effect of dietary pretreatment and obesity on

165
The Pennsylvania State University The Graduate School Department of Food Science EFFECT OF DIETARY PRETREATMENT AND OBESITY ON (-)- EPIGALLOCATECHIN-3- GALLATE (EGCG) MEDIATED HEPATOTOXICITY AND THE UNDERLYING MECHANISM A Dissertation in Food Science by Karma James 2016 Karma James Submitted in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy May 2016

Upload: others

Post on 19-May-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

The Pennsylvania State University

The Graduate School

Department of Food Science

EFFECT OF DIETARY PRETREATMENT AND OBESITY ON (-)-

EPIGALLOCATECHIN-3- GALLATE (EGCG) MEDIATED HEPATOTOXICITY AND

THE UNDERLYING MECHANISM

A Dissertation in

Food Science

by

Karma James

2016 Karma James

Submitted in Partial Fulfillment

of the Requirements

for the Degree of

Doctor of Philosophy

May 2016

Page 2: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

The dissertation of Karma James was reviewed and approved* by the following:

Joshua Lambert

Associate Professor of Food Science

Co-Director, Center for Plant and Mushroom Foods for Health

Disseration Advisor

Chair of Committee

Ryan Elias

Frederick Sr. and Faith E. Rasmussen Professor of Food Science

Associate Professor of Food Science

Jairam Vanamala

Associate Professor of Food Science

Faculty Member, The Penn State Hershey Cancer Institute

Faculty Member, Center for Molecular Immunology and Infectious Disease

Gary Perdew

John T. and Paige S. Smith Professor in Agricultural Sciences

Director of the Center for Molecular Toxicology and Carcinogenesis

Robert Roberts

Professor of Food Science

Head of the Department of Food Science

*Signatures are on file in the Graduate School

Page 3: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

iii

ABSTRACT

A growing body of evidence suggests the hepatotoxic potential of high doses of EGCG.

In vitro several mechanisms of EGCG cytotoxicity have been examined; however potential

mechanisms of in vivo toxicity remain understudied. Additionally factors that can potentiate

EGCG induced hepatotoxicity, such as pre-existing physiological conditions that attenuate liver

function have not been investigated. With the lack of data to support an association between

dietary supplements and liver toxicity, I hypothesized that dietary pretreatment with EGCG

markedly decreases EGCG bioavailability and can potentially mitigate the toxic potential of

EGCG, and that ORFLD increases susceptibility to high dose EGCG-mediated

hepatotoxicity. In addition to the effect of these factors on high dose EGCG hepatotoxicity, I

further hypothesized that the underlying mechanism by which EGCG mediates

hepatotoxicity is through induction of mitochondrial oxidative stress and increased

mitochondrial dysfunction.

This study investigated the effect of dietary pretreatment with EGCG in male CF-1 mice.

EGCG pretreated (EP) mice were pretreated with 3.2mg/kg EGCG in their diet for 2 weeks, a

dietary dose that is equivalent to 500 mg/kg body weight. Non-pretreated (NP) and negative

control (NC) mice were given a basal diet. NP mice showed increased levels of ALT and

phosphorylated histone 2Ax compared to control mice. Pretreatment with EGCG reduced these

elevated levels. NP mice showed reduced levels of glutathione, which was partially ameliorated

in EP mice. In addition to blunting the markers of oxidative stress dietary pretreatment enhanced

hepatic level of antioxidant genes and reduced plasma and tissue concentrations of unconjugated

EGCG indicating that EGCG impacted its own bioavailability. In summary, these results show

that dietary pretreatment with EGCG reduced the bioavailability and hepatotoxic potential of

subsequent acute high dose oral bolus EGCG.

Page 4: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

iv

In another study we examined the effect of pre-existing obesity and ORFLD on EGCG-

mediated hepatotoxicity in mice. EGCG treatment decreased survival rates of both lean and obese

mice, but the effects were more pronounced in obese mice. EGCG caused increased liver injury

as seen in elevated levels of liver ALT in all treatment groups. EGCG treatment increased DNA

oxidative damage in both lean and obese mice. Markers of liver tissue damage assessed using

histopathology showed that EGCG caused a dose dependent increase in tissue apoptosis, necrosis,

hemorrhage, inflammation and hepatic lipidosis in both lean and obese mice. Lipid peroxidation

increased following EGCG treatment, and depleted glutathione levels were observed. Obese mice

exhibited significantly lower levels of glutathione compared to lean mice indicating elevated

levels of oxidative stress caused by EGCG treatment. This study showed that EGCG treatment

caused hepatotoxicity in lean and obese mice in a dose dependent manner, with the effects being

more pronounced in obese mice.

Our third study examined the effects of EGCG on hepatic markers of antioxidant

response and mitochondrial biogenesis/function in lean and obese mice. EGCG significantly

reduced hepatic antioxidant capacity. Lean and obese mice showed reduce gene and protein

expression, and activity of antioxidant enzymes. EGCG treatment significantly reduced

mitochondrial superoxide dismutase (Sod2) mRNA levels in lean mice; as well as catalase mRNA

levels in both lean and obese mice. Obese mice showed lower catalase activity and protein

expression following EGCG treatment. A significant decrease in glutathione peroxidase 1 (Gpx1)

protein expression was also seen. mRNA expression of glutathione transferase zeta 1 (Gstz1) and

peroxiredoxin were significantly decreased by EGCG. In response to oxidative stress induced by

EGCG, increases in gene expression of metallothionein transcription factor 1 (MTF-1) and

metallothionein I and II were observed. Reduced antioxidant activity was a result of the

impairment of antioxidant regulators by EGCG treatment. EGCG reduce mRNA levels of Sirtuin

3 (Sirt3), forkhead box O3 (Foxo3a) and nuclear factor (erythroid-derived 2)-like 2 (Nrf 2).

Page 5: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

v

EGCG treatment reduced mitochondrial biogenesis by decreasing mRNA levels of transcription

factor nuclear respiratory factor 1 (Nrf1), co-activator peroxisome proliferator-activator receptor

coactivator-1a (Pgc-1α) and mtDNA copy number in both lean and obese mice. EGCG impaired

mitochondrial function by reducing gene expression of mitochondrial transcription factors A,

(Tfam), B1 (Tfb1m) and B2 (Tfb2m), as well as subunits of mitochondrial Complex I and

mitochondrial complex III.

Overall, based on our results and observations, EGCG treatment induced hepatotoxicity

in lean and obese mice in a dose dependent manner and it induced oxidative stress by inhibiting

antioxidant response. Mitochondrial function was impaired based on reduced biogenesis and

inhibition of complexes following EGCG treatment. However, EGCG was shown to mitigate its

toxic effects by reducing its own bioavailability following chronic dietary treatment.

Page 6: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

vi

TABLE OF CONTENTS

List of Figures ......................................................................................................................... ix

List of Tables ........................................................................................................................... xi

Acknowledgements .................................................................................................................. xii

Literature Review and Research Objectives............................................................ 1

Green Tea Processing and Chemistry .............................................................................. 1 Putative Health Benefits of Green Tea and EGCG .......................................................... 4

Antioxidant Activity of EGCG ................................................................................ 5 Pro-oxidant Activity of EGCG ................................................................................. 5 Anti-Obesity Properties ............................................................................................ 7 Anti-Inflammatory Properties .................................................................................. 9 Anti-Diabetes Properties .......................................................................................... 10

Dietary supplements ......................................................................................................... 12 Impact of dosage form on bioavailability ........................................................................ 14 Potential Toxic Effects of EGCG and Green Tea Extracts .............................................. 19

Human studies/Case Reports .................................................................................... 19 Laboratory Studies ................................................................................................... 30

Dietary and Lifestyle Factors Affecting Risk of Green Tea-Related Liver Toxicity ....... 33 Research Hypothesis and Specific Aims .......................................................................... 38

Dietary Administration of the Green Tea Polyphenol, (-)- Epigallocatechin-3-

gallate Reduces the Bioavailability and Hepatotoxicity of Subsequent High Oral

Bolus Doses of (-)-Epigallocatechin-3-gallate ................................................................. 39

Abstract ............................................................................................................................ 39 Introduction ...................................................................................................................... 41 Materials and methods ..................................................................................................... 42

Chemicals and reagents ............................................................................................ 42 Animal studies .......................................................................................................... 44

Hepatotoxicity Studies ..................................................................................... 45 Bioavailability Studies ..................................................................................... 45

Determination of hepatotoxicity and oxidative stress .............................................. 46 Gene expression studies ........................................................................................... 46 HPLC analysis of EGCG .......................................................................................... 47 Statistical Analysis ................................................................................................... 47

Results .............................................................................................................................. 49 Effect of EGCG pretreatment on EGCG-induced hepatotoxicity and oxidative

stress ................................................................................................................. 49 Hepatic expression of antioxidant responsive genes ................................................ 50 Effect of EGCG pretreatment on EGCG bioavailability .......................................... 52

Discussion ........................................................................................................................ 54

Page 7: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

vii

Effect of Obesity on High Dose Oral EGCG-mediated Liver toxicity .................... 58

Abstract ............................................................................................................................ 58 Introduction ...................................................................................................................... 60 Materials and methods ..................................................................................................... 63

Chemicals and reagents ............................................................................................ 63 Animal care and treatment ....................................................................................... 64 Western blot analysis ............................................................................................... 65 Determination of hepatotoxicity ............................................................................... 66 Examination of Mitochondrial Structure .................................................................. 66 Measurement of hepatic oxidative stress .................................................................. 67 Statistical Analysis ................................................................................................... 68

Results .............................................................................................................................. 70 Effect of EGCG on general markers of toxicity ....................................................... 70 Effect of EGCG on general markers of liver toxicity............................................... 71 Histological evaluation of the hepatotoxic effect of EGCG ..................................... 73 Effect of EGCG on Mitochondrial Structure ........................................................... 76 Effect of EGCG on Markers of Oxidative Stress ..................................................... 79

Discussion ........................................................................................................................ 82

Effect of High Dose Oral (-)-Epigallocatechin-3-gallate (EGCG) on Hepatic

Mitochondrial Antioxidants, Biogenesis and Function .................................................... 91

Abstract ............................................................................................................................ 91 Introduction ...................................................................................................................... 93 Materials and methods ..................................................................................................... 96

Animal studies .......................................................................................................... 96 Gene expression studies ........................................................................................... 96 Western blot analysis ............................................................................................... 99 Antioxidant enzyme activity .................................................................................... 99

Glutathione peroxidase (GPx) .......................................................................... 99 Superoxide dismutase (SOD) ........................................................................... 100 Catalase Activity .............................................................................................. 100

Statistical Analysis ................................................................................................... 101 Results .............................................................................................................................. 102

Effect of EGCG on Antioxidant Response ............................................................... 102 Effect of EGCG on regulators of antioxidant genes ................................................. 108 Effect of EGCG on mitochondrial biogenesis .......................................................... 110 Effect of EGCG on factor involved in mitochondrial function ................................ 111

Discussion ........................................................................................................................ 115

Conclusions and Recommendation for Future Work .............................................. 128

Conclusions ...................................................................................................................... 128 Future Work ..................................................................................................................... 132

Effect of Other Lifestyle Factors on EGCG Mediated Liver Toxicity ..................... 132 Determination of Hepatotoxicity of Green Tea Extract ........................................... 133 Inhibition of Mitochondrial Proteins Involved in Mitochondrial Antioxidant

Response and Function by EGCG .................................................................... 134

Page 8: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

viii

Effect of EGCG Metabolites on EGCG Mediated Liver Toxicity ........................... 136

References ................................................................................................................................ 138

Page 9: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

ix

LIST OF FIGURES

Figure 1-1. Structures of Green Tea Catechins. [5] ................................................................ 2

Figure 1-2. Redox Activity of EGCG adapted from Yang et al [9] ........................................ 3

Figure 1-3. Biotransformation of the green tea catechins. [51] .............................................. 15

Figure 1-4. Tissue concentration of EGCG following oral and intragastric administration ... 18

Figure 1-5. Illustration of drug induced liver injury in obesity. [72] ...................................... 35

Figure 1-6. Mechanisms of lipid-induced cellular injury in ORFLD. [76] ............................. 36

Figure 2-1. Hepatotoxic effects of acute oral bolus EGCG in naive or dietary EGCG-

pretreated mice. ................................................................................................................ 49

Figure 2-2. Effect of dietary pretreatment with EGCG on the expression of antioxidant

response genes in mice following subsequent acute oral bolus dosing with EGCG. ....... 51

Figure 2-1. Effect of dietary pretreatment with EGCG on the oral bioavailability of a

single oral bolus EGCG in mice....................................................................................... 52

Figure 3-1. Survival rate and changes in body weight of male C57BL/6J mice following

treatment with EGCG. ...................................................................................................... 70

Figure 3-2. Plasma alanine aminotransferase levels and liver weight in male C57BL/6J

mice treated with oral EGCG. .......................................................................................... 71

Figure 3-1. Representative images of histopathological analysis of stained liver tissue

from male C57BL/6J mice. .............................................................................................. 73

Figure 3-4. Quantification of markers of liver tissue damage based on histopathological

analysis. ............................................................................................................................ 74

Figure 3-5. Representative images from transmission electron microscopy (TEM)

analysis of liver tissue of male C57BL/6J mice. .............................................................. 76

Figure 3-6. Quantification of mitochondrial parameters from TEM analysis. ........................ 77

Figure 3-7. Hepatic levels of reduced glutathione (GSH) were measured using HPLC and

normalized to tissue weight. ............................................................................................. 79

Figure 3-8. Hepatic levels of γ-H2AX determined by western blot analysis. ......................... 80

Figure 3-9. Hepatic levels of malondialdehyde (MDA). ........................................................ 81

Figure 3-10. Relationship between mitochondrial morphology and function. [112] .............. 86

Page 10: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

x

Figure 4-1. Effect of EGCG on hepatic gene expression, protein expression and activity

of superoxide dismutase 2 (Sod2) in male C57BL/6J mice. ............................................ 102

Figure 4-2. Effect of EGCG on hepatic gene expression, protein expression and activity

of catalase in male C57BL/6J mice. ................................................................................. 103

Figure 4-1. Effect of EGCG on hepatic gene expression, protein expression and activity

of glutathione peroxidase in male C57BL/6J mice. ......................................................... 104

Figure 4-4. Effect of EGCG on hepatic gene expression of glutathione-S-transferase zeta

1 and peroxiredoxin 3 in male C57BL/6J mice................................................................ 105

Figure 4-5. Changes in metallothionein expression in male C57BL/6J mice. ........................ 106

Figure 4-6. Effect of EGCG on hepatic gene and protein expression of Sirtuin 3 (Sirt3) in

male C57BL/6J mice. ....................................................................................................... 108

Figure 4-7. Effect of EGCG on the hepatic gene expression of forkhead box O3 and

nuclear factor (erythroid-derived 2)-like 2 (Nrf 2) in male C57BL/6J mice. ................... 109

Figure 4-8. Effect of EGCG on the hepatic gene expression of nuclear respiratory factor

1 (Nrf1) and peroxisome proliferator-activator receptor coactivator-1a (Pgc-1α) in

male C57BL/6J mice. ....................................................................................................... 110

Figure 4-9. Effect of EGCG on mitochondrial copy number in male C57BL/6J mice. .......... 111

Figure 4-10. Effect of EGCG on mitochondrial transcription factors A, (Tfam), B1

(Tfb1m) and B2 (Tfb2m) in male C57BL/6J mice............................................................ 112

Figure 4-11. Effect of EGCG on the hepatic gene expression of mitochondrial Complex I

and mitochondrial complex III in male C57BL/6J mice. ................................................. 113

Figure 4-12. Effect of EGCG on mitochondrial uncoupling protein 2 (Ucp2) in male

C57BL/6J mice. ............................................................................................................... 114

Figure 4-11. Scheme of mitochondrial antioxidant pathways and gene regulators. ............... 115

Page 11: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

xi

LIST OF TABLES

Table 1-1. Effects of green tea polyphenols on markers of diabetes1. [28] ............................. 11

Table 1-2. Case reports of hepatotoxicity with green tea-based dietary supplements. [44] ... 20

Table 2-1. Composition of Experimental Diets. ...................................................................... 43

Table 2-2. Primer sequences for qPCR analysis of hepatic antioxidant response in male

CF-1 mice. ........................................................................................................................ 44

Table 2-3. Comparative maximum concentration and exposure to EGCG in pretreated

and non-pretreated CF-1 mice. ......................................................................................... 52

Table 3-1. Composition of Experimental Diets. ...................................................................... 63

Table 4-1. Primer sequences for qPCR analysis of gene expression in liver tissue. ............... 97

Page 12: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

xii

ACKNOWLEDGEMENTS

I would like to express my sincere gratitude to my advisor Dr. Lambert. I appreciate the

guidance and support he gave me during my research tenure, particularly when I got stuck or was

pessimistic about my results. He always pointed out the value of my work and helped me see the

brighter side of what I was doing. Dr. Lambert maintained a positive attitude, was very

approachable, and made himself readily available to discuss any issues that arose no matter how

busy he was. I was fortunate to have an advisor with his background that was genuinely interested

in my project’s findings, which bolstered my own interest in this project and provided ideas and

direction for my future endeavors. I enjoyed his candor and views on research, science and life as

an academic. My research would not have been possible without the funding I received, so I

would also like to thank Dr. Lambert and the Department of Food Science for my assistantship

and tuition.

A special thank you to my other committee members, Dr. Elias for his input on the

chemistry aspects of my work and encouraging me to think about the chemical processes taking

place and their significance to my project; Dr. Perdew for his thought provoking questions that

ensured that I considered many perspectives and covered all my bases, and Dr. Vanamala for

encouraging me to always look at the bigger picture and broader implications of my work. I am

very grateful for their insight and approaches to problem solving; they helped me produce my

best work.

