI] [PubMed] [Google Scholar]
  • 80.Weitberg AB, Corvese D. The effect of epigallocatechin galleate and sarcophytol A on DNA strand breakage induced by tobacco-specific nitrosamines and stimulated human phagocytes. Journal of experimental & clinical cancer research: CR. 1999;18:433–7. [PubMed] [Google Scholar]
  • 81.Witschi H, Espiritu I, Yu M, Willits NH. The effects of phenethyl isothiocyanate, N-acetylcysteine and green tea on tobacco smoke-induced lung tumors in strain A/J mice. Carcinogenesis. 1998;19:1789–94. doi: 10.1093/carcin/19.10.1789. [DOI] [PubMed] [Google Scholar]
  • 82.Yang CH, Lin CY, Yang JH, Liou SY, Li PC, Chien CT. Supplementary catechins attenuate cooking-oil-fumes-induced oxidative stress in rat lung. The Chinese journal of physiology. 2009;52:151–9. doi: 10.4077/cjp.2009.amh022. [DOI] [PubMed] [Google Scholar]
  • 83.Zhou R, Zhou Y, Chen D, Li S, Haug A. Effects of soaking temperature and soaking time during preparation of water extract of tea on anticlastogenicity against environmental tobacco smoke in the sister-chromatid exchange assay. Toxicology letters. 2000;115:23–32. doi: 10.1016/s0378-4274(00)00171-5. [DOI] [PubMed] [Google Scholar]
  • 84.Yang CS, Wang X. Green tea and cancer prevention. Nutr Cancer. 2010;62:931–7. doi: 10.1080/01635581.2010.509536. [DOI] [PubMed] [Google Scholar]
  • 85.Bennett JW. Mycotoxins, mycotoxicoses, mycotoxicology and Mycopathologia. Mycopathologia. 1987;100:3–5. doi: 10.1007/BF00769561. [DOI] [PubMed] [Google Scholar]
  • 86.Bennett JW, Klich M. Mycotoxins. Clinical microbiology reviews. 2003;16:497–516. doi: 10.1128/CMR.16.3.497-516.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Squire RA. Ranking animal carcinogens: a proposed regulatory approach. Science. 1981;214:877–80. doi: 10.1126/science.7302565. [DOI] [PubMed] [Google Scholar]
  • 88.Khlangwiset P, Shephard GS, Wu F. Aflatoxins and growth impairment: a review. Critical reviews in toxicology. 2011;41:740–55. doi: 10.3109/10408444.2011.575766. [DOI] [PubMed] [Google Scholar]
  • 89.Groopman JD, Kensler TW, Wild CP. Protective interventions to prevent aflatoxin-induced carcinogenesis in developing countries. Annual review of public health. 2008;29:187–203. doi: 10.1146/annurev.publhealth.29.020907.090859. [DOI] [PubMed] [Google Scholar]
  • 90.Gonzalez HH, Martinez EJ, Pacin AM, Resnik SL, Sydenham EW. Natural co-occurrence of fumonisins, deoxynivalenol, zearalenone and aflatoxins in field trial corn in Argentina. Food additives and contaminants. 1999;16:565–9. doi: 10.1080/026520399283704. [DOI] [PubMed] [Google Scholar]
  • 91.Peraica M, Radic B, Lucic A, Pavlovic M. Toxic effects of mycotoxins in humans. Bulletin of the World Health Organization. 1999;77:754–66. [PMC free article] [PubMed] [Google Scholar]
  • 92.Hour TC, Liang YC, Chu IS, Lin JK. Inhibition of eleven mutagens by various tea extracts, (−)epigallocatechin-3-gallate, gallic acid and caffeine. Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association. 1999;37:569–79. doi: 10.1016/s0278-6915(99)00031-9. [DOI] [PubMed] [Google Scholar]
  • 93.Huang TYJTLLHBMWZLYLY, Wang K, Shi J, Cui Y, Zhang ZQ, Zhang L, Sun S, Martin C, Wang JS. Phase IIa Chemoprevention Trial of Green Tea Polyphenols in High-Risk Individuals of Liver Cancer: I. Design, Clinical Outcomes, and Baseline Biomarker Data. Int J Cancer Prev. 2004;1:269–80. [Google Scholar]