I am thankful to my lab mates Shannon Glisan, Weslie Khoo, Mingyao Sun, Kuier Zhao,

Ben Chrsifield, and Qiaoqioa Dai for their support, guidance, input, critiques, and discussions

throughout my project and during my defense and seminar. Special thanks to Ben Chrisfield for

his help with my experiments when my workload became overwhelming. Everyone played an

important role in my journey.

Page 13: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

xiii

I want to thank my friends in State College for being there for me, supporting me, helping

me grow, recognize my strengths, being my family and making this one of the most enriching

experiences of life. Last but not least my heart goes out to my family for their continued love and

support, especially my dad to whom I dedicate all of this work and because of whom all of this

was possible. Without him I would not be the woman I am today. His love and support was a

source of motivation, strength and sustenance.

Page 14: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

Literature Review and Research Objectives

Green Tea Processing and Chemistry

Tea is derived from the processing and brewing of leaves from the Camellia sinensis

plant. Drinking of the brew is a practice that has been around for centuries and valued for its

medicinal properties. Tea consumption, which originated in China and Southeast Asia, is very

high, and represents the second most popular beverage in the world [1][2]. Green tea is one of

three different types of tea produced based on processing, and it is the most popular and widely

studied for its health benefits. A typical cup of green tea (1.5 g in 150 mL water contains

approximately 450 – 675 mg of water extractable solids. Many of these solids are polyphenolic

compounds such as quercetin and myricitin [3]. Characteristic of green tea are catechins (Figure

1-1), which are polyphenols that make up 30 – 40 % of the water extractable solids [2]. (-)-

Epigallocatechin-3-gallate (EGCG) is the most abundant and potent catechin found in green tea,

making up approximately 50 % of catechins by weight [4].

Page 15: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

2

Figure 1-1. Structures of Green Tea Catechins. [5]

Processing of tea leaves produces three types of tea based on the method; green tea, black

tea and oolong tea. Green tea is processed in such a way as to minimize oxidation and retain the

major components that are found in the fresh tea leaf. Quickly after plucking, green tea leaves are

either steamed, pan fried, rolled or air dried at high temperature to inactivate polyphenol oxidase,

the oxidizing enzyme [5]. As a result, green has been shown to have the highest amount of

catechins (Graham 1992,Bhagwat et al. 2011). Black tea which is more commonly consumed

and manufactured compared to green tea is processed in order facilitate enzymatic oxidation of

the catechins in a process known as fermentation. Oxidation results in a decreased level of

catechins, and yields theaflavins, which are catechin dimers, and polymers called thearubigins

[5], [7]. Oolong tea processing allows for partial oxidation of tea leaves to occur and as a result it

contains varying levels of catechins. Oolong tea is considered an intermediate between green and

black tea and as such has components of both types of teas, but in different proportions. The

Page 16: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

3

partial oxidation process also yields components that are not found in either green or black tea,

for example, theasinensins (bisflavanols) [5].

Catechins have been widely studied for their antioxidant activities. The hydroxyl groups

on the A and B rings of the catechins are at the center of the antioxidant activity. This antioxidant

capacity is further increased by the presence of the gallate ester group (D-ring) as found in ECG

and EGCG. Based on this EGCG has been shown to exhibit the highest antioxidant capability of

the tea catechins [8].

The antioxidant capabilities of EGCG in biological systems have been extensively

explored. EGCG functions as a chain-breaking antioxidant that can trap peroxyl radicals and

interrupt detrimental reactions such as lipid peroxidation in vitro (Figure 1-2) (reviewed in [4]).

EGCG has also been shown to be effective in scavenging reactive oxygen, nitrogen and chlorine

species in vitro [4], [9]. EGCG antioxidant capacity extends to chelating transition metals, which

participate in the Fenton reaction and can initiate lipid peroxidation [4], [8], [10]. The relevance

of these direct antioxidant effects of EGCG in vivo remain unclear.

Figure 1-2. Redox Activity of EGCG adapted from Yang et al [11]

Page 17: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

4

Recent studies have demonstrated that EGCG has the propensity to exhibit pro-oxidant

activities as well, and this has been implicated in its anti-cancer capabilities (reviewed in [9]. In a

review by Lambert and Elias polyphenols such as EGCG were shown to undergo oxidation and

produce ROS in a process known as auto-oxidation, summarized in Figure 1-2.

Figure 1-2 shows EGCG reacting with molecular oxygen to produce the superoxide anion

and an EGCG semiquinone radical. The superoxide anion produced can further react with EGCG

to produce more EGCG semiquinone radicals and hydrogen peroxide (H2O2). The semiquinone

radical is very reactive and it can either react with EGCG to form reactive dimers or produce a

more stable quinone. Transition metals such as copper and iron are also able to initiate catechin

oxidation and can catalyze EGCG auto-oxidation. This contributes to the in vitro and in vivo pro-

oxidant effects of EGCG [4].

Putative Health Benefits of Green Tea and EGCG

Green tea and EGCG have been extensively studied for their numerous health benefits.

Numerous studies have demonstrated the effects of green tea catechins in treatment and

prevention of a host of diseases. They have been shown to have significant chemopreventative

effects, play a role in the treatment of cardiovascular disease, diabetes, arthritis, and

neurodegenerative disorders; and exhibit anti-inflammatory and anti-obesity effects [3], [12].

Many of those health benefits have been attributed to the antioxidant capabilities of EGCG.

Recent studies have shown that the pro-oxidant activities are very important as well, particularly

in the treatment of cancer [4], [9].

Page 18: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

5

Antioxidant Activity of EGCG

The antioxidant capabilities of EGCG have been explained by its ability to scavenge

ROS, chelate transition metal ions, inhibit redox-sensitive transcription factors, inhibit “pro-

oxidant” enzymes and induce phase II detoxification enzymes [12]. Several studies and reviews

have shown the antioxidant effects of EGCG in vitro and to a lesser extent in vivo [4], [9], [12].

For example, Nakagawa et al showed the direct scavenging of nitric oxide and

superoxide by green tea catechins in in vitro systems generating these reactive oxygen species

[13]. One study using 2,2′-azobis(2-amidinopropane) dihydrochloride (AAPH) to generate

radicals in renal epithelial cells showed that green tea catechins protected the cells from oxidative

damage with EGCG showing the highest antioxidant activity [14]. Kumamoto et al showed the

metal chelating abilities of EGCG when its antioxidant capacity was measured in the presence of

13 metals. EGCG was shown to have the highest antioxidant activity against copper (II) ions [15].

A study by Khan et al showed mice fed 0.2% (w/v) GTP for 30 days had significantly increased

activities of glutathione peroxidase, catalase, and quinone reductase in small bowel, liver, and

lungs, and glutathione S-transferase in small bowel and liver. Additionally there was considerable

enhancement of glutathione reductase activity in the liver [16].

Pro-oxidant Activity of EGCG

Depending on experimental conditions, green tea polyphenols undergo auto-oxidation

and generate ROS. For example, the addition of 50 µM EGCG to cell culture media caused the

production of hydrogen peroxide in time dependent manner to reach a maximum of 25 µM after

120 minutes [17]. The pro-oxidant activity of EGCG has been widely studied, particularly its

role in cancer prevention. Lambert and Elias, and Forester and Lambert have conducted reviews

Page 19: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

6

detailing the pro-oxidant activities of green tea polyphenols as it pertains to cancer prevention [4],

[9]. In vitro, EGCG was shown to induce apoptosis and inhibit cell proliferation in various cell

lines, as well as affect key cell signaling pathways that induce EGCG-mediated oxidative stress

[4]. Tao et al showed that preceding apoptosis, EGCG induced mitochondrial ROS and

mitochondrial dysfunction in oral cancer cells. SCC-25 cancer cells treated with 0 – 200 µM

EGCG for 0 – 6 hours resulted in formation of extracellular reactive oxygen species (ROS) and

increased the production of mitochondrial hydrogen peroxide in SCC-25 cells (0–6 h) before the

induction of apoptosis. Subsequent opening of the mitochondrial transition pore and a decrease in

mitochondrial membrane potential were observed. Additionally, EGCG down regulated

expression of genes related to antioxidant defense including superoxide dismutase 2/3 and

thioredoxin reductase 2 [18].

The pro-oxidant activity of EGCG in vivo has not been widely studied. One study by Li

et al showed that oral administration of EGCG to H1299 human lung cancer xenograft-bearing

nu/nu mice dose-dependently inhibited tumor growth and induced tumor cell apoptosis. EGCG

treatment resulted in tumor cell-specific increases in 8-hydroxy-2-deoxyguanosine (8-OHdG) and

phosphorylated histone 2A variant X (γ-H2AX) [19]. Lambert et al found that salivary H2O2

levels increased when healthy subjects either held tea solution (0.1–0.6%) in the oral cavity or

chewed green tea leaves. This study provides some support for the pro-oxidant activity of green

tea polyphenols [20]. These data suggest that pro-oxidant activity of polyphenols is impacted by

concentration and experimental conditions. Further studies are warranted to identify mechanisms

of pro-oxidant activity in vivo.

Page 20: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

7

Anti-Obesity Properties

The anti-obesity properties of green tea and EGCG have also been extensively studied

(reviewed in [21]. EGCG was shown to stimulate BAT thermogenesis, reduce adipocyte

differentiation and proliferation; lipogenesis, fat absorption, plasma levels of triglycerides, free

fatty acids, and increase beta-oxidation in in both cell culture and animal models of obesity [21].

High-fat fed C57BL/6J mice supplemented with 0.2-0.5% tea catechin in the diet showed

reduced body fat (epididymal, retroperitoneal, perirenal fat) as well as a significant reduction of

high-fat diet-induced body weight gain, visceral and liver fat accumulation [22], [23]. Green tea

reduced body fat accumulation in male Sprague–Dawley rats a high-fat diet for 14 days and

increased in brown adipose tissue thermogenesis through adrenoceptor activation [24]. High-fat

fed C57BL/6J mice treated with 0.32% dietary EGCG for 16 weeks saw a reduction in body

weight gain, percent body fat, and visceral tissue weight compared to high fat-fed controls [25].

Additionally, EGCG was shown to decrease liver weight, liver triglycerides, and plasma alanine

aminotransferase concentrations in high-fat fed mice. C57BL/6J mice on a high fat diet

supplemented with 1% w/w TEAVIGO (90% EGCG) for 5 months showed lower body weight

gain and reduced levels subcutaneous and epididymal adipose tissue [26]. In the same study,

obese Sprague-Dawley rats treated with 1% w/w TEAVIGO (90% EGCG) for 1 month showed a

reversal of obesity. In other study, the beneficial effects of EGCG on high-fat/Western-style diet-

induced obesity in C57BL/6J mice were examined. Treatment with 0.32% EGCG for 17 weeks

significantly lowered body weight gain weight of brown adipose tissue, total visceral adipose

tissue, levels of plasma cholesterol and plasma alanine aminotransferase compared to high-

fat/Western-style-fed mice [27].

EGCG has been shown to affect fat metabolizing enzymes and effect changes in

expression of genes related to fat metabolism and oxidation. Changes in these factors represent

Page 21: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

8

the mechanism by which EGCG causes the changes seen in body weight. A recent publication

using animal models showed that EGCG reduces body weight gain in high fat-fed mice by

inhibiting pancreatic lipase which is essential for the breakdown and absorption of fats in the

intestines [28]. Another study using high fat-fed C57bl/6J mice treated with 0.32% dietary EGCG

for 16 weeks observed that EGCG can increase the expression of genes related to lipid oxidation,

such as nuclear respiratory factor (nrf)1, medium chain acyl coA decarboxylase (mcad),

uncoupling protein (ucp)3, and peroxisome proliferator responsive element (ppar)α. The authors

also found that EGCG treated mice had reduced body weight gain and final body weight

compared to high fat-fed controls. EGCG-treatment decreased fasting blood glucose, plasma

insulin, and insulin resistance by 18.5%, 25.3%, and 33.9%, respectively [29]. New Zealand

black mice fed a high-fat diet supplemented with (0.5 and 1%) EGCG purified from green tea

(TEAVIGO) showed a dose-dependent decrease of body fat accumulation, reduction of gene

expression of stearoyl-CoA desaturase-1 (SCD1), malic enzyme (ME), and glucokinase GK and

an increase in UCP2. These data support a decrease in lipogenesis and an increase in fat oxidation

by EGCG treatment [30]. Changes in expression of genes related to fat metabolism and fatty acid

oxidation were measured in Male C57BL/6J mice fed a high fat diet for 8 weeks then

supplemented with 0, 0.2 or 0.5% EGCG for another 8 weeks. EGCG treatment reduced body

weight, and plasma triglyceride and liver lipid levels. Additionally, EGCG significantly reduced

mRNA levels of as peroxisome proliferator-activated receptor-γ (PPAR-γ), CCAAT enhancer-

binding protein-α (C/EBP-α ), regulatory element-binding protein-1c (SREBP-1c), adipocyte

fatty acid-binding protein (aP2), lipoprotein lipase (LPL) and fatty acid synthase (FAS), and

increased mRNA levels of carnitine palmitoyl transferase-1 (CPT-1) and uncoupling protein 2

(UCP2) [31].

In many cases of obesity the progression of hepatic steatosis or obesity related fatty liver

disease (ORFLD) is seen. A number of studies have reported the effect of green tea polyphenols

Page 22: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

9

on ORFLD in animal models. Sae-Tan et al showed that EGCG reduced liver injury marker,

alanine aminotransferase (ALT) in high fat fed mice[32]. Murase et al showed that 0.5% green

tea extract in the diet for 11 months significantly decreased liver lipid accumulation caused by a

high-fat diet in C57BL/ 6J mice [23]. Bose et al have also shown that in C57BL/6J mice fed high

fat diets supplement with 0.32% EGCG for 16 weeks, EGCG was able to ameliorate high fat-diet

induced ORFLD [25].

Anti-Inflammatory Properties

Inflammation is an important immune response to trauma and infection and aids in the

host’s recovery process. Chronic inflammation is cause for concern as it can result in DNA

damage, cell death, and cancer promotion [33]. Similar to studies demonstrating the anti-

carcinogenic effects of green tea polyphenols, laboratory studies have generally supported anti-

inflammatory effects of green tea polyphenols. EGCG has been shown to exert its inflammatory

effects by inhibiting inducible nitric oxide synthase (iNOS) gene expression and enzyme activity;

protecting against neutrophil-mediated inflammation induced by topical application of 12-O-

tetra- decanoylphorbol-13-acetate, in the ears of SENCAR mice; inhibiting infiltration of

leukocytes and markers of oxidative stress (H2O2 and NO) in the skin of UVB-treated C3H/HeN

mice following topical application; and preventing infiltration of macrophages and neutrophils

on UVB- irradiated human skin, also following topical application [34]–[37]. Kawai et al in a

recent study showed that EGCG was able to induce apoptosis in monocytes. Monocytes were

isolated from blood incubated without or with catechin; Epicatechin (EC), epigallocatechin

(EGC), epicatechin gallate (ECG), and EGCG. Apoptosis was evaluated by annexin V and

propidium iodide double-staining or terminal deoxynucleotidyl assay. Of the four catechins

evaluated, EGCG was the only one to induce apoptosis [38].

Page 23: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

10

In another study, EGCG was shown to have an inhibitory effect on T-cell–mediated

inflammation. CD8+ T cells incubated without or with catechin; epicatechin gallate (ECG), and

EGCG, were assessed for changes in the surface expression of integrin molecules and the direct

binding of catechin to CD11b molecule. Also, the effect of catechin on the ability of CD8+ T

cells to bind intracellular adhesion molecule 1 and to migrate in response to chemokines was

evaluated. Both catechins bound directly to CD11b expressed on CD8+ T cells, and caused a

decrease of CD11b expression, but EGCG was more prominent. This resulted in decreased ability

of CD8+ T cells to adhere intercellular adhesion molecule 1, and consequently decreased

migration in response to chemokines [38].

In addition to its anti-inflammatory effects on monocytes and T-cells, EGCG has been

shown to inhibit neutrophil elastase and apoptosis of neutrophils. Additionally EGCG inhibited

chemokine-induced neutrophil chemotaxis in vitro, blocked neutrophil-mediated angiogenesis in

vivo, and enhanced resolution in a pulmonary inflammation model, significantly reducing

consequent fibrosis [39].

Anti-Diabetes Properties

Insulin resistance, like obesity is a symptom of metabolic syndrome, which has been

shown to increase the risk of diseases such as cardiovascular disease, diabetes and cancer [40].

The International Diabetes Federation defines metabolic syndrome as having central adiposity

based on waist circumference (> 102 cm in men and > 88 cm in women) and two or more of the

following factors: elevated serum of triglycerides, dysglycemia, elevated blood pressure, and

reduced concentration of high-density lipoprotein (HDL) associated cholesterol [41]. A

comorbidity of metabolic syndrome is ORFLD. Data from clinical, experimental and

epidemiological studies indicate that ORFLD may be the hepatic manifestation of metabolic

Page 24: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

11

syndrome [42]. Insulin resistance is a key pathogenic factor in the development of type II diabetes

and has been shown to be associated with obesity [29].

Many laboratory, epidemiological and intervention studies have shown the role of green

tea polyphenols in the treatment and prevention of symptoms of metabolic syndrome (reviewed in

[43]). In the previous section we discussed the anti-obesity effects of green tea polyphenols. Here

we look at the role of green tea polyphenols in prevention and treatment of insulin resistance and

type II diabetes. Several studies have examined the effects of green tea polyphenols in type I and

type II diabetes in animal model. Sae-Tan et al conducted an extensive review of these findings,

and their data was summarized as seen in Table 1-1.

Table 1-1. Effects of green tea polyphenols on markers of diabetes1. [29]

Page 25: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

12

1db/db, leptin-receptor deficient; GTC, green tea catechins; GTE, green tea extract; SDR,

Sprague-Dawley rat; STZ, streptozotocin

These data suggest that green tea polyphenols are capable of improving symptoms of

insulin resistance and diabetes.

Dietary supplements

With all these seemingly beneficial effects of EGCG in cancer prevention and obesity

prevention shown mostly in animal models and cell lines, as well as its consumption as beverage

with no adverse effect, green tea and EGCG have been considered safe. Studies show that the oral

bioavailability of EGCG in humans is low compared to in vitro systems, and as such it has no

serious adverse effects when consumed in low doses, for example in green tea beverage [44]. The

relative safety of EGCG and its anti-obesity properties have in recent years lead to the translation

of green tea and EGCG into dietary supplements, for a variety of indications, particularly for their

potential to manage body weight [45].

US Food and Drug administration data shows that dietary supplements, including

vitamins, were consumed by 158 million Americans in the year 2000 compared with a previous

1997 survey that showed that alternative medical therapies, principally herbals, were used by 83

million people. The sales of dietary supplements in the United States doubled after passage of the

Dietary Supplement Health and Education Act (DSHEA) in 1994, to $17.1 billion in 2000, and

are anticipated to continue increasing by 10% per year [46]. According to the Nutrition Business

Journal, dietary supplement sales have reached $36 billion as of 2014 and projected to continue to

increase [47]. Regulation of dietary supplements is governed by the U.S. Food and Drug

Administration (FDA). According to DSHEA, a dietary supplement is a product taken by mouth

that contains a "dietary ingredient" intended to supplement the diet. The "dietary ingredients" in

Page 26: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

13

these products may include: vitamins, minerals, herbs or other botanicals, amino acids, and

substances such as enzymes, organ tissues, glandulars, and metabolites. Dietary supplements are

not as strictly regulated as drugs in that Under DSHEA, a firm is responsible for determining that

the dietary supplements it manufactures or distributes are safe and that any representations or

claims made about them are substantiated by adequate evidence to show that they are not false or

misleading. This means that dietary supplements do not need approval from FDA before they are

marketed [48]. This aspect can give rise to many inconsistencies in the products found on the

market, including variations in the concentrations of the different products and raises issues

regarding the safety of these products. By law, manufacturers may make three types of claims for

their dietary supplement products: health claims, structure/function claims, and nutrient content

claims. If a dietary supplement label includes a claim other than those mentioned, it must state in

a "disclaimer" that FDA has not evaluated this claim. The disclaimer must also state that this

product is not intended to "diagnose, treat, cure or prevent any disease," because only a drug can

legally make such a claim [48]. This often leads to manufacturers making claims the do not

necessarily have sufficient scientific evidence to support them.

Green tea based dietary supplements are especially popular for weight loss. For example,

Sales of green tea-based dietary supplements in the US totaled (USD) 5.6 million in 2005, an

increase of 94% from 2004 [49]. Green tea supplements can contain 0.4–8 mmol EGCG with a

recommend dosing of 1–2 capsules up to 3 times/d. giving rise to a total recommended dose that

may be up to 5.2 mmol/d. Green tea is commonly consumed as a beverage, and a typical cup of

green tea contains about 0.4 mmol EGCG [50]. Whereas no reported adverse effects associated

with normal green tea beverage consumption, green tea-based dietary supplements represent a

different dosage form and have the potential to deliver a much higher dose of catechins than

green tea beverages. For example Lambert et al showed that treatment of mice with a single oral

bolus dose of 1.1 mg/kg EGCG result in peak plasma concentrations of 2.0 mM, whereas the

Page 27: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

14

same total daily dose given via the diet resulted in plasma concentrations of 0.5 mM [50]. This

shows the stark difference in dosage forms and their potential implications in the safety of dietary

supplements, which I discuss further in the next section.

Impact of dosage form on bioavailability

Whereas historical exposure to EGCG has been in the form of a beverage, these dietary

supplements are generally in the form of capsules or pills. The safety of these alternative

formulations is generally based on the historical safety of green tea beverage. However, when

compared to green tea beverages, green tea based dietary supplements can deliver between 700

and 2100 mg of green tea polyphenols based on recommended dosing. These concentrations are

several times higher than what is found in a cup of tea, 240-320 mg of catechins, thus

supplements can deliver a higher dose of green tea catechins [4], [50]. Equally as important to

consider is the dosage form. Previous studies have shown that oral bolus dosing, such as capsules

results in greatly increased peak plasma concentrations of EGCG compared with dietary

administration of the same total daily dose [51]. This data suggests that the dosing form impacts

the bioavailability of the polyphenols. In this section I will discuss the metabolism and

bioavailability of EGCG

The metabolic pathways of green tea catechins have been shown to be methylation,

glucuronidation, sulfation and ring fission metabolism [8]. EGCG is methylated by catechol-O-

methyltransferase (COMT) to form 4″-O-methyl- (-)-EGCG and 4′,4″-O-dimethyl-(-)-EGCG and

was found to be time- and concentration-dependently sulfated in human, mouse and rat liver. But,

EGCG-4’’-O-glucuronide is the major metabolite formed in human, mouse and rat microsomes

[8], [52]. Following i.p. or i.g. administration of EGCG mouse urine have shown that methylated

EGCG (or glucuronidated or sulfated EGCG) can be further glucuronidated and/or sulfated (or

Page 28: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

15

methylated) to form related mixed EGCG metabolites [8]. In addition to these conjugation

reactions, the tea catechins undergo metabolism in the gut to form the ring fission products 5-

(3’,4’,5’-trihydroxyphenyl)-γ-valerolactone (M4) and 5-(3’,4’-dihydroxyphenyl)-γ-valerolactone

(M6). These products were present in human urine and plasma approximately 13 hours following

oral ingestion of 20 mg/kg decaffeinated green tea [8]. The extensive biotransformation of tea

catechins results in low systemic bioavailability. Figure 1-3 summarizes the biotransformation of

tea catechins.

Figure 1-3. Biotransformation of the green tea catechins. [53]

4’-MeEGC, 4’-O-methyl-(-)-epigallocatechin; 4’,4’’-di-O-methyl EGCG, 4’,4’’-di-O-methyl-(-)-

epigallocatechin-3-gallate; COMT, catechol-O-methyltransferase; EGC, (-)-epigallocatechin;

EGCG, (-)-epigallocatechin-3-gallate; SAH, S-adenosylhomocysteine; SAM, S-

adenosylmethionine; SULT, sulfotransferase; UGT, UDP-glucuronosyltransferase

Page 29: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

16

Studies have found that the oral bioavailability of EGCG and other green tea catechins in

humans is low [44]. For example, a study conducted by Nakagawa et al in which human

volunteers were given 3, 5 or 7 capsules of green tea corresponding to 225, 375 and 525 mg of

EGCG showed that 90 minutes after ingestion plasma levels of free EGCG concentrations

increased to 300, 1970 and 2020 ng/ml in the subjects who received 3, 5 and 7 capsules

respectively [54]. Chow et al showed that the plasma levels seen in the previous study by

Nakagawa et al were higher than levels obtain in their studies, although not high enough to exert

any potential toxicity. In their study healthy subjects were randomly assigned doses of 200, 400,

600, and 800 mg based on EGCG content. Analysis of blood and urine samples 24 hours after

oral administration showed plasma levels of EGCG observed were 73.7, 111.8, 169.1 and 438.5

ng/mL after 200, 400, 600, and 800 mg dose of EGCG respectively [44], [55]. Further research

by Chow et al investigated the effect of dosing conditions on green tea catechin availability, and

found that oral bioavailability of EGCG in humans increased under fasting conditions, when

compared to fed conditions. Fasting subjects given 400, 800 and 1200 mg of EGCG showed 450,

420, and 270 %, respectively increase in plasma concentrations of free catechins when compared

to fed subjects. [44]

Some studies show that mice and humans have a greater ability to glucuronidate EGCG

compared to rats and are thus may represent a better animal model for EGCG biotransformation

in humans compared to rats. Lambert et al examined EGCG bioavailability in mice following

intravenous and intragastric administration. Plasma and urine levels of EGCG were analyzed. It

was found that bioavailability of EGCG after intragastric administration was 12.4-25.6%. Plasma

levels of EGCG after intravenous and intragastric administration were 2.7 and 0.28 µmol/L

respectively. Results showed that EGCG was present in the plasma mainly in the conjugated form

after intragastric administration. This high level of conjugated EGCG and not free EGCG was

Page 30: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

17

attributed to the fact that intragastric administration results in the EGCG entering the small

intestines and liver prior to reaching the plasma. In these organs enzyme activity results in

conjugation of EGCG. In intravenous administration EGCG reached all the tissues at the same

time. As a result EGCG levels in various organs such as the lung, liver, small intestines, colon

and spleen was dependent on method of EGCG administration. It was concluded from this study

that mice and humans have similar EGCG bioavailability compared to rats and that EGCG

bioavailability is impacted most by glucuronidation of EGCG. This is consistent with research

that has shown that EGCG-4’’-O-glucuronide is the major formed in human, mouse and rat

microsomes and also takes into highlight species differences in EGCG biotransformation [56].

Based on these studies, and a review by Yashin et al it appears that EGCG bioavailability is

greatly impacted by the method of EGCG administration, and that bioavailability is increased in

this order, oral < intragastric < intravenous [57]. Figure 1-4 illustrates peak levels of EGCG

following route of administration.

Page 31: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

18

Figure 1-4. Tissue concentration of EGCG following oral and intragastric administration

The red peak represents gavage administration and the blue peak represents dietary

administration.

Figure 1-4 shows that following intragastric administration tissue concentrations are

significantly higher when compared to oral administration. This suggests that intragastric

administration has the potential to cause concentrations to increase pass a safe level thus

becoming unsafe or toxic. Whereas oral administration, especially dietary administration does not

increase concentrations nearly as high mainly because dietary administration occurs in

increments, allowing the concentration to fluctuate. In a study by Henning et al thirty healthy

subjects were randomly assigned to 3 different sequences of green tea, black tea, or a green tea

extract supplement in a 3 x 3 crossover design with a 1-wk washout period. They showed that

catechin absorption was enhanced when tea polyphenols were administered as a green tea

supplement in capsule form and led to a small but significant increase in plasma antioxidant

activity compared with when tea polyphenols were consumed as black tea or green tea [58].

Page 32: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

19

Understanding the mechanisms behind EGCG bioavailability will prove useful in understanding

the potential for EGCG toxicity.

Potential Toxic Effects of EGCG and Green Tea Extracts

A growing number of human cases have been reported citing adverse effects, mainly liver

toxicity associated with consumption of dietary supplements containing high levels of EGCG and

green tea extract. As previously stated, EGCG has the highest antioxidant potential among green

tea catechins because of the presence of the gallate ester, which increases the number of hydroxyl

groups in the compound; a moiety that is at the center of the antioxidant activity of catechins [8].

These functional groups in addition to increasing antioxidant capacity also increase the pro-

oxidant capabilities of EGCG and thus it’s overall redox activity. Due to this high redox activity

of EGCG, it has the greatest potential to cause toxicity when compared to other green tea

catechins. In addition to published cases of liver toxicity in humans, laboratory studies have

reported toxicity associated with administration of high doses EGCG in animal models. In the

following sections I will discuss data supporting EGCG mediate hepatotoxicity with a focus on

laboratory studies as well as human studies.

Human studies/Case Reports

Human studies for evaluating EGCG toxicity are very limited. Hepatotoxicity has not

been widely observed in controlled human intervention studies [59]. Many of the studies done do

not provide consistent findings. This indicates that there is a need to better understand the factors

which determine sensitivity to EGCG-induced hepatotoxicity, and the mechanism by which it

Page 33: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

20

occurs. With a large number of green tea based dietary supplements available on the market, and

more being produced, many of which contain high levels of EGCG (as high as 90%), the risk for

EGCG-mediated hepatotoxicity is a major cause for concern. Case reports are the common source

of data, but they do not provide conclusive evidence for hepatotoxicity from green tea. In a

review completed by Mazzanti et al 34 reported cases of liver toxicity believed to be associated

with green tea were reviewed Table 1-2[modified from [45]].

Table 1-2. Case reports of hepatotoxicity with green tea-based dietary supplements. [45]

Sex,

age

(years)

Herbal product

composition (Brand

name)

Time

(duration of

treatment in

weeks)

Evidence of

Toxicity

Dechallenge

/rechallenge

Reference

f, 81

Green tea dry aqueous

extract (90% EGCG)

4

ALT-

1996/2368

HEP

+/No

[45]

f, 72 12

ALT>700

HEP

Cholestatic

hepatitis +/No

m, 28 Green tea extract;

(Hydroxycut) 12

ALT-

1049/2272

HEP +/NR

Shim et al.

2008

Page 34: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

21

Sex,

age

(years)

Herbal product

composition (Brand

name)

Time

(duration of

treatment in

weeks)

Evidence of

Toxicity

Dechallenge

/rechallenge

Reference

f, 60

Green tea

8

62N/73N

MIX

NR +/NR

f, 56 12

8N/41N

MIX

NR +/NR

f, 64 5

77N/89N

HEP

NR +/NR

Bjornsson et al

2007

f, 35 11

55N/95N

HEP

NR +/NR

m, 40 20

33N/25N

MIX

Centrilobular

hepatocytes

drop out with

bridging

necrosis,

heavy

inflammatory

reaction +/NR

Federico et al

2007

Page 35: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

22

Sex,

age

(years)

Herbal product

composition (Brand

name)

Time

(duration of

treatment in

weeks)

Evidence of

Toxicity

Dechallenge

/rechallenge

Reference

f, 51 Green tea infusion (NR) 260

4-5N

COL

Mild

cholestasis +/+

f, 20

NR

∼ 60

COL

NR +/No

f, 44

Green tea extract,

Vitamin E, wheat germ

oil, soy oil, beeswax,

glycerol esters of fatty

acids (NR) 26

2393/3583

HEP

Hepatocellula

r necrosis,

mixed

inflammatory

infiltrates.

The patient

underwent

orthotopic

liver

transplant NR/No

Molinari et al

2006

f, 46 Green tea infusion (NR) 31

1188/1100

HEP

NR NR/NR

Javaid et al

2006

f, 26

Green tea (75%),Menta

piperita (25%) infusion

(Té verde Hacendado,

Mercadona, Valencia,

Spain) 18

1813/3314

HEP

Toxic liver

disease +/+

Martinez-

Sierra et al

2006

Page 36: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

23

Sex,

age

(years)

Herbal product

composition (Brand

name)

Time

(duration of

treatment in

weeks)

Evidence of

Toxicity

Dechallenge

/rechallenge

Reference

m, 45 Green tea infusion (NR) 18

1037/1613

HEP

NP +/+

Jimenez-Saenz

et al 2006

f, 37

Green tea extract

(Tegreen 97:

polyphenols 97%,

cathechins

64%),Magnolia

officinalis,Epimedium

koreanumand Lagerstro

emia speciosa extract,

calcium, chromium, L-

Theanine, β-sitosterol,

vanadium (USA) 18

1783/1788

HEP

Marked

interface

necrosis, mild

lobular

inflammation +/+

Bonkovsky et

al 2006

f

, 52

Green

tea, Citrus

aurantium, Citrus

paradisi, Cynara

scolymus,Petroselinum

sativumextracts (X-

elles) 1.2

2.5N/

6.5N

COL

Portal

inflammation,

mild

cholestasis,

mixed

centrolobular

inflammatory

infiltrates,

necrosis +/No

Mathi

eu et al 2005

Page 37: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

24

Sex,

age

(years)

Herbal

product composition

(Brand name)

Time

(duration of

treatment in

weeks)

Evide

nce of

Toxicity

Dec

hallenge/rec

hallenge

Reference

f

, 48

Green tea

extract (AR25: EGCG

25%), caffeine (19%)

(Exolise, Arkopharma,

Carros, France) 8

140N/

102N

HEP

Mixe

d portal

inflammatory

infiltrates,

necrotic and

steatosic

lobular

hepatocytes

NR/

No

Gloro et al

2005

f

, 35

See Gloro, 2005

[20] (Exolise,

Arkopharma, Carros,

France) 1.2

1191/

2885

HEP

NP +/No

Abu el Wafa et

al 2005

m

, 27

Green tea

extract (cathechins 70%,

EGCG 45%); Garcinia

cambogia, Gymnema

silvestre, Salixspp.and P

aullinia cupana extract,

calcium, chromium,

potassium,

glucomannan, α-lipoic

acid, L-carnitine,

caffeine, gelatin, silica,

cellulose (Hydroxycut,

Iovate Health Sciences

Research, Mississauga,

Canada) 5

1808/

3131

HEP

NR +/No

Stevens et al

2005

Page 38: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

25

Sex,

age

(years)

Herbal product

composition (Brand

name)

Time

(duration of

treatment in

weeks)

Evidence of

Toxicity

Dechallenge

/rechallenge

Reference

m, 30

0.6

59/45

COL

Cholestasis,

portal

inflammation +/No

f, 53

Green tea leaves,Ananas

sativuspowder, citric

acid, maltodextrine,

magnesium stearate,

silicium dioxide

(Fitofruits grasas

acumuladas) 2

927/1259

HEP

NP +/No

Porcel et al

2005

f, 25

Green tea leaves

micronized powder

(caffeine >2%)

(Camilina Arkocapsulas

Arkopharma, Carros,

France) 8

1943/2398

HEP

NP +/No

Garcia-Moran

et al 2004

f, 42

Green tea

leaves,Gynostemma

pentaphyllum, Aloe sp.

juice, Raphanus

sativus semen,Crataegu

s sp fruitc(Slim 10) 18

NR

Sub-massive

hepatocellular

necrosis. Post

mortem:

extensive

areas of dense

fibrotic tissue death

Lau et al 2004

f, 35

See Gloro, 2005 [20]

(Exolise, Arkopharma,

Carros, France) 5

1108/1558

MIX

NP +/No

Dueñas

Sadornil et al

2004

Page 39: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

26

Sex,

age

(years)

Herbal product

composition (Brand

name)

Time

(duration of

treatment in

weeks)

Evidence of

Toxicity

Dechallenge

/rechallenge

Reference

f, 56

Green tea hydroalcholic

extract,Cassia sp.