  • 94.Ito Y, Ohnishi S, Fujie K. Chromosome aberrations induced by aflatoxin B1 in rat bone marrow cells in vivo and their suppression by green tea. Mutation research. 1989;222:253–61. doi: 10.1016/0165-1218(89)90141-9. [DOI] [PubMed] [Google Scholar]
  • 95.Luo H, Tang L, Tang M, Billam M, Huang T, Yu J, et al. Phase IIa chemoprevention trial of green tea polyphenols in high-risk individuals of liver cancer: modulation of urinary excretion of green tea polyphenols and 8-hydroxydeoxyguanosine. Carcinogenesis. 2006;27:262–8. doi: 10.1093/carcin/bgi147. [DOI] [PubMed] [Google Scholar]
  • 96.Luo HC SB, Gao W, Yu J, Tang L, Wang JS. Metabolic profiling in validation of plasma biomarkers for green tea polyphenols. Metabolomics. 2006;2:235–41. [Google Scholar]
  • 97.M IYI. Suppressive Effect of (−)-Epigallocatechin Gallate on Aflatoxin B1-induced Chromosome Aberrations in Rat Bone Marrow Cells. J Health Sci. 2001;47:248–57. [Google Scholar]
  • 98.Marnewick JL, Batenburg W, Swart P, Joubert E, Swanevelder S, Gelderblom WC. Ex vivo modulation of chemical-induced mutagenesis by subcellular liver fractions of rats treated with rooibos (Aspalathus linearis) tea, honeybush (Cyclopia intermedia) tea, as well as green and black (Camellia sinensis) teas. Mutation research. 2004;558:145–54. doi: 10.1016/j.mrgentox.2003.12.003. [DOI] [PubMed] [Google Scholar]
  • 99.Marnewick JL, van der Westhuizen FH, Joubert E, Swanevelder S, Swart P, Gelderblom WC. Chemoprotective properties of rooibos (Aspalathus linearis), honeybush (Cyclopia intermedia) herbal and green and black (Camellia sinensis) teas against cancer promotion induced by fumonisin B1 in rat liver. Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association. 2009;47:220–9. doi: 10.1016/j.fct.2008.11.004. [DOI] [PubMed] [Google Scholar]
  • 100.Mo HZ, Zhang H, Wu QH, Hu LB. Inhibitory effects of tea extract on aflatoxin production by Aspergillus flavus. Letters in applied microbiology. 2013;56:462–6. doi: 10.1111/lam.12073. [DOI] [PubMed] [Google Scholar]
  • 101.Qin G, Gopalan-Kriczky P, Su J, Ning Y, Lotlikar PD. Inhibition of aflatoxin B1-induced initiation of hepatocarcinogenesis in the rat by green tea. Cancer letters. 1997;112:149–54. doi: 10.1016/s0304-3835(96)04568-5. [DOI] [PubMed] [Google Scholar]
  • 102.Qin G, Ning Y, Lotlikar PD. Chemoprevention of aflatoxin B1-initiated and carbon tetrachloride-promoted hepatocarcinogenesis in the rat by green tea. Nutr Cancer. 2000;38:215–22. doi: 10.1207/S15327914NC382_11. [DOI] [PubMed] [Google Scholar]
  • 103.Snijman PW, Swanevelder S, Joubert E, Green IR, Gelderblom WC. The antimutagenic activity of the major flavonoids of rooibos (Aspalathus linearis): some dose-response effects on mutagen activation-flavonoid interactions. Mutation research. 2007;631:111–23. doi: 10.1016/j.mrgentox.2007.03.009. [DOI] [PubMed] [Google Scholar]
  • 104.Tang L, Tang M, Xu L, Luo H, Huang T, Yu J, et al. Modulation of aflatoxin biomarkers in human blood and urine by green tea polyphenols intervention. Carcinogenesis. 2008;29:411–7. doi: 10.1093/carcin/bgn008. [DOI] [PubMed] [Google Scholar]
  • 105.Tulayakul P, Dong KS, Li JY, Manabe N, Kumagai S. The effect of feeding piglets with the diet containing green tea extracts or coumarin on in vitro metabolism of aflatoxin B1 by their tissues. Toxicon: official journal of the International Society on Toxinology. 2007;50:339–48. doi: 10.1016/j.toxicon.2007.04.005. [DOI] [PubMed] [Google Scholar]