(Mincifit, Arkopharma,

Carros, France) 2

33N/54N

HEP

NP – Rech:

cholestasis,

mild necrosis,

steatosis of

20%

hepatocytes +/+e

Peyrin-

Biroulet et al

2004

f, 46

See Gloro, 2005 [20]

(Exolise, Arkopharma,

Carros, France) 13

61N/75N

HEP

NP +/+

Vial et al 2003

f, 35

See Gloro, 2005 [20]

(Exolise, Arkopharma,

Carros, France)

5

976/1558

MIX

NP +/No

f, 34 Several

NR

NR NR/No

f, 69 5

3N/4N

COL

NP +/No

Pedros et al

2003

f, 29 6.5

1023/1674

HEP

NR +/No

Page 40: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

27

Sex,

age

(years)

Herbal product

composition (Brand

name)

Time

(duration of

treatment in

weeks)

Evidence of

Toxicity

Dechallenge

/rechallenge

Reference

m, 44

As in Stevens, 2005

[22] + Ephedra

sinicaextract

(Hydroxycut Iovate

Health Sciences

Research, Mississauga,

Canada) 16

2046/3600

HEP

Mild

periportal

fibrosis,

periportal and

parenchymato

us

inflammatory

infiltrates +/No

Bajaj et al

2003

f, 52

Green tea,Gynostemma

pentaphyllum,Nelumbo

sp.,Chrysanthemum sp.,

Lycium

barbarum,Crataegus

monogyna,Citrus

aurantium,Cassia

mimosoides,Raphanus

sativus, beer yeast, Blc

Golden tang,

raifukushid(Be-petite) 9

1539/1920

MIX

Dramatic

portal

inflammation

with

characteristic

fibrosis +/No

Kanda et al

2003a

f, 31

Green tea

leaves,Gynostemma

pentaphyllum,

barbaloin, total saponin,

polyphenold(Onshidou-

genbi-kounou) 4

9310/8820

HEP

NR +/No

Kanda et al

2003b

f, 39

Green tea (7%), Oolong

tea, Cassia

angustifolia leaves,Mom

ordica

grosvenori, Malva

verticillata (Ooloon tea

fine tonic) 2

1.9N/3.9N

COL

NP +/No

Thiolet et al

2002

Page 41: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

28

Sex,

age

(years)

Herbal product

composition (Brand

name)

Time

(duration of

treatment in

weeks)

Evidence of

Toxicity

Dechallenge

/rechallenge

Reference

f, 50

See Gloro, 2005 [20]

(Exolise, Arkopharma,

Carros, France) 4

31N/40N

HEP

Marked

periportal

hepatocytes

necrosis,

lobular

inflammatory

infiltrates,

marked

centrolobular

cholestasis +/No

Seddik et al

2001

f, 19

Green tea powdered

leaves (Arkocapsulas,

Arkopharma, Carros,

France) 8

1130/1110

HEP

Necrosis of

hepatocytes,

mixed

inflammatory

infiltrates +/+

Gavilan et al

1999

f, Female; m, male; AST, alanine aminotransferase; NR, not reported; No, not done; NP, not

performed; HEP, hepatocellular; COL, cholestatic; MIX, mixed; -, not evaluable; ECGC, (-)-

epigallocatechin gallate a x N, x times the upper normal value; b Recovery time (weeks); cCapsules were adulterated with N-nitrosofenfluramine; dAdulteration with N-nitrofenfluramine is

hypothesized; eRechallenge was performed with Dynasvelte (Camellia sinensis + Coffea

arabica + chromium)

Elevated serum levels of aspartate aminotransferase (AST), alanine aminotransferase

(ALT), bilirubin and alkaline phosphatase were reported. In most of the cases reviewed in which

histological analysis was conducted, hepatic tissue necrosis was very common. In several of the

reported cases symptoms abated or ceased all together when consumption of the dietary

supplement was discontinued. Re-injury was reported when consumption of supplement was

resumed (reviewed in [45]). In another report, a 37-year-old woman suffered from abdominal

Page 42: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

29

pain, nausea and jaundice following consumption of a green tea extract weight loss supplement

for 4 months [60]. Her serum AST, serum alanine ALT, serum bilirubin and serum alkaline

phosphatase levels were all much higher than the respective reference ranges.

In a case report, a 44 year old woman was hospitalized for progressive illness, the onset

of abdominal pain and jaundice [1]. Blood tests revealed elevated serum bilirubin, serum AST,

serum ALT and gamma glutamyl transferase levels. Other bodily functions and vital signs were

normal. Investigation into the possible cause for her conditions revealed that the woman had been

engaged in a weight loss program that required the intake of green tea extract containing dietary

supplement with a dosage of 720 mg/day. Subsequent toxicology and hepatitis tests were all

negative. Treatment with intravenous infusion acetylcysteine resulted in worsening of some of

her conditions. A liver biopsy was performed and it revealed severe hepatocellular necrosis

among other finding. Continued deterioration of her medical conditions resulted in a liver

transplant. Further investigation prompted chemical analysis of the green tea extract supplements.

The results were not conclusive, but the medical personnel attributed the acute liver failure to the

green tea supplement the patient consumed as part of her weight loss regimen.

Federico et al presented the case of a 51 year old woman with a history of abnormal liver

function tests; increased serum aminotransferases (four- to fivefold), γ-glutamyl-transpeptidase

(∼ 200 U/L; normal range 0–50 U/L), and alkaline phosphatase (over 200 U/L; normal range 53–

128 U/L) spanning a period of 5 years. Clinical tests were not able to identify the cause of the

liver issues, but an investigation into her history revealed that she consumed green tea every day

for the past 5 years. Cessation of tea consumption for two months improved liver function test.

When she resumed consumption, liver function test reverted to being abnormal. Cessation again

returned liver function tests to normal [61].

A 45-year-old man diagnosed with jaundice was reported to consume six cups/day of a

marketed green tea infusion over the previous 4 months with no other medication. Laboratory

Page 43: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

30

tests showed elevated serum AST 1033 U/l, ALT 1613 U/l (<40), lactate dehydrogenase 602 U/l

(<425), alkaline phosphatase 310 U/l (<110), GGT 394 U/l (<30), total bilirubin 119 μg/dl (<17)

and direct bilirubin 102 μg/dl. Discontinuation of the product led to normal liver function tests.

Resumption of the product resulted in a significant increase of liver enzymes levels. Withdrawal

again normalized liver function tests [62].

These data though not conclusive indicate the potential of dietary supplements containing

high levels of EGCG to cause liver toxicity. One factor to consider is the bioavailability of

EGCG, which can vary from one individual to the next. In the next section I discuss EGCG

bioavailability and its impact on EGCG mediated liver toxicity.

Laboratory Studies

Animal models and cell lines have been used in laboratory studies to investigate the

potential toxicity of EGCG. Hepatotoxicity of high oral dose EGCG in mice was studied.

Lambert et al showed that high orally administered doses of EGCG in male CF-1 mice resulted in

hepatotoxicity [63] Mice were fasted and treated with either single dose of 50-2000 mg/kg EGCG

through oral administration or once-daily dose of 500 or 750 mg/kg EGCG through oral

administration for 2-7 days. Blood and liver samples were collected and analyzed by both

histological and biochemical methods, with plasma ALT used as the biochemical marker for

hepatotoxicity. Results showed that EGCG toxicity was time and dose-dependent. In mice given

once daily doses of 1500 mg/kg EGCG, and mice given two once-daily 750 mg/kg EGCG

through oral administration resulted in high mortality (85%) and toxic potency of EGCG

increased with this dose. Hepatic necrosis and apoptotic cells were observed in liver samples

from mice given two once-daily 750 mg/kg EGCG through oral administration. Plasma ALT

levels, showed an 8 and 108 fold increase in plasma ATL levels after 24 and 48 hours after

Page 44: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

31

administration of that dose. Increased oxidative stress was also observed after treatment with

1500 mg/kg EGCG through oral administration and two once-daily treatments with 750 mg/kg

EGCG through oral administration. A similar study in Beagle dogs observed a dose-dependent

toxicity of EGCG (in Polyphenon E) when administered at 200, 500, and 1000 mg/kg/day to

fasted dogs [64].

These results from Lambert et al suggest that at some threshold, or at a certain

concentration present in the cells, EGCG its exhibits pro-oxidant activities, which induces

oxidative stress and results in hepatotoxicity in the mouse. The toxicity levels in mice (500-1500

mg/kg) translates to (30-90 mg/kg) in humans, which is equivalent to 10.5-32 cups of green tea

[63]. Although these levels may not pose a significant risk for hepatotoxicity from tea

consumption alone, risk levels increase with the introduction of green tea dietary supplements.

Some supplements may contain sufficient levels of EGCG to reach the toxic threshold, or the

persons taking more than the recommended dosage could potentially reach the toxic threshold.

Galati et al studied the cytotoxicity of green tea catechins and phenolic acids [65].

Hepatocytes from male rats were obtained and treated with various stock solutions of the phenolic

compounds. Mitochondrial membrane potentials, hepatocyte GSH and GSSG, and reactive

oxygen species (ROS) formation were all measured. They found that the most cytotoxic tea

phenolic was EGCG. EGCG collapsed the hepatocyte mitochondrial membrane potential proving

to be the most effective mitochondrial toxin. They also found that EGCG was the most effective

at inducing ROS formation. The H2O2 generated caused oxidative damage to isolated and cellular

DNA in the rat hepatocytes. The same group studied hepatotoxicity in vivo in male mice and

found that EGCG increased plasma ALT levels in a dose-dependent manner, which indicates liver

injury, and EGCG treatment of 150 mg/kg i. p. resulted in mice death less than 24 hrs after

treatment. No actual references to hepatotoxicity in healthy cells were made in this study, making

it difficult to form conclusions on the actual hepatotoxicity of EGCG. The in vivo study reports

Page 45: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

32

that the toxic level of EGCG in male mice is 150 mg/kg when administered intraperitoneally.

This data though essential, is not relevant when assessing the potential toxicity of EGCG as it

pertains to green tea extract containing dietary supplements, which are administered orally.

In this study, Schmidt et al investigated the possible hepatotoxicity from hydro-alcoholic

green tea extracts as well as the components involved [66]. Hepatocytes isolated from adult rats

were treated with hydro-alcoholic green tea extracts of varying concentrations of catechins and

polyphenols and cytotoxicity was assessed. Results based on the two methods of analysis used to

determine cytotoxicity; EGCG was found to be mostly responsible compared to other green tea

components tested, for causing cytotoxicity in rat hepatocytes. This study is relevant to EGCG

toxicity as it pertains to green tea extract containing dietary supplements. Many supplements are

prepared using hydro-alcoholic extracts from green tea leaves. Exolise® is one such example, and

it is responsible for many of the reported cases of liver toxicity, resulting in its removal from

French and Spanish markets [45]. In their study Schmidt et al showed that the cytotoxicity of

EGCG occurred at levels ≥ 1000 µg/ml of green tea extract, which according to the authors is a

very high concentration, when compared to other hepatotoxins such as acetaminophen, which

cause toxicity at concentrations of 1-50 µM [66]. Based on their findings, the authors conclude

that hydro-alcoholic green tea extracts display low cytotoxicity in rat hepatocytes in vitro and

most of this toxicity is due to EGCG. The authors also concluded that due to EGCG’s low

bioavailability in humans, it does not appear to be the cause of liver toxicity associated with

supplements containing hydro-alcoholic green tea extracts. However, several of the studies that I

have looked at directly contradict this conclusion.

Kapetanovic et al examined the exposure and toxicity of green tea in fasted and non-

fasted dogs over the course of 13 week. Beagle dogs were treated with 0, 200, 500, and 1000

mg/kg/day of unformulated Polyphenon E, a highly purified and standardized green tea extract

containing 85-95% total catechins (56-72% EGCG) in gelatin capsules. The dogs were fasted

Page 46: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

33

prior to treatment with Polyphenon E. Blood and urine samples were collected from the fasted

dogs, and analyzed. The high dose of 1000 mg/kg/day had to be discontinued due to observed

levels of toxicity and subsequent death of one dog. During the 9 month study, the 1000

mg/kg/day administration resulted in the highest mortality (100 %), with the majority of deaths

occurring in the first 13 weeks. Overall there were high levels of mortality in the fasted dogs.

Some of the toxicities observed after analysis of samples include decreased red blood cell, and

increase in white blood cells, lesions in GI tract, lymph nodes, liver, kidney, lung, heart and

tonsils. Necrosis was also observed in several organs. The authors concluded that fasting

increased bioavailability of EGCG resulting in enhanced toxicity [64]

The underlying mechanism of EGCG-induced hepatotoxicity in vivo remains to be

studied. Lambert et al in a study showed that high doses of EGCG caused oxidative stress in liver

tissue resulting in hepatotoxicity [63]. The pro-oxidant activities of EGCG have been studied and

have been implicated in its chemopreventative effects. Studies by Galati et al and Tao et al show

that EGCG induces ROS in cancer cells resulting in cytotoxicity. Tao et al also showed that at the

doses used normal cells were unaffected by EGCG. The ROS including activities of EGCG in

vitro could have implications in vivo and could provide insight into high does EGCG induced

liver toxicity, which warrants further study.

Dietary and Lifestyle Factors Affecting Risk of Green Tea-Related Liver Toxicity

The benefits of EGCG particularly its ability to modulate body weight, has made it

popular as a dietary or weight loss supplements. In a study by Bailey et al, which examined the

use of supplements in adults and the reason for taking them, it was found that 45% of the adults

surveyed took supplements to improve and 33% to maintain overall health [67]. The study

Page 47: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

34

highlighted that many adult took multiple supplements for a variety of reason, and many were not

recommended by a physician or health care provider. As was discussed, many of these

supplements are capable of delivering a higher dose of green tea catechins compared to the

traditional beverage. When consumed with limited supervision, especially when not

recommended by a physician or health care provider the risk of improper use of supplements

increases. Many supplement takers do not consider the risks involved as some are taking multiple

supplements at once and many are of the belief that supplements are natural and are therefore

safe. As such, lifestyle factors that are potentially harmful are not considered when taking

supplements. Users have not evaluated potential health risks if supplements are taken with pre-

existing physiological conditions, or how taking multiple supplements or other aspects of the diet

may affect the safety of the supplements.

Previous studies have shown that chronic administration of EGCG to mice subsequently

decreased EGCG bioavailability [68]. Rats and mice treated with a 0.6% green tea polyphenol

preparation as the drinking fluid for 28 days showed an initial increase in plasma catechin levels

from Day 1 and peaked on day 14. The plasma levels of tea catechins the decreased, to Day 1

values on Day 28. The results suggest that consumption of tea by rodents could induce adaptive

responses affecting blood and tissue levels of tea catechins with time. Studies using other

compounds, for example alcohol, which is liver toxin, have shown that chronic consumption of

alcohol elicit an adaptive response over time, essentially allowing test subjects to develop a

tolerance [69]. With supplements capable of delivering high doses of EGCG and inducing

hepatotoxicity, the potential of chronic dietary administration of EGCG in modulating these

effects warrants further investigation.

Green tea supplements are marketed primarily for weight loss because of a pre-existing

obese condition. As I will discuss, obesity has the potential to increase drug induced liver injury

and the potential role it play in EGCG induced liver toxicity warrants further investigation.

Page 48: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

35

Numerous studies show that obesity is a major issue not only in the United States, but

worldwide and is considered a major public health problem. Obesity has been shown to be a

significant risk factor for many diseases such as type 2 diabetes, cardiovascular disease, obesity-

related fatty liver disease (ORFLD), and certain cancers [70]. Obesity is highly associated with

non-alcoholic fatty liver disease (NAFLD), which is the most common type of liver disease.

NAFLD can develop into non-alcoholic steatohepatitis (NASH), and in some cases hepatocellular

carcinoma (HCC), making obesity a risk factor for HCC [71], [72]. Studies have shown that

ORFLD effects changes in liver physiology and function. This impacts drug metabolism and it

has been shown to increase instances of drug induced liver injury (DILI) [73], [74]. Obesity has

been shown to increase blood flow to the liver and the activity of cytochrome P450 (CYPs)

metabolizing enzymes which can subsequently increase the generation of toxic metabolites. This

increases the risk for drug induced liver injury by inducing oxidative stress, mitochondrial

dysfunction and tissue necrosis. This is illustrated in Figure 1-5 [73]–[75].

Figure 1-5. Illustration of drug induced liver injury in obesity. [74]

Page 49: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

36

In addition to altering CYP activity, obesity has been shown to increase oxidative stress

by depleting glutathione (GSH) levels, which can inhibit the mitochondrial respiratory chain

(MRC) and lead to mitochondrial dysfunction. In addition depleted GSH levels in the

mitochondrial can prevent the removal of CYP-generated toxic metabolites, further impairing the

MRC [76].

Many studies support the role of obesity as a risk factor for DILI. In studies conducted

by Corcoran et al. obesity was shown to increase the risk of acetaminophen and furosemide

induced liver injury. Acetaminophen and furosemide, both of which generate toxic metabolites

when metabolized by CYPs, caused dose dependent increases in liver tissue necrosis when fed to

obese mice [77].

A review by Browning et al, (summarized in Figure 1-6) showed that in ORFLD there is

an increase in the release of free fatty acids (FFA) which can lead to insulin resistance, increase

oxidative stress and induce proinflammatory cytokines and eventually promote liver injury [78].

Figure 1-6. Mechanisms of lipid-induced cellular injury in ORFLD. [78]

Page 50: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

37

As stated earlier, EGCG has been well-regarded for it anti-obesity properties, so much so

that dietary supplements containing high levels of EGCG have been market particularly for

weight loss [21], [50]. With the growing body of evidence of high dose EGCG mediated liver

toxicity, and studies showing increased DILI in obesity, obesity is likely to increase susceptibility

to high dose EGCG mediated hepatotoxicity.

With a large number of green tea based dietary supplements available on the market, and

more being produced, many of which contain high levels of EGCG (as high as 90%), EGCG

toxicity and the risk of hepatotoxicity are a major cause for concern. Understanding the

mechanism by which EGCG mediates liver toxicity and the potential risk factors associated with

EGCG induced hepatotoxicity are significant, particularly from a public health perspective.

Consumers are exposed to doses of EGCG that are potentially toxic. With many of the green tea

supplements listing other ingredients such as acai berry, known for its antioxidant properties,

consumers are more inclined to purchase these supplements. Many are under the misconceptions

that more is better and naturally derived products are 100% safe. Also, with the growing trend of

eating and being healthy, especially in the face of an obesity epidemic many consumers believe

that taking dietary supplements will help them lead a healthy life. As such, studies investigating

the potential mechanism of EGCG mediated liver toxicity and the effect of dietary and

physiological conditions on toxicity are warranted to identify factors which increase susceptibility

to green tea supplement-associated hepatotoxicity.

Page 51: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

38

Research Hypothesis and Specific Aims

A growing body of evidence suggests the hepatotoxic potential of high doses of EGCG.

In vitro several mechanisms of EGCG cytotoxicity have been examined; however potential

mechanisms of in vivo toxicity remain understudied. Additionally factors that can potentiate

EGCG induced hepatotoxicity, such as pre-existing physiological conditions that attenuate liver

function have not been investigated. With the lack of data to support an association between

dietary supplements and liver toxicity, I hypothesized that dietary pretreatment with EGCG

markedly decreases EGCG bioavailability and can potentially mitigate the toxic potential of

EGCG, and that ORFLD increases susceptibility to high dose EGCG-mediated

hepatotoxicity. In addition to the effect of these factors on high dose EGCG hepatotoxicity, I

further hypothesized that the underlying mechanism by which EGCG mediates

hepatotoxicity is through induction of mitochondrial oxidative stress and increased

mitochondrial dysfunction.

To test this hypothesis the following specific aims were proposed:

1. To determine the effect of dietary pretreatment with EGCG on the bioavailability and

hepatotoxicity of subsequent oral bolus dosing with EGCG in mice.

2. To determine the effect of pre-existing obesity and ORFLD on EGCG-mediated

hepatotoxicity in mice.

3. To examine the effect of EGCG on hepatic markers of antioxidant response and

mitochondrial biogenesis/function in lean and obese mice.

Page 52: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

Dietary Administration of the Green Tea Polyphenol, (-)- Epigallocatechin-3-

gallate Reduces the Bioavailability and Hepatotoxicity of Subsequent High

Oral Bolus Doses of (-)-Epigallocatechin-3-gallate

Abstract

The green tea catechin, (-)-Epigallocatechin-3-gallate (EGCG), may have obesity

preventive effects. Recent case-studies, however, have reported an association between green tea-

based dietary supplement use and hepatotoxicity in human subjects. Additionally, laboratory

studies have demonstrated the hepatotoxicity of an acute high-dose oral bolus EGCG in mice and

dogs. Here, we examined the effect of pretreatment with dietary EGCG on the bioavailability and

hepatotoxicity of acute oral bolus EGCG in male CF-1 mice. EGCG (750 mg/kg, i.g., once daily

for 3 days) increased plasma levels of alanine aminotransferase (80-fold increase), decreased both

reduced (59% decrease) and total (33% decrease) hepatic glutathione, and increased hepatic

levels of γ-histone 2AX compared to control mice. Pretreatment with dietary EGCG (3.2 mg/g

diet) for 2 wk significantly blunted these markers of hepatotoxicity. Acute oral bolus dosing

with EGCG also decreased mRNA expression of glutathione reductase and glutathione

peroxidase (Gpx) 2 compared to control mice: dietary pretreatment prevented these decreases

and significantly increased expression of Gpx3, Gpx5, and Gpx7. We hypothesized that these

effects were related to modulation of EGCG bioavailability. We found that pre-treatment with

dietary EGCG significantly reduced the peak plasma (38% reduction) and hepatic (57%

reduction) EGCG concentrations following oral bolus dosing compared to mice that were not pre-

treated. In conclusion, it appears that EGCG can modulate its own bioavailability and that

Page 53: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

40

chronic exposure to dietary EGCG may reduce the toxic potential of acute high oral bolus doses

of EGCG. These data may help explain the variation in hepatotoxic response to green tea-

containing dietary supplements.

Page 54: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

41

Introduction

(-)–Epigallocatechin-3-gallate (EGCG), is the most abundant polyphenol in green tea

(Camellia sinensis), and has been widely-studied for its potential health benefits including

modulation of body weight gain [26], [28], [79]. One result of these reports of positive health

effects has been the proliferation of green tea-based dietary supplements for a variety of

indications. Whereas historical exposure to EGCG has been as a beverage, green tea-based

dietary supplements are generally capsules or pills. The result is the ability to deliver an

equivalent dose of green tea components in a much smaller volume. The safety of these

alternative formulations is generally based on the historical safety of green tea beverage,

however, there is increasing data to suggest that this assumption may not be valid [45].

Laboratory studies in mice and dogs have demonstrated the potential hepatotoxicity of

acute oral bolus dosing with EGCG or green tea extracts. We have previously reported that

EGCG induces dose-dependent hepatotoxicity in CF-1 mice [63]. Once daily dosing with EGCG

(750 mg/kg, i.g.) induced oxidative liver damage and hepatic necrosis. Similar results were

observed in fasted Beagle dogs treated with oral bolus doses of Polyphenon E, a defined tea

polyphenol mixture containing 60% EGCG [64]. Although the underlying mechanisms of

EGCG-induced hepatotoxicity remains understudied, it has been proposed that the acute oral

doses of EGCG, depending on dose, can result in oxidative stress leading to liver injury [63],

[80].

More than 34 case-reports have associated human hepatotoxicity with the use of green

tea-containing dietary supplements since 1999 [reviewed in [45]]. Although the causative role of

these supplement in the hepatotoxicity has not been clearly established, several case-reports have

indicated that cessation of supplement use led to resolution of symptoms and re-challenge led to

renewed liver injury [45]. It is interesting to note that hepatotoxicity has not been widely

Page 55: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

42

observed in controlled human intervention studies [55], [59]. These inconsistent findings indicate

that genetic or life-style factors may play a role in susceptibility to EGCG-mediated

hepatotoxicity.

Previous studies have shown that chronic, dietary administration of green tea can impact

the oral bioavailability of the tea polyphenols. Kim et al., have previously reported that long term

treatment of rats and mice with dietary green tea resulted in an initial increase in plasma EGCG

levels over the first 14 d of treatment [68]. Over the subsequent 14 d, plasma levels of EGCG

decreased with continued treatment and eventually reached baseline levels again. In addition,

EGCG has been shown to induce the expression of genes related to antioxidant response and

Phase II biotransformation in vivo [81]. These changes have been shown to alter the metabolic

profile of toxicants and carcinogens [82]. Given that EGCG, itself, is subject to Phase II

metabolism, it is plausible that chronic administration of EGCG or green tea may influence the

biotransformation and bioavailability of EGCG.

In the present study we determined the effect of dietary pretreatment EGCG on the

bioavailability and hepatotoxicity of subsequent oral bolus dosing with EGCG in mice. This

study was meant to mimic the potential hepatotoxic effects of high oral bolus dosing with EGCG

in chronic tea consumers to those in non-consumers, and should aid in identifying factors which

predispose subjects to green tea supplement-associated hepatotoxicity.

Materials and methods

Chemicals and reagents

EGCG (93% pure) was purchased from Taiyo Green Power (Wuxi, Jiangsu, China).

Diets (Table 2-1) were prepared by Research Diets, Inc. (New Brunswick, NJ).

Page 56: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

43

Table 2-1. Composition of Experimental Diets.

NC NP EP

Macronutrient Composition

Protein, % kcal

Carbohydrate, % kcal

Fat, % kcal

Ingredient, g/kg

Casein, 30 Mesh

DL-Methionine

Corn Starch

Sucrose

Cellulose, BW 200

Corn Oil

aMineral Mix S10001

bVitamin Mix V10001

Choline Bitartrate

EGCG

20.8

67.7

11.5

200

3

150

500

50

50

35

10

2

0

20.8

67.7

11.5

200

3

150

500

50

50

35

10

2

0

20.8

67.7

11.5

200

3

150

500

50

50

35

10

2

3.2

aMineral mix adds the following components (per g mineral mix): sodium chloride, 259

mg; magnesium oxide, 41.9 mg; magnesium sulfate, 257.6 mg; chromium K sulfate, 1.925 mg;

cupric carbonate, 1.05 mg; sodium fluoride, 0.2 mg; potassium iodate, 0.035 mg; ferric citrate, 21

mg; manganous carbonate, 12.25 mg; ammonium molybdate, 0.3 mg; sodium selenite, 0.035 mg;

zinc carbonate, 5.6 mg. bVitamin mix adds the following components (per g vitamin mix):

vitamin A palmitate, 400 IU; vitamin D3, 100 IU; vitamin E acetate, 5 IU; menadione sodium

bisulfite, 0.05 mg; biotin, 0.02 mg; cyanocobalamin, 1 µg; folic acid, 0.2 mg; nicotinic acid 3 mg;

calcium pantothenate, 1.6 mg; pyridoxine-HCl, 0.7 mg; riboflavin, 0.6 mg; thiamin HCl, 0.6 mg

Page 57: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

44

Primers for quantitative reverse transcriptase (q)PCR (Table 2-2) were synthesized by the

Genomics Core Facility at The Pennsylvania State University (University Park, PA).

Phosphorylated histone 2AX (γH2AX) and glyceraldehyde-3-phosphate dehydrogenase (Gapdh)

were purchased from Cell Signaling Technolgies (Danvers, MA). All other chemicals were of

the highest grade commercially-available.

Table 2-2. Primer sequences for qPCR analysis of hepatic antioxidant response in male CF-1

mice.

Gene Forward Primer Reverse Primer

Gsr GACACCTCTTCCTTCGACTACC CACATCCAACATTCACGCAAG

Gstz1 CGCTGAAAATTGATGGAATCACC GATGCCACTAGCGATGAGGT

Gpx1 CCACCGTGTATGCCTTCTCC AGAGAGACGCGACATTCTCAAT

Gpx2 GCCTCAAGTATGTCCGACCTG GGAGAACGGGTCATCATAAGGG

Gpx3 CCTTTTAAGCAGTATGCAGGCA CAAGCCAAATGGCCCAAGTT

Gpx4 GCCTGGATAAGTACAGGGGTT CATGCAGATCGACTAGCTGAG

Gpx5 TCTAGCCAGCTATGTGCAGAC TCCTTCCCATTAAGAGACAGAGC

Gpx7 TCCGAGCAGGACTTCTACGAC TCTCCCTGTTGGTGTCTGGTT

Sod2 CAGACCTGCCTTACGACTATGG CTCGGTGGCGTTGAGATTGTT

Animal studies

All animal studies were approved by the Institutional Animal Care and Use Committee of

the Pennsylvania State University (University Park, PA, IACUC 37115). Mice were maintained

on a 12 h light-dark cycle at 21o C and 38% relative humidity. Mice were acclimatized for 1 wk

Page 58: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

45

prior to the start of experiments and had ad libitum access to food and water. The dietary dose of

EGCG (3.2 mg/g diet) is equivalent to a total daily dose of 500 mg/kg, body weight. The dietary

EGCG dose and the oral bolus EGCG dose (750 mg/kg, body weight) are equivalent to 10 and 16

cups of green tea prepared using a standard method (2.5 g tea leaves in 250 mL) [56].

Hepatotoxicity Studies

Male CF-1 mice (6 – 8 wks old, Charles River Laboratory, Wilmington, MA) were

randomized into three treatment groups; EGCG pretreatment (EP), no pretreatment (NP), and

negative control (NC). EP mice were fed AIN76A diet supplemented with EGCG (3.2 mg/g) for

14 d, whereas NP and NC mice received AIN76A diet. Following the pretreatment period, mice

were fasted for 7 h (0700 – 1400 h) and then given a single dose of EGCG (750 mg/kg, i.g.) once

daily for 3 d. On d 3, 1 h post-gavage, mice were anesthetized and blood was collected by cardiac

puncture. Plasma was prepared by centrifugation at 3200 g for 15 min and frozen at -80oC. The

livers were harvested, washed with ice-cold PBS and frozen at -80o for later analysis.

Bioavailability Studies

Male CF-1 mice (n = 24 per group) were randomized into EP, NP and NC pre-treatment

groups. After a pretreatment period of 14 days, NP and EP mice were fasted for 7 h and then

given a single dose of EGCG (750 mg/kg, i.g.). At 0.5, 1.5, 4 and 6 h post-gavage, mice were

anesthetized and blood was collected via cardiac puncture. Plasma was prepared as above,

combined with 0.1 vol of ascorbate preservative (20% ascorbic acid: 0.1% EDTA) and frozen at -

800C. Livers were harvested and frozen as above.

Page 59: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

46

Determination of hepatotoxicity and oxidative stress

Plasma alanine aminotransferase (ALT) levels were determined spectrophotometrically

(λmax = 340 nm) using a commercially-available assay (CATACHEM Bridgeport, CT) according

the manufacturer’s instructions. Expression of γ-H2AX was determined by western blot. Protein

was extracted from liver samples using Tissue Protein Extraction Reagent (TPER) containing 1%

(v/v) phosphatase and protease inhibitor cocktails (Thermo-Scientific, Rockford, IL). Protein

samples (30 μg protein) were resolved by SDS-polyacrylamide gel electrophoresis (PAGE) and

transferred to a nitrocellulose membrane. Membranes were probed with γH2AX primary antibody

and a fluorescent-conjugated secondary antibody (Li-Cor Co., Lincoln, NE). Bands were

visualized using a Licor Odyssey Imaging System (LI-COR Corporate). GAPDH was used as a

protein loading control. Total and reduced glutathione levels were determined using the GSH-

GloTM Glutathione assay kit (Promega, Madison, WI, USA) according to the manufacturer’s

instructions.

Gene expression studies

The mRNA expression of genes related to antioxidant response (Table 2) was examined

by qPCR. RNA was isolated from liver tissue using Tri-reagent (Sigma Chemical Co. St. Louis,

MO) according to the manufacturer’s instructions. Isolated RNA was quantified using the

NanoDrop ND-1000 spectrophotometer. cDNA was reverse transcribed and amplified using

SYBR Green PCR Master Mix and an ABI Prism 7000 sequence detection system (Applied

Biosystems, Foster City, CA) . Standard curves were made by using serial dilutions from pooled

cDNA samples. mRNA levels were normalized to the house-keeping gene glyceraldehyde-3-

phosphate dehydrogenase (Gapdh).

Page 60: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

47

HPLC analysis of EGCG

EGCG levels in plasma and liver samples were determined as described previously [56].

In brief, plasma samples were combined with 0.4 M sodium phosphate monobasic (pH 6.8) and

20% ascorbic acid and then extracted with dichloromethane and ethyl acetate. The ethyl acetate

fraction was dried under vacuum and resuspended in 10% aqueous acetonitrile for HPLC

analysis. The resultant samples represented unconjugated EGCG. To determine the total EGCG,

samples were treated with β-glucuronidase (250 U) and sulfatase (1 U) prior to extraction. Liver

samples were homogenized in a 1:1 mixture of hydrosulfite buffer (0.4 M sodium phosphate

monobasic containing 0.3 M sodium hydrosulfite and 0.1% disodium EDTA) and methanol:ethyl

acetate (2:1, v:v). The supernatant was collected after centrifugation for 4 min at 16 000 g. After

removing the methanol and ethyl acetate under vacuum, samples were treated in a manner

analogous to plasma. All samples were analyzed using a Shimadzu HPLC system equipped with

two LC-20AD pumps (Shimadzu Co, Columbia, MD), an ESA 5600A Coularray detector

(Chelmsford, MA), and a Supelcosil LC18 column (4.6 x 150 mm, 5 µm particle size, Supelco,

Bellefonte, PA).

Statistical Analysis

All experiments were repeated twice with at least two replicates for each treatment and

time point. Maximum plasma concentrations (Cmax) and exposure (AUC06h) were determined

using GraphPad Prism 5.0 (San Diego, CA). One-way ANOVA with Dunnett’s post-test was

used to compare differences in plasma ALT, hepatic glutathione, and gene expression data.

Pharmacokinetic parameters were compared using the Student’s t test. Statistical analysis was

Page 61: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

48

performed using GraphPad Prism 5.0. Data are expressed as mean ± SEM and significance was

reached at P<0.05.

Page 62: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

49

Results

Effect of EGCG pretreatment on EGCG-induced hepatotoxicity and oxidative stress

Figure 2-1. Hepatotoxic effects of acute oral bolus EGCG in naive or dietary EGCG-pretreated

mice.

(A) Plasma ALT levels were determined following administration of 3 daily doses of 750 mg/kg,

i.g. EGCG to male CF-1 mice pretreated with dietary EGCG [3.2 mg/g diet] for 14 days [EP] or

not [NP] using spectrophotometric methods. (B) Hepatic levels of reduced glutathione [GSH] and

(C) total glutathione in EP and NP mice following 3 daily doses of 750 mg/kg, i.g. EGCG using a

luminescence-based assay. N = 8–16.(D) Hepatic levels of γH2AX were determined by western

blot analysis in EP and NP mice following 3 daily doses of 750 mg/kg, i.g. EGCG. Expression

was determined by densitometry and normalized to GAPDH. The blot shows representative

expression patterns. The histogram shows quantification of all samples analyzed (N = 10 per

treatment group). Error bars represent SEM. Data were analyzed by one-way ANOVA with

Dunnett’s post-test (* = P < 0.05; ** = P < 0.01]. All biomarkers were compared to

vehicletreatedcontrol mice [NC].