  • 106.Wang JS, Luo H, Wang P, Tang L, Yu J, Huang T, et al. Validation of green tea polyphenol biomarkers in a phase II human intervention trial. Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association. 2008;46:232–40. doi: 10.1016/j.fct.2007.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wang ZY, Cheng SJ, Zhou ZC, Athar M, Khan WA, Bickers DR, et al. Antimutagenic activity of green tea polyphenols. Mutation research. 1989;223:273–85. doi: 10.1016/0165-1218(89)90120-1. [DOI] [PubMed] [Google Scholar]
  • 108.de Boer JG. Protection by dietary compounds against mutation in a transgenic rodent. The Journal of nutrition. 2001;131:3082S–6S. doi: 10.1093/jn/131.11.3082S. [DOI] [PubMed] [Google Scholar]
  • 109.Li Y. Comparative study on the inhibitory effect of green tea, coffee and levamisole on the hepatocarcinogenic action of diethylnitrosamine. Zhonghua zhong liu za zhi [Chinese journal of oncology] 1991;13:193–5. [PubMed] [Google Scholar]
  • 110.Guyton KZ, Kensler TW. Prevention of liver cancer. Current oncology reports. 2002;4:464–70. doi: 10.1007/s11912-002-0057-4. [DOI] [PubMed] [Google Scholar]
  • 111.Petriello MC, Newsome B, Hennig B. Influence of nutrition in PCB-induced vascular inflammation. Environ Sci Pollut Res Int. 2014;21:6410–8. doi: 10.1007/s11356-013-1549-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Han SG, Han SS, Toborek M, Hennig B. EGCG protects endothelial cells against PCB 126-induced inflammation through inhibition of AhR and induction of Nrf2-regulated genes. Toxicol Appl Pharmacol. 2012;261:181–8. doi: 10.1016/j.taap.2012.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Morita K, Matsueda T, Iida T. [Effect of green tea (matcha) on gastrointestinal tract absorption of polychlorinated biphenyls, polychlorinated dibenzofurans and polychlorinated dibenzo-p-dioxins in rats] Fukuoka Igaku Zasshi. 1997;88:162–8. [PubMed] [Google Scholar]
  • 114.Newsome BJ, Petriello MC, Han SG, Murphy MO, Eske KE, Sunkara M, et al. Green tea diet decreases PCB 126-induced oxidative stress in mice by up-regulating antioxidant enzymes. J Nutr Biochem. 2014;25:126–35. doi: 10.1016/j.jnutbio.2013.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ramadass P, Meerarani P, Toborek M, Robertson LW, Hennig B. Dietary flavonoids modulate PCB-induced oxidative stress, CYP1A1 induction, and AhR-DNA binding activity in vascular endothelial cells. Toxicol Sci. 2003;76:212–9. doi: 10.1093/toxsci/kfg227. [DOI] [PubMed] [Google Scholar]
  • 116.Weisburger JH, Nagao M, Wakabayashi K, Oguri A. Prevention of heterocyclic amine formation by tea and tea polyphenols. Cancer letters. 1994;83:143–7. doi: 10.1016/0304-3835(94)90311-5. [DOI] [PubMed] [Google Scholar]
  • 117.Acharyya N, Chattopadhyay S, Maiti S. Chemoprevention against arsenic-induced mutagenic DNA breakage and apoptotic liver damage in rat via antioxidant and SOD1 upregulation by green tea (Camellia sinensis) which recovers broken DNA resulted from arsenic-H2O2 related in vitro oxidant stress. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2014;32:338–61. doi: 10.1080/10590501.2014.967061. [DOI] [PubMed] [Google Scholar]
  • 118.Acharyya N, Sajed Ali S, Deb B, Chattopadhyay S, Maiti S. Green tea (Camellia sinensis) alleviates arsenic-induced damages to DNA and intestinal tissues in rat and in situ intestinal loop by reinforcing antioxidant system. Environ Toxicol. 2015;30:1033–44. doi: 10.1002/tox.21977. [DOI] [PubMed] [Google Scholar]
  • 119.Calatayud M, Devesa V, Montoro R, Velez D. In vitro study of intestinal transport of arsenite, monomethylarsonous acid, and dimethylarsinous acid by Caco-2 cell line. Toxicology letters. 2011;204:127–33. doi: 10.1016/j.toxlet.2011.04.023. [DOI] [PubMed] [Google Scholar]
  • 120.Kim JY, Choi JY, Lee HJ, Byun CJ, Park JH, Park JH, et al. The Green Tea Component (−)-Epigallocatechin-3-Gallate Sensitizes Primary Endothelial Cells to Arsenite-Induced Apoptosis by Decreasing c-Jun N-Terminal Kinase-Mediated Catalase Activity. PLoS One. 2015;10:e0138590. doi: 10.1371/journal.pone.0138590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lee TC, Cheng IC, Shue JJ, Wang TC. Cytotoxicity of arsenic trioxide is enhanced by (−)-epigallocatechin-3-gallate via suppression of ferritin in cancer cells. Toxicol Appl Pharmacol. 2011;250:69–77. doi: 10.1016/j.taap.2010.10.005. [DOI] [PubMed] [Google Scholar]
  • 122.Messarah M, Saoudi M, Boumendjel A, Kadeche L, Boulakoud MS, El Feki A. Green tea extract alleviates arsenic-induced biochemical toxicity and lipid peroxidation in rats. Toxicol Ind Health. 2013;29:349–59. doi: 10.1177/0748233711433934. [DOI] [PubMed] [Google Scholar]
  • 123.Nakazato T, Ito K, Ikeda Y, Kizaki M. Green tea component, catechin, induces apoptosis of human malignant B cells via production of reactive oxygen species. Clin Cancer Res. 2005;11:6040–9. doi: 10.1158/1078-0432.CCR-04-2273. [DOI] [PubMed] [Google Scholar]
  • 124.Raihan SZ, Chowdhury AK, Rabbani GH, Marni F, Ali MS, Nahar L, et al. Effect of aqueous extracts of black and green teas in arsenic-induced toxicity in rabbits. Phytotherapy research: PTR. 2009;23:1603–8. doi: 10.1002/ptr.2827. [DOI] [PubMed] [Google Scholar]
  • 125.Sinha D, Bhattacharya RK, Siddiqi M, Roy M. Amelioration of sodium arsenite-induced clastogenicity by tea extracts in Chinese hamster v79 cells. J Environ Pathol Toxicol Oncol. 2005;24:129–40. doi: 10.1615/jenvpathtoxoncol.v24.i2.60. [DOI] [PubMed] [Google Scholar]
  • 126.Sinha D, Dey S, Bhattacharya RK, Roy M. In vitro mitigation of arsenic toxicity by tea polyphenols in human lymphocytes. J Environ Pathol Toxicol Oncol. 2007;26:207–20. doi: 10.1615/jenvironpatholtoxicoloncol.v26.i3.50. [DOI] [PubMed] [Google Scholar]
  • 127.Sinha D, Roy M. Antagonistic role of tea against sodium arsenite-induced oxidative DNA damage and inhibition of DNA repair in Swiss albino mice. J Environ Pathol Toxicol Oncol. 2011;30:311–22. doi: 10.1615/jenvironpatholtoxicoloncol.v30.i4.40. [DOI] [PubMed] [Google Scholar]
  • 128.Sinha D, Roy M, Dey S, Siddiqi M, Bhattacharya RK. Modulation of arsenic induced cytotoxicity by tea. Asian Pacific journal of cancer prevention: APJCP. 2003;4:233–7. [PubMed] [Google Scholar]
  • 129.Sinha D, Roy M, Siddiqi M, Bhattacharya RK. Arsenic-induced micronuclei formation in mammalian cells and its counteraction by tea. J Environ Pathol Toxicol Oncol. 2005;24:45–56. doi: 10.1615/jenvpathtoxoncol.v24.i1.50. [DOI] [PubMed] [Google Scholar]
  • 130.Sinha D, Roy S, Roy M. Antioxidant potential of tea reduces arsenite induced oxidative stress in Swiss albino mice. Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association. 2010;48:1032–9. doi: 10.1016/j.fct.2010.01.016. [DOI] [PubMed] [Google Scholar]
  • 131.Mizoi M, Takabayashi F, Nakano M, An Y, Sagesaka Y, Kato K, et al. The role of trivalent dimethylated arsenic in dimethylarsinic acid-promoted skin and lung tumorigenesis in mice: tumor-promoting action through the induction of oxidative stress. Toxicology letters. 2005;158:87–94. doi: 10.1016/j.toxlet.2005.03.009. [DOI] [PubMed] [Google Scholar]

RESOURCES