Page 63: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

50

Treatment of naïve male CF-1 mice (NP) with oral bolus EGCG (750 mg/kg, i.g., once

daily) for 3 days resulted in an 80-fold increase in plasma ALT levels compared to vehicle-treated

control mice (NC) (Figure 2-1A). By contrast, pretreatment with EGCG (3.2 mg/g diet

[equivalent to 500 mg/kg, body weight total daily dose], EP) for 2 weeks reduced this elevation in

plasma ALT by 75%. Treatment with oral bolus EGCG (750 mg/kg, i.g. once daily for 3 d)

decreased both reduced (GSH) and total glutathione in NP mice by 57% and 33%, respectively,

compared to vehicle-treated controls (Figure 2-1B and 2-1C). These reductions were ameliorated

in EP mice (Figure 2-1B and 2-1C). Western blot analysis showed that the hepatic levels of the

oxidative stress marker, phosphorylated histone 2AX (γ-H2AX), were increased in NP mice

compared to NC mice (Figure 2-1D). By contrast, hepatic γ-H2AX expression was mitigated in

EP mice.

Hepatic expression of antioxidant responsive genes

A panel of genes related to antioxidant response was examined in the hepatic tissue of

mice treated with daily oral bolus EGCG (Figure 2-2). The expression of glutathione reductase

(Gsr) and glutathione peroxidase (Gpx)2 were significantly suppressed in NP compared to NC

mice (Figure 2-2). These decreases in expression were ameliorated in EP mice. In addition,

hepatic expression of Gpx3, Gpx5, and Gpx7 was increased in EP mice compared to NP mice

(Figure2-2).

Page 64: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

51

Figure 2-2. Effect of dietary pretreatment with EGCG on the expression of antioxidant response

genes in mice following subsequent acute oral bolus dosing with EGCG.

Hepatic gene expression of the genes of interest were determined following administration of 3

daily doses of 750 mg/kg, i.g. EGCG to male CF-1 mice pretreated with 3.2 mg/g dietary EGCG

for 14 days [EP] or not [NP]. Levels were determined 0.5 and 1.5 hours after the last EGCG

treatment and normalized to Gapdh expression. Each bar represents the mean of N = 4–8. Error

bars represent SEM. Data were analyzed by one-way ANOVA with Dunnett’s post-test (*P = <

0.05, ** = P < 0.01).

Page 65: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

52

Effect of EGCG pretreatment on EGCG bioavailability

Figure 2-1. Effect of dietary pretreatment with EGCG on the oral bioavailability of a single oral

bolus EGCG in mice.

(A) Plasma and (B) liver levels of unconjugated EGCG were determined following a single oral

bolus dose of 750 mg/kg, i.g. EGCG to male CF-1 mice pretreated with 3.2 mg/g dietary EGCG

for 14 days [EP] or not [NP]. EGCG levels were determined by HPLC with electrochemical

detection. Data represent the mean of N = 5–6 mice. Error bars represent SEM. Data were

analyzed by one-way ANOVA with Dunnett’s post-test (*P = < 0.05, ** = P < 0.01).

Table 2-3. Comparative maximum concentration and exposure to EGCG in pretreated and non-

pretreated CF-1 mice.a

Parameter Units EP NP

Plasma

Cmax

µM 14.5 7.4 33.7 7.6

AUC06h

µM · h 36.2 8.4* 83.3 12.5

Liver

Cmax

µg/g 0.34 0.11 0.79 0.35

AUC06h

µg/g · h 0.9 0.2† 3.1 0.7

a Data represent the mean ± SEM.

* Indicates P < 0.05.

† Indicates P = 0.0637.

Page 66: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

53

Plasma and liver concentrations of unconjugated EGCG were determined by HPLC in

both EP and NP mice (Figure 2-3). We found that following a single oral bolus dose of 750

mg/kg, i.g. EGCG, the maximum plasma concentration (Cmax) of and exposure (AUC06h) to

unconjugated EGCG was 1.3-fold higher in NP mice compared to EP mice (Table 2-3). Similar

to the plasma, EGCG Cmax and AUC06h were higher in the livers of NP mice than EP mice

(Table 2-3).

Page 67: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

54

Discussion

In the present study we investigated the effect of dietary pretreatment with EGCG on

hepatotoxicity induced by high oral bolus dose EGCG mediated in male CF-1 mice. These

experiments were meant to evaluate dietary conditions which mitigate the hepatotoxic effects of

high oral bolus doses of EGCG. They were also meant to model a comparison of the potential

hepatotoxic effects of a high oral bolus dose of EGCG, as might be expected through use of green

tea-based dietary supplements, in regular tea consumers (EP mice in this study) to naïve subjects

(NP mice in this study). Such circumstances of exposure to green tea-based dietary supplements

are likely to occur in free-living human subjects.

The doses of EGCG selected for the present study, 500 mg/kg total daily dose (3.2 mg/g

in the diet) or 750 mg/kg, i.g. bolus dose are equivalent to a human dose of 20 or 30 mg/kg body

weight, respectively, based on allometric scaling [63]. These doses are somewhat higher than the

typical recommended dose of EGCG derived from commercially-available supplements (2 – 12

mg/kg/d), but are well within the range achievable in humans subjects who exceed the

recommended dose either inadvertently or due to a belief that higher doses of EGCG will provide

additional benefit.

Previous studies by our laboratory and others have reported that high oral bolus doses of

EGCG induced liver toxicity in the livers of mice and Beagle dogs, whereas dietary dosing with

up to 1000 mg/kg body weight/d was not toxic (Lambert et al. 2010; Isbrucker et al. 2006).

These differences likely result from the fact that the Cmax of EGCG achieved following oral bolus

dosing with EGCG is significantly higher than that achieved by the same dose give in the diet

(Lambert et al. 2006; Klaus et al. 2005).

Here, we observed that oral bolus dosing with EGCG (750 mg/kg once daily with 3 d)

elevated plasma ALT levels in non-pretreated (NP) mice compared to vehicle-treated controls

Page 68: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

55

(NC). These effects are consistent with our previous studies[63]. By contrast, dietary pretreatment

with EGCG for 2 weeks (EP) reduced this elevation in plasma ALT by 75%, indicating a

reduction in hepatotoxicity induced by oral bolus EGCG.

EGCG has been shown to exhibit pro-oxidant activity both in vitro and in vivo (reviewed

in ([9]), and previous studies have shown that EGCG-mediated liver toxicity was associated with

increased hepatic oxidative stress (Lambert et al. 2010; Galati et al. 2006). Here, we found that

both reduced (57 % decrease) and total glutathione (33 % decrease) was significantly diminished

in NP mice compared to vehicle-treated NC mice, which is consistent with previous studies

demonstrating EGCG-mediated increases in hepatic oxidative stress. By contrast, the reductions

in both GSH and total glutathione were blunted in EP mice.

Increased phosphorylation of histone 2AX (γH2AX) is a marker of oxidative stress and

double strand DNA breaks. Previously we found that EGCG treatment could increase hepatic

levels of γH2AX [63]. Consistent with this previous work, we found that NP mice exhibited

increased hepatic levels of γ-H2AX compared to NC mice. By contrast, pretreatment with dietary

EGCG mitigated this increase in hepatic γ-H2AX. Taken together, these results suggest that

pretreatment with dietary EGCG can ameliorate oxidative stress induced by high oral bolus doses

of EGCG.

In addition to blunting the expression of biomarkers of oxidative stress, dietary

pretreatment also preserved or enhanced the hepatic expression of several genes related to

antioxidant response in mice following treatment with a single high dose oral bolus of EGCG.

Following oral bolus dosing with EGCG, hepatic expression of glutathione reductase (Gsr),

glutathione-S-transferase zeta 1 (Gstz1), and glutathione peroxidase (Gpx)2, Gpx3, Gpx5, and

Gpx7 was higher in mice that had been pretreated with dietary EGCG compared to those that did

not receive EGCG pretreatment. Increased expression of these genes would be expected to

translate into reduced hepatic oxidative stress and are similar to the results of previous studies.

Page 69: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

56

Previous studies have shown that oral dosing with EGCG can modulate hepatic

antioxidant gene expression and function. For example, oral bolus administration of non-toxic

doses of EGCG (200 mg/kg once daily) has been shown to enhance the expression of a number of

antioxidant-related genes in mice in a Nuclear factor (erythroid-derived 2)-like 2-dependent

manner [81]. Another study found that dietary green tea extract could enhance glutathione-

mediated detoxification of aflatoxin in piglets [86]. Taken together, the present results and these

previous studies provide a putative mechanism by which dietary EGCG can mitigate the pro-

oxidant effects of toxic oral bolus doses of EGCG.

Previous studies in mice and rats have indicated that longer term treatment with dietary

green tea can reduce the overall plasma and tissue bioavailability of EGCG and the other green

tea polyphenols [68]. These authors reported that in A/J mice given 0.6% green tea solids as the

sole source drinking fluid, plasma and liver levels of EGCG increased from day 0 to day 4 and

then declined over the subsequent 10 days of treatment. Consistent with these results, we found

that following a single oral bolus dose of 750 mg/kg, i.g. EGCG, the plasma Cmax and

AUC0→6h of unconjugated EGCG was 1.3-fold higher in NP mice compared to EP mice.

Similarly, the hepatic EGCG Cmax and AUC0→6h were higher in NP mice than EP mice. These

results indicate that EGCG can impact its own bioavailability and that chronic exposure can result

in lower plasma and tissue levels of unconjugated EGCG.

Unconjugated EGCG has been reported to have greater biological activity than

glucuronidated and methylated conjugates of EGCG [52], [87], therefore this observed decrease

in bioavailability, coupled with increased expression of hepatic antioxidant genes, provides a

mechanism by which dietary pretreatment with EGCG can reduce acute oral bolus-related

hepatotoxicity. Further studies are needed to determine the molecular mechanism by which

EGCG can modulate its own bioavailability; however, it is reasonable to speculate that EGCG

may induce Phase II metabolic enzymes as well as active efflux transporters. We and others have

Page 70: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

57

previously shown that the bioavailability of EGCG is modulated by uridine diphosphate

glucuronyltransferases, catechol-Omethyltransferase, and the multidrug resistance-associated

proteins in the liver or small intestine [17], [52], [56], [88], [89].

Previous studies have shown that these systems are inducible by other compounds [90].

Further studies are needed to determine if chronic oral EGCG can induce such changes and the

extent to which such induction may influence the oral bioavailability of acute oral bolus doses of

EGCG. Of note, there have been a limited number of studies that have examined the impact of

repeating dosing on the bioavailability of EGCG in human subjects. Chow et al. have reported

that daily dosing with 800mg EGCG for 4 weeks increased both the plasma Cmax and AUC [55].

No such increase was observed at a lower dose (400 mg per day). The underlying mechanism for

these results remains unclear, and no further studies have reported similar findings. It is possible

that the differences between this study in humans and our present study in mice are related to

species differences in the response of inducible Phase II and active transport systems.

Alternatively, the differences could result from the dosage form for the chronically administered

EGCG. In our study, it was incorporated into the diet, and mice are to be exposed continuously to

lower concentrations throughout the feeding period. In the human study, EGCG was administered

in pill form once or twice daily. The result would be higher peak plasma concentrations and more

frequent periods of very low levels. To date there have been no studies to determine the impact of

such differences on bioavailability of EGCG over long periods of time.

Page 71: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

Effect of Obesity on High Dose Oral EGCG-mediated Liver toxicity

Abstract

Fatty liver disease is a co-morbidity of obesity which is characterized by increased

hepatic oxidative stress and increased sensitivity to drug induced liver injury (DILI). Green tea-

based dietary supplements containing high levels of the polyphenol (-)-epigallocatechin-3-gallate

(EGCG) have become popular for weight loss. Case-studies have reported hepatotoxicity

associated with consumption of some of these supplements and laboratory studies have shown the

potential hepatotoxicity of EGCG in animals. With the growing body of evidence demonstrating

the hepatotoxic potential of high oral doses of EGCG, and studies showing increased sensitivity

to DILI in obese subjects, it is reasonable to hypothesize that obesity may increase susceptibility

to EGCG-mediated hepatotoxicity. We tested this hypothesis in high fat-fed obese C57BL/6J

mice. Obese and age-matched lean controls were treated with 0, 250, 500, or 750 mg/kg EGCG

by oral gavage, once daily for 2 days. Lean and obese mice treated with 750 and 500 mg/kg

EGCG suffered many casualties over the course of the treatment period. Obese mice treated with

750 mg/kg and 500 mg/kg EGCG had survival rates of 25% and 68% respectively, while lean

mice had 91% and 86% survival rates. At 500 mg/kg, EGCG treatment increased plasma alanine

aminotransferase (ALT) and hepatic malondialdehyde (MDA) levels to a greater extent in obese

mice compared to lean, age-matched controls. By contrast, plasma ALT and hepatic MDA levels

were greater in lean mice treated with 750 mg/kg EGCG compared to obese mice. EGCG

treatment reduced hepatic levels of reduced glutathione more significantly in obese mice

compared to lean mice. Obese mice also had significantly higher liver weights. EGCG treatment

Page 72: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

59

dose dependently increased liver toxicity, makers of liver tissue and DNA damage and in both

lean and obese mice.

Page 73: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

60

Introduction

The incidence of obesity has increased in recent years. Also on the rise are obesity

associated disorders such obesity related fatty liver disease (ORFLD) which is a potential risk

factor for diabetes, cardiovascular disease and cancer [71]. As a result, many obese patients take

more drugs compared to non-obese patients to manage some of the associated disorders [74].

Many drugs have been shown to induce liver injury (reviewed in [91]). Additionally there is a

growing body of evidence showing that obesity and ORFLD can increase the risk of drug induced

liver injury (DILI), which could make obese subjects more susceptible to DILI [74].

Obesity related DILI can occur in a few ways. Some drugs such as tamoxifen can

aggravate ORFLD in subjects with a pre-existing condition, while others such as acetaminophen

can induce acute hepatitis in obese subjects [92]. Obesity has been shown to increase oxidative

stress in the liver due to an increase in fatty acid oxidation [93], [94]. This results in a decrease in

antioxidant capacity, such as glutathione levels. When combined with drug induced oxidative

stress the already reduced antioxidant capacity accelerates the progression of DILI [74].

(-)-Epigallocatechin-3-gallate (EGCG) is the most abundant and studied polyphenol in

green tea [5], [95]. Many health benefits have been attributed to EGCG such as the prevention of

cancer, obesity, diabetes and neurodegenerative diseases [96]. In many cases, these biological

effects of green tea have been associated with the antioxidant and a pro-oxidant effects of EGCG

[4], [97], [98]. EGCG has been shown to scavenge reactive oxygen species (ROS) such as

superoxide anions, hydroxyl radicals as well as reactive nitrogen species such as nitric oxide and

peroxynitrite in vitro [4], [9]. By contrast EGCG can also generate ROS by undergoing auto-

oxidation [4].

Many studies have focused on the ability of EGCG to modulate obesity. In a review by

Sae-Tan et al on the effects of green tea polyphenols on body weight and metabolic syndrome,

Page 74: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

61

EGCG was shown to significantly reduce the gaining of body or adipose tissue weight in mice

[29]. Many studies used mice on high-fat diets to demonstrate the anti-obesity effects of EGCG.

In one study for example, mice fed a high-fat (60% of calories) diet, supplemented with 3.2 g/kg

EGCG for 16 weeks showed significantly reduced body weight gain, percent body fat, and

visceral fat weight compared to mice without EGCG treatment [25]. EGCG treatment also

attenuated insulin resistance, and plasma cholesterol, and ameliorated high-fat-diet-induced

hepatic steatosis in mice in mice on the high-fat diet.

The effects of EGCG on body weight in human subjects have been studied in a number of

small, randomized controlled trials (RCTs). Reviews and meta-analyses have been conducted by

Hursel et al in (covering 11 RCTs) and by Phung et al (covering 15 RCTs). The effects of EGCG

(in green tea extract containing caffeine) were studied for 8 to 12 weeks in subjects with normal

weight or who were overweight. As compared with caffeine-free controls, most studies observed

a lowering of body weight [99], [100]. A review by Rains et al on intervention studies showed

that consumption of green tea catechins has beneficial effects on body composition [101]. A

randomized placebo-controlled trial with moderately overweight Chinese subjects showed that

daily consumption of 458 to 886 mg of green tea catechins (with less than 200 mg caffeine) for

90 days reduced total body fat and percent body fat [102]. Although caffeine has been shown to

be a confounding variable in many of these studies, the effects of EGCG on modulating body

weight are observed.

The suggested beneficial effects of EGCG against obesity have encouraged the

consumption of EGCG as a dietary and weight loss supplement. Laboratory and human case

reports have since made an association been these supplements and liver injury. A review by

Mazzanti et al reported several instances where persons showed liver injury as result of taking

EGCG supplements. Symptoms abated when supplementation ceased [45]. Laboratory studies

have demonstrated the hepatotoxic effects of EGCG in mice. For example, Lambert et al showed

Page 75: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

62

a dose dependent increase in toxicity in mice treated with once daily dosing of 750 mg/kg i.g.

EGCG [63]. As discussed, many people consume EGCG supplements for weight loss purposes

and this can cause liver injury. Obesity has also been shown to increase instances of drug induced

liver injury. For example, obesity has been shown to increase toxicity of acetaminophen, a known

hepatotoxin in rats [77]. To date the potential of obesity to increase susceptibility to EGCG

induced hepatotoxicity has not been studied. In the present study, I investigate the effect of

obesity on EGCG mediated hepatotoxicity.

Page 76: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

63

Materials and methods

Chemicals and reagents

EGCG (93% pure) was purchased from Taiyo Green Power (Wuxi, Jiangsu, China).

Diets (Table 3-1) were prepared by Research Diets, Inc. (New Brunswick, NJ).

Table 3-1. Composition of Experimental Diets.

LF HF

Macronutrient Composition

Protein, % kcal

Carbohydrate, % kcal

Fat, % kcal

Ingredient, g/kg

Casein

L-Cystine

Corn Starch

Maltodextrin 10

Sucrose

Cellulose, BW 200

Soybean Oil

Lard

aMineral Mix

20

70

10

200

3

315

35

350

50

25

20

10

20

20

60

200

3

0

125

68.5

50

25

245

10

Page 77: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

64

Table 3-1 continued

Dicalcium Phosphate

Calcium Carbonate

Potassium Citrate. 1 H2O

bVitamin Mix

Choline Bitartrate

13

5.5

16.5

10

2

13

5.5

16.5

10

2

aMineral mix adds the following components (per g mineral mix): sodium chloride, 259

mg; magnesium oxide, 41.9 mg; magnesium sulfate, 257.6 mg; chromium K sulfate, 1.925 mg;

cupric carbonate, 1.05 mg; sodium fluoride, 0.2 mg; potassium iodate, 0.035 mg; ferric citrate, 21

mg; manganous carbonate, 12.25 mg; ammonium molybdate, 0.3 mg; sodium selenite, 0.035 mg;

zinc carbonate, 5.6 mg. bVitamin mix adds the following components (per g vitamin mix):

vitamin A palmitate, 400 IU; vitamin D3, 100 IU; vitamin E acetate, 5 IU; menadione sodium

bisulfite, 0.05 mg; biotin, 0.02 mg; cyanocobalamin, 1 µg; folic acid, 0.2 mg; nicotinic acid 3 mg;

calcium pantothenate, 1.6 mg; pyridoxine-HCl, 0.7 mg; riboflavin, 0.6 mg; thiamin HCl, 0.6 mg

Animal care and treatment

All animal studies were approved by the Institutional Animal Care and Use Committee of

the Pennsylvania State University (University Park, PA, IACUC Protocol 37115). Male

C57BL/6J mice (4 – 5 wks old) were purchased from Jackson Laboratory (Bar Harbor, ME) and

maintained on a 12 h light-dark cycle at 21o C and 38% relative humidity. After a 2 week

acclimation period, mice were randomized into two groups and given ad libitum access to either a

high fat (HF- 60% kcal from fat) diet or a low fat (LF – 10 % kcal from fat) diet. After 9 weeks,

mice were fasted for 7 h (0700 – 1400 h) and then given a single oral bolus dose of saline (0

mg/kg) or EGCG (250, 500 or 750 mg/kg, i.g.) once daily for 2 days. On day 2, mice were

anesthetized and blood was collected by cardiac puncture 1 hour post-gavage. Plasma was

Page 78: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

65

prepared by centrifugation at 3200 g for 15 min and frozen at -80oC. The livers were harvested,

washed with ice-cold PBS, a section was fixed in formalin for histopathology analysis, a 1mm3

section was fixed in modified Karnovsky’s fixative (2.5 % formaldehyde, 1.5 % gluturaldehdye,

0.1 M sodium Cacodylate buffer, 4 % sucrose) for transmission electron microscopy (TEM)

analysis and the remainder was frozen at -80o for later analysis. Formalin-fixed liver tissues were

embedded, sectioned 1 micron thick and stained with hematoxylin and eosin by the Animal

Diagnostics Laboratory, Pennsylvania State University (University Park, PA). The oral bolus

EGCG doses (250, 500 and 750 mg/kg, body weight) are equivalent to 5, 10 and 16 cups of green

tea prepared using a standard method (2.5 g tea leaves in 250 mL) [56].

Western blot analysis

Expression of proteins of interest was determined by western blot. Protein was extracted

from liver samples using Tissue Protein Extraction Reagent (TPER) containing 1% (v/v)

phosphatase and protease inhibitor cocktails (Thermo-Scientific, Rockford, IL). After

homogenization and centrifugation the protein concentration in supernatant was determined by

the Bradford assay (Sigma-Aldrich, St.Louis, MO). Lysates were then combined with an equal

volume of laemmli sample buffer (Bio-Rad, Hercules, CA) and denatured at 90°C for 5 min.

Protein samples (75 μg protein) were resolved by SDS-polyacrylamide (15 %) gel

electrophoresis (PAGE) and transferred to a nitrocellulose membrane (Bio-Rad, Hercules, CA).

Membranes were probed with primary antibody for protein of interest (1:1000 dilution) (Cell

Signaling Technology, Danvers, MA) overnight at 4 °C. After incubation with a fluorescent-

conjugated secondary antibody (1:10, 000 dilution) (Li-Cor Co., Lincoln, NE), bands were

visualized using a Licor Odyssey Imaging System (LI-COR Corporate). Band density was

Page 79: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

66

quantified using Odyssey Application Software Version 3.0. Beta actin was used as a protein

loading control.

Determination of hepatotoxicity

Plasma alanine aminotransferase (ALT) levels were determined spectrophotometrically

(λmax = 340 nm) using a commercially-available assay (CATACHEM Bridgeport, CT) according

the manufacturer’s instructions.

Images of hematoxylin and eosin stained liver tissue taken using under microscope were

visually analyzed for instances of inflammation, necrosis, apoptosis, hepatic lipidosis and

hemorrhage. Each image, 5 each representing 9 – 12 livers per treatment group was scored on a

scale from 0 – 4 based on increase in severity for each pathology. Scores were averaged

according to diet and EGCG treatment to determine overall severity of the different pathologies.

Examination of Mitochondrial Structure

Formaldehyde/glutaraldehyde-fixed samples were washed in 0.1 M Cacodylate buffer, 10

mins each. Liver samples were incubated for 1 hour in 1 – 2 % Osmium tetroxide in 0.1 M

Cacodylate buffer to fix lipids in the sample. This was followed by 2 washes with 0.1 M

Cacodylate buffer and 1 wash in milliQ water. En Bloc staining for 1 hour in 2 % Uranyl acetate

was performed followed by dehydration for 5 – 15 min each in 50, 70, 85, 90, 95, and 100%

ethanol. Three washes for 5 – 15 mins each in EM grade ethanol came next, followed by three

washes with acetone, also for 5 – 15 mins each. Samples were infiltrated with Eponate resin

Page 80: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

67

overnight (50 % acetone, 50 % Eponate). Resin was removed and samples were infiltrated with

100 % Eponate resin three times each for 2 hours. Samples were placed in molds and covered

with fresh 100 % resin and allowed to polymerize overnight at 60 °C. Blocks containing samples

were first trimmed and cut into 50 – 100 nm thick sections with a Leica UC6 ultramicrotome

(Leica Microsystems, Wetzlar, Germany). Slices were collected on mesh grids, stained with 3.4

mg/mL lead citrate for 12 mins, rinsed and allowed to dry prior to imaging with Transmission

electron microscope.

TEM images were analyzed using NIH Image J software. Area of mitochondria was

determined by tracing each whole mitochondrion in the field, and then averaged per field and

number of samples. Density was determined as the number of mitochondria per field and

averaged based on number of fields per sample. Lipid droplets per field were counted and

averaged per sample. Disruption of cristae was determined based on visual analysis of fields and

scored a scale of 0 – 4, with 0 representing absolutely no disruption, 1 = 0 – 25 % disruption, 2 =

25 – 50 % disruption, 3 = 50 – 75 % disruption and 4 = 75 – 100 % disruption. These values were

averaged according to diet and EGCG treatment.

Measurement of hepatic oxidative stress

Malondialdehyde (MDA) levels were determined using a standard Thiobarbituric acid

reactive substances test modified from McDonald and Hultin 1987. Briefly, liver homogenate

prepared for western blot analysis (see above) were also used for MDA analysis. Homogenate

was combined with 6 volume of 100: 3 ratio of A (15% TCA and 0.375% (w/v) in 0.25M HCl)

and B (2% BHT in ethanol (w/v)). Mixture was heated at 95 °C for 15 mins and cooled on ice for

10 mins. Sample was centrifuged at 14 000 g for 15 mins and then incubated for 10 mins at room

Page 81: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

68

temperature. Absorbance was measured at 532 nm using a plate reader. Quantification was made

based on a MDA standard curve.

Reduced glutathione levels were determined using a modified HPLC method from [103].

Briefly, liver tissue (10 – 30 mg) was perfused with PBS containing heparin (0.15 mg/mL).

Tissue was homogenized in 1 – 2 mL PBS containing 2 mM EDTA, centrifuged at 13, 000 g for

10 mins at 4 °C, and the supernatant was collected. The supernatant was combined with 2 vol of

PBS and 3 vol of 1 % metaphosphoric acid, and centrifuged at 13, 000 g for 10 mins at 4 °C. The

resultant supernatant was analyzed by HPLC. Samples were resolved using a SUPELCOSIL LC-

18 column (15 cm x 4.6 mm x 5 µm) and an isocratic mobile phase of 50 mM NaH2PO4 (pH 3.0).

The column was maintained at 30 °C, the flow rate was 0.3 mL/min and the injection volume was

10 µL. GSH detection was carried out on a four channel ESA 5600 CoulArray detector (ESA Inc.

Chelmsford MA) with increasing potentials of 250, 400, 600, and 800 mV applied. Quantified

was based on comparison to a standard curve of GSH in metaphosphoric acid at 600 mV.

Statistical Analysis

All experiments were repeated twice with at least three replicates for each treatment.

GraphPad Prism 5.0 (San Diego, CA) was used for statistical analysis. In this experiment two

outcomes were focused on; the effect of varying doses of EGCG and the difference between diets

at each dose of EGCG. To determine differences between control (0 mg/kg) and varying doses of

EGCG (250, 500, and 750 mg/kg), one-way ANOVA with Dunnett’s post-test was used to

compare differences in plasma ALT, hepatic glutathione, MDA levels, liver weight and severity

of tissue damage induced by different doses of EGCG, (* = P < 0.05, ** = P < 0.01, *** = P <

0.001). The Student’s t-test was used to compare differences between diets (HF and LF) at each

Page 82: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

69

dose of EGCG, (# = P < 0.05, ## = P < 0.01, ### = P < 0.001). Data are expressed as mean ±

SEM and significance was reached at P<0.05.

Page 83: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

70

Results

Effect of EGCG on general markers of toxicity

Figure 3-1. Survival rate and changes in body weight of male C57BL/6J mice following

treatment with EGCG.

Mice we fed either a high fat or low fat diet for mice 9 weeks and then treated with 0,250, 500, or

750 mg/kg b.w. EGCG once daily for 2 days.

At the end of the first day of treatment lean and obese mice given 0 and 250 mg/kg

EGCG, and lean mice given 500 mg/kg EGCG suffered no casualties (100 % survival) (Figure 3-

1). By contrast, obese mice given 500 and 750 mg/kg EGCG, and lean mice give 750 mg/kg

EGCG had 77%, 67% and 91% survival rates, respectively. After the second day of treatment

survival rates for lean and obese mice given 0 and 250 mg/kg EGCG stayed the same at 100 %,

but lean mice given 500 mg/kg had an 86% survival rate. Lean mice given 750 mg/kg EGCG had

a 91 % survival rate. By contrast, obese mice given 500 and 750 mg/kg EGCG had rates of 68%

and 25%. Body weight for male C57BL/6J mice treated with 0, 250, 500, or 750 mg/kg b.w.

EGCG were higher in obese mice compared to lean mice. Obese mice treated with 750 mg/kg

EGCG showed the largest decrease in body weight over the 2 day treatment period, but it was not

Page 84: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

71

significant. Obese mice treated with 250 and 500 mg/kg also showed decreasing trends in body

weight, but these too were not significant. Body weight of obese control mice remained constant.

An insignificant decreasing trend in body weight can be seen in lean mice treated with 750 and

500 mg/kg EGCG. Lean mice treated with 0 and 250 mg/kg EGCG remained fairly constant over

the 2 day period.

Effect of EGCG on general markers of liver toxicity

Figure 3-2. Plasma alanine aminotransferase levels and liver weight in male C57BL/6J mice

treated with oral EGCG.

ALT levels and liver weights were determined following administration of 2 once daily doses of

EGCG by oral gavage. Each bar represents the mean of N= 8-18 and error bars represent SEM.

ALT levels represent mice treated with 0, 250, 500, or 750 mg/kg b.w. EGCG. Liver weights

represent mice treated with 0, 250 or 500 mg/kg b.w. EGCG. Mice were fed either a low fat (LF)

or a high fat (HF) diet. Liver tissue was weighed immediately after it was excised for the mouse.

For liver weight each bar represents the mean of N= 6 – 10. Error bars represent SD. Data were

analyzed by one-way ANOVA based on dose with Dunnett’s post-test and compared to controls

(* = P < 0.05, ** = P < 0.01, *** = P < 0.001) and t-test for diets (# = P < 0.05, ## = P < 0.01,

### = P < 0.001).

Page 85: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

72

Alanine aminotransferase (ALT), a marker of liver injury was measured in plasma

samples of lean and obese mice treated with 0, 250, 500, 750 mg/kg EGCG (Figure 3-2). A dose

dependent increase was observed in lean mice. ALT levels increased 50 and 150 fold at the 500

and 750 mg/kg dose respectively, in lean mice. In mice fed a HF diet a significant increase in

ALT can only be seen in mice given a 500 mg/kg dose, (80 fold) when compared to controls. A

55 fold increase occurred for mice that received 750 mg/kg EGCG, but it was not significant. The

decrease seen at the 750 mg/kg dose in obese mice could be as a result of the mice that

succumbed to EGCG treatment during the study, so plasma from these subjects were not

available for analysis at that dose (refer to survival data, Figure 3-1). These mice potentially had

higher levels of ALT compared to the ones that survived, hence the reason they did not survive,

and could account for why the remaining mice had lower ALT levels. There were no significant

differences between diet groups at the different doses of EGCG. Treatment with EGCG had no

significant effect on liver weights of obese or lean mice. In general obese mice had higher liver

weights than lean mice with significant differences seen in mice treated with 250 and 500 mg/kg

EGCG. At these two doses obese mice had significantly higher liver weights than lean mice.

Page 86: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

73

Histological evaluation of the hepatotoxic effect of EGCG

Figure 3-1. Representative images of histopathological analysis of stained liver tissue from male

C57BL/6J mice.

Mice were fed either a low fat (LF) or a high fat (HF) diet and treated with 0, 500, or 750 mg/kg

b.w. EGCG. Tissue was fixed in formalin and slices were stained with hematoxylin and eosin for

imaging and analysis. Images are shown at 200 x magnification. Arrows indicate blood vessels

(central veins) and circles indicate area necrotic tissue.

Page 87: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

74

Images of liver tissue stained with hematoxylin and eosin were analyzed to examine the

effect of treatment with EGCG. Representative images (Figure 3-3) show normal liver tissue for

control obese and lean mice (HF-0 and LF-0). Clear and defined cellular structures are present as

well as blood vessels such as portal and central veins (denoted by arrows). Effects of EGCG can

be seen in the HF-500 image, where the circled area denotes the presence of necrosis and

hemorrhage. Compared to HF-500, the image for LF-500 does not show any effects of EGCG.

Effects of EGCG can be seen in the images for both LF-750 and HF-750, indicated by circled

regions. Based on these images a dose dependent effect of EGCG can be seen, but is not

sufficient to demonstrate the extent of EGCG toxicity. Quantification of the markers of liver

tissue damage will provide a more comprehensive analysis of the hepatotoxic effects of EGCG

(Figure 3-4).

Figure 3-4. Quantification of markers of liver tissue damage based on histopathological analysis.

Inflammation, necrosis, apoptosis, hemorrhage, hepatic Lipidosis were assessed on a scale of 0-4

for images of each section, where 0 was no presence and 4 was highest presence. Severity was

determined as the average presence within a treatment group. Each bar represents the mean of N=

Page 88: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

75

8-12 images. Error bars represent SD. Data were analyzed by one-way ANOVA based on dose

with Dunnett’s post-test and compared to controls (* = P < 0.05, ** = P < 0.01, *** = P < 0.001).

Quantification of stained liver tissue severity of inflammation, necrosis, apoptosis,

hepatic lipidosis and hemorrhage, showed an overall dose dependent increase in severity for all

pathologies. Both obese and lean mice showed significant increases in inflammation (Figure 3-4)

at the 750 mg/kg dose compared to the controls (0 mg/kg). The increase in severity of tissue

necrosis was significant for both the 500 and 750 mg/kg doses in lean and obese mice compared

to controls. EGCG caused significant increases in apoptosis at both the 500 and 750 mg/kg doses

compared to controls for mice fed a HF diet. Mice fed a LF diet showed a significant increase in

apoptosis only when given a 750 mg/kg dose of EGCG. There was a dose dependent increase in

hemorrhaging for mice fed both a HF and LF diet. The increase was only significant for mice

given a 750 mg/kg dose of EGCG. Hepatic lipidosis increased significantly for both diet groups.

There was a significant increase for the obese mice only at the 750 mg/kg dose, but for the lean

mice the increase was significant at both the 500 and 750 mg/kg doses. No significant differences

were observed between HF treated mice and LF treated mice at the different doses of EGCG.

Page 89: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

76

Effect of EGCG on Mitochondrial Structure

Figure 3-5. Representative images from transmission electron microscopy (TEM) analysis of

liver tissue of male C57BL/6J mice.

Mice were fed either a low fat (LF) or a high fat (HF) diet and treated with 0, or 500 mg/kg b.w.

EGCG. Liver tissue was fixed formaldehyde and glutaraldehyde, treated with Osmium tetroxide,

negatively stained with Uranyl acetate, dehydrated with ethanol, infiltrated with resin,

polymerized, sectioned, stained positively with lead citrate and imaged. Images are shown at

1000 x magnification. Arrows indicate fat droplets. Circles indicate mitochondria.

Transmission electron microscopy enabled us to obtain images of the mitochondria in

obese and lean mice treated with 0, or 500 mg/kg EGCG at 1000 x magnification (Figure 3-5).

These images were analyzed using Image J software and for lean and obese mice treated with 0

mg/kg EGCG the most significant difference observed is in the lipid droplet content. Lipid

droplets, indicated with an arrow, are in a larger quantity in obese mice when compared to lean

Page 90: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

77

mice. At that dose, the density of mitochondria; mitochondria are indicated with a circle, appears

to be higher in lean mice. At 500 mg/kg EGCG, obese mice showed a higher number of lipid

droplets compared to lean mice and a higher mitochondrial density. Changes in mitochondrial

area and density, disruption of cristae, and number of lipid droplets are shown in Figure 3-6.

Figure 3-6. Quantification of mitochondrial parameters from TEM analysis.

Images were analyzed for mitochondrial area, density, number of lipid droplets, and cristae

disruption. Quantification was done using Image J software. Each bar represents the mean of N=

5-10 fields. Error bars represent SEM. Data were analyzed by one-way ANOVA based on dose

with Dunnett’s post-test and compared to controls (* = P < 0.05, ** = P < 0.01, *** = P < 0.001)

and t-test for diets (# = P < 0.05, ## = P < 0.01, ### = P < 0.001).

The areas of the mitochondria were measured for mice fed both diets that received the

doses of EGCG (Figure 3-6). Although not significant, the data shows a decreasing trend in areas

of mitochondria of obese mice and an increasing trend in areas of mitochondria of lean mice.

There was a significant difference in areas between lean and obese not treated with EGCG.

Page 91: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

78

Obese mice had significantly larger mitochondria when compared to lean control mice. Treatment

with 500 mg/kg EGCG yielded no difference between mice.

Another parameter measured was the density of mitochondria in the images obtained

(Figure 3-6). Mitochondrial density defined as the number of mitochondria per image revealed no

difference for obese mice at the various doses of EGCG. Lean mice, however, had significantly

less mitochondria per image at 500 mg/kg EGCG, when compared to controls (P < 0.05). Also at

this dose, obese mice had significantly more mitochondria than lean mice.

The cristae of the mitochondria, which are formed by the inner mitochondrial membrane,

were analyzed to measure how intact they were. Disruption of cristae is a marker of

mitochondrial dysfunction. There was no significant difference in obese mice from 0 to 500

mg/kg EGCG. Lean mice showed a decreasing trend, but it was not significant. When compared,

control lean mice appear to have higher levels of cristae disruption compared to their obese

counterparts. This difference however, was not significant. No significant difference was seen

between lean and obese mice for the 500 mg/kg dose of EGCG.

The number of lipid droplets in each image was measure for lean and obese mice. Figure

3-6 shows that obese mice had significantly lower number of lipid droplets at 500 mg/kg when

compared to controls. There was no significant difference for lean mice in the number of lipid

droplets at the different doses of EGCG, but an increasing trend from 0 to 500 mg/kg EGCG can

be seen. Additionally, HF control mice had a significantly higher number of lipid droplets when

compared to lean control mice.

Page 92: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

79

Effect of EGCG on Markers of Oxidative Stress

Hepatic levels of reduced glutathione (GSH) were lower in obese mice compared to mice

fed a LF diet regardless of the EGCG treatment (Figure 3-7). This difference between diets was

significant only at the 0 and 500 mg/kg doses (P < 0.001 and P < 0.05 respectively). EGCG

treatment caused an insignificant dose dependent decrease in levels of GSH in lean mice. Similar

results were observed in obese mice from 250 to 750 mg/kg EGCG, but that too was not

significant.

0 250 500 7500.00

0.05

0.10

0.15

0.20HF

LF###

#

Dose (mg/kg)

GS

H (

uM

/mg

Tis

su

e)

Figure 3-7. Hepatic levels of reduced glutathione (GSH) were measured using HPLC and

normalized to tissue weight.

Each bar represents the mean of N= 8-18. Error bars represent SEM. Data were analyzed by one-

way ANOVA based on dose with Dunnett’s post-test and compared to controls, and t-test for

diets (# = P < 0.05, ## = P < 0.01, ### = P < 0.001).

Phosphorylated histone 2Ax, a marker for DNA oxidative damage increased in a dose

dependent manner for mice in both diet groups (Figure 3-8). No expression of γH2Ax was

observed at the low dose, 250 mg/kg. A significant increase (P < 0.01) was seen for mice given a

750 mg/kg dose of EGCG in both diet groups when compared to controls.

Page 93: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

80

Figure 3-8. Hepatic levels of γ-H2AX determined by western blot analysis.

γ-H2AX was determined in mice following 2 daily doses of 0, 250, 500, or 750 mg/kg, i.g.

EGCG. Expression was determined by densitometry and normalized to β-actin. The blot shows

representative expression patterns. The histogram shows quantification of all samples analyzed

(N = 8-18 per treatment group). Error bars represent SEM. Data were analyzed by one-way

ANOVA based on dose with Dunnett’s post-test and compared to controls (* = P < 0.05, ** = P <

0.01, *** = P < 0.001) and t-test for diets.

Levels of MDA in liver tissue which correlated to the extent of lipid peroxidation in cells

increased significantly for mice in both diet groups (Figure 3-9). The increase was dose

dependent for mice on a LF diet with those given a 750 mg/kg dose having the most significant

increase compared to controls. Mice on a HF diet had a significant increase in MDA levels when

given a 500 mg/kg dose of EGCG. This increase was also significantly higher than mice on the

LF on that dose. An increase was observed for HF mice given a 750 mg/kg dose of EGCG, but it

was not significant.

Page 94: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

81

0 250 500 7500

2

4

6HF

LF**

*

#

Dose (mg/kg)

uM

MD

A/m

g p

rote

in

Figure 3-9. Hepatic levels of malondialdehyde (MDA).

MDA was measured using the standard thiobarbituric acid-reactivity test as a marker of lipid

peroxidation and peroxidative tissue injury. Measurements were normalized to tissue protein

concentrations. Each bar represents the mean of N= 8-18. Error bars represent SEM. Data were

analyzed by one-way ANOVA based on dose with Dunnett’s post-test and compared to controls

(* = P < 0.05, ** = P < 0.01, *** = P < 0.001) and t-test for diets (# = P < 0.05, ## = P < 0.01,

### = P < 0.001).

Page 95: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

82

Discussion

In the present study we examined the effect of obesity on EGCG mediated hepatotoxicity.

Survival data shows that EGCG treatment induced toxicity most in obese mice treated with 750

mg/kg, evident by a 25 % survival rate at the end of the two day treatment when compared to all

other mice (Figure 3-1). Toxic effects were also seen in obese mice treated with 500 mg/kg

EGCG as they had a survival rate of 68 % at the end of the treatment period. When compared to

the lean mice at the 500 and 750 mg/kg doses the survival data shows that obese mice had lower

survival rates indicating that they were more susceptible to EGCG toxicity.

Using a decrease in body weight as a general marker of toxicity, it can also be seen in the

data that obese mice treated with 750 mg/kg EGCG showed the most decrease in body weight

(Figure 3-1). Decreases in body weight after the 2 day treatment period was evident for all obese

mice except controls. When compared to lean mice EGCG treatment caused slight decreases only

in lean mice treated with 500 and 750 mg/kg EGCG. This also indicates that obese mice were

more affected by the EGCG treatment.

As previously stated, obesity has been shown to increase instances of drug induced liver

injury. We examined the effects of EGCG treatment on general markers of liver injury. Plasma

alanine aminotransferase (ALT) levels increased for both diet groups at the 500 and 750 mg/kg

doses of EGCG (Figure 3-2). This data is consistent with findings from previous studies [63]. At

the 500 mg/kg dose of EGCG, mice on a HF diet had higher levels of plasma ALT compared to

lean. Though not significant, this difference indicates that HF mice were more susceptible to the

toxic effects of EGCG at that dose. This is in keeping with studies that have shown that obesity

can increase drug induced liver injury. In contrast, at the 750 mg/kg dose, EGCG appeared to

have more toxic effects on mice fed a LF diet. Based on the survival data, obese mice treated with

750 mg/kg EGCG had a low survival rate (25%) at the end of the 2 treatment period. The loss of

Page 96: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

83

mice during this time decreased the number of samples obtained for plasma based analyses, but

not liver tissue based analyses. As a result this affected the changes seen at the 750 mg/kg dose

for markers measured using plasma samples. The death of these mice suggests they succumbed to

EGCG mediated toxicity at this dose, which is not reflected in the data due to loss of these mice.

In the case of ALT levels it can be seen at the 750 mk/kg dose of EGCG, obese mice had lower

levels compared to lean mice. This could be explained by the fact that obese mice given a 750

mg/kg dose of EGCG and having higher levels of plasma ALT, succumbed to the toxic effects

prior to completion of the study. As a result these measurements could not be reflected in data,

thus showing lower ALT levels at this dose, compared to the higher levels that were expected.

Liver weight data showed no differences between obese and lean control mice. However,

when treated with 250 and 500 mg/kg EGCG obese mice had significantly higher liver weight,

indicative of larger livers. This can be attributed to obese mice developing hepatomegaly as a

result of EGCG treatment. Hepatomegaly has been shown to be increased in instances of DILI

[104]. This data is consistent with that obtained so far, and supports the conclusion that EGCG

has caused more liver injury in obese mice compared to lean mice.

Representative images of stained liver tissue of obese and lean mice treated with 0, 500,

or 750 mg/kg EGCG showed markers of liver tissue damage evidenced by signs of hemorrhaging,

the presence of apoptotic bodies, necrosis, inflammation and hepatic lipidosis. Based on images,

the severity of tissue damage increased with dose of EGCG in both diet groups, Figure 3-3. These

observations correlate well with data from previous studies done in our lab, where EGCG caused

a dose dependent increase in liver tissue damage [63], [105]. However, no discernable

differences were observed between diet groups based solely on images.

Data obtained from quantification of histological analysis supported the observations

seen in tissue images. It can be seen from Figure 3-4 that EGCG caused significant increases in

severity of tissue inflammation, necrosis, apoptosis, hemorrhage and hepatic lipidosis in mice fed

Page 97: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

84

HF and LF diets. Closer examination of the data showed that there was no difference between

obese and lean mice in severity of inflammation when mice received 0, 500, or 750 mg/kg dose

of EGCG, indicating that EGCG affected mice on both diets in much the same way when

triggering an inflammatory response. A similar effect was seen in the severity of tissue necrosis

caused by EGCG, whereby there were no significant differences between mice on HF diet or LF

diet.

The effect of EGCG on the severity of apoptosis, though not significant, can be seen to

vary between lean and obese mice at the different doses of EGCG. Mice on HF diet recorded no

incidence of apoptosis when they received 0 mg/kg EGCG. This observation is interesting to note

as obesity has been shown to induce apoptosis in both liver tissue and skeletal muscle [106],

[107], but this was not observed here. We expected to see more significant differences in

apoptosis between obese and lean mice because of the ability of an obese diet to induce apoptosis,

and when compounded with the toxic effects of EGCG, the severity would be higher in obese

mice, but we did not see that. Obesity has been shown to increase blood flow to the liver as a

result of increased lipid accumulation. Consequently, this increased blood flow and increased fat

deposition compromises blood vessels and increases the risk of rupture [73]. This suggests that

obese mice are at increased risk of tissue hemorrhage. However, we observed no significant

differences between lean and obese mice.

In mice treated with acetaminophen, a known hepatotoxin, hepatic lipidosis was shown to

increase along with hepatic necrosis [108]. Similar results were seen in this study, confirming the

hepatotoxic effects of EGCG. As expected high fat fed control mice had higher levels of hepatic

lipidosis. Interestingly, the severity of hepatic lipidosis in mice fed a LF diet was not significantly

different from HF fed mice as one would expect. Several studies have shown that a high

carbohydrate diet can also lead to lipid accumulation in the liver and contribute to nonalcoholic

fatty liver disease (NAFLD) [109][110] [111]. Additionally, the type of carbohydrate in the diet

Page 98: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

85

has also been shown to cause fat accumulation in the liver with rapidly absorbed carbohydrates

(RAC) contributing more significantly to hepatic steatosis [112]. Based on the diet composition

from Table 3-1 it can be seen that mice on LF diet had significantly more carbohydrates in their

diet than mice on HF diet, (LF = 70 kcal from carbs, HF = 20 kcal from carbs). Also from Table

3-1 we see that the LF diet has 315 g/kg corn starch vs 0 g/kg for the HF diet. Corn starch is made

up mostly of amylopectin, which is a RAC, and as studies have shown, these contribute

significantly to fat accumulation in the liver. The high carbohydrate content may only be part of

the reason we see no differences between lean and obese mice at the 500 and 750 mg/kg doses,

because lean control mice had no hepatic lipidosis. It could be that EGCG treatment exacerbates

hepatic lipidosis in mice on a high carbohydrate diet.

To get a better understanding of the hepatotoxic effects of EGCG, we examined the effect

of EGCG on mitochondrial structure. Transmission electron micrographs of treated liver tissue

were assessed for changes in mitochondria area, density, circularity, lipid droplet size and

disruption of mitochondria cristae. EGCG has been shown to increase mitochondrial ROS and

trigger mitochondrial dysfunction [18]. Changes in mitochondrial structure may be able to

confirm the hepatotoxic capabilities of EGCG. Mitochondrial are constantly undergoing fusion

and fission, which can impact their size and density at any given time, illustrated in Figure 3-10.

Page 99: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

86

Figure 3-10. Relationship between mitochondrial morphology and function. [113]

Fusion and fission work to maintain the mitochondrial function especially under

environmental stress such as an increase in ROS. While fusion results in larger, more elongated

mitochondria, fission results in fragmented or smaller mitochondrial, and the rates typically

balance each other [114], [115]. Fusion of mitochondria requires intact membranes and stable

membrane potential, interruption of fusion can lead to increased fission. This can be induced by

ROS or apoptosis [116]. Fusion appears to play a role in protecting cells from cell death or in

mitigating stress, in that mitochondria with damaged DNA or defects can fuse with other

mitochondria to allow complementing of DNA for improved function [114]. Fission is generally

triggered by apoptosis, and can result from loss of mitochondrial membrane potential. This can

lead to swelling and opening of mitochondrial permeability transition pore, and is necessary for

the removal of damaged mitochondrial in times of stress, and for the formation of new

mitochondria [115]–[117]. Non-apoptotic cells treated with 3.3, 9.8 or 16.3 mM hydrogen

Page 100: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

87

peroxide for 4, 24 and 48 hours showed a dose dependent increase in fragmentation after 4 hours

of treatment. After 24 hours, there was intermediate fragmentation and some recovery of

mitochondria, evident by the increase in tubular (normal) shaped mitochondria. 48 hours of

treatment showed recovered mitochondria [118]. In the same study, treatment with hydrogen

peroxide caused an increase in both fission and fusion, and an increase in cristae disruption. An

increase in spherical mitochondrial was also seen and was associated with increase fragmentation.

Our examination of mitochondrial area in electron microscopy images showed no

significant differences in obese mice treated with 0 or 500 mg/kg EGCG. Obese control mice

showed significantly larger mitochondrial areas compared to lean control mice. Since there is no

effect of EGCG treatment to consider, the difference seen could be an effect of diet. Larger

mitochondrial areas generally indicate the mitochondria are undergoing fusion. Fusion has been

shown to be an adaptive mechanism when some mitochondria are damaged, and before they are

destroyed they can complement other mitochondria [114]. In this case, stress brought on by an

obese diet could be causing some mitochondrial damage, but not sufficient to induce mitophagy,

so the mitochondria are probably complementing each other in response to the stress and as result

they have larger areas.

Mitochondrial density, indicative of the number of mitochondria on the image, showed

no differences in obese mice with respect to EGCG treatment. Lean mice given 500 mg/kg

showed a significant decrease in density compared to control, which can indicate an increase in

mitochondrial fusion. As previously discussed, increased mitochondrial density is an indication of

mitochondrial fusion. This can occur when there is a need for mtDNA complementation. In this

case, EGCG treatment could have caused changes in mitochondrial oxidative environment

resulting in mtDNA damage, and warrant mitochondrial fusion. The significant difference

observed between lean and obese mice at 500 mg/kg EGCG could also be explained by a

difference in the fusion and fission cycles at that point.

Page 101: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

88

Cristae disruption is a marker of mitochondrial dysfunction. Here we see that there is no

effect of EGCG on cristae disruption in obese mice, which suggests the EGCG had no effect on

obese mice. This conflicts with the data we have obtained thus far, which clearly indicates the

hepatotoxic effects of EGCG. In lean mice, although a slight decrease in cristae disruption is

observed, it is not significant.

The number of lipid droplets was determined from the TEM images. Obese mice showed

a significant decrease in the number of lipid droplets at 500 mg/kg EGCG, when compared to

control. This could be as a result of the anti-obesity effects of EGCG, as seen in previous studies,

where EGCG was shown to reduce lipid accumulation and increase fecal lipid content in high fat

fed mice [28]. Also seen, is a significant difference in the number of lipid droplets between obese

and lean controls. This data is supported by the TEM images (Figure 3-5) and shows that for the

samples chosen obese mice had more lipid droplets. This is also consistent with hepatic lipidosis

data (Figure 3-4).

Hepatic levels of reduced glutathione (GSH) were evaluated as a marker for oxidative

stress. Previous studies have shown that EGCG-mediated liver toxicity was associated with

increased hepatic oxidative stress [85][63]. Here, we found that EGCG dose dependently depleted

GSH levels in both diet groups (Figure 3-7). This data is consistent with previous studies [105].

When comparing GSH levels between diets, GSH levels were significantly lower in mice fed a

HF diet. Studies have shown that fatty acid oxidation is a source of ROS in fatty livers, which

causes oxidative stress and can lead to depleted GSH levels in cells [74], [78], [119]. Our

findings support what has been seen in the literature.

Phosphorylation of histone 2Ax (γ-H2Ax) occurs as a result of double strand breaks to

DNA, and is used a biochemical marker for DNA damage. In this study we measured protein

expression of γ-H2Ax to assess the effect of EGCG treatment on cellular DNA as a marker of

liver toxicity. As can be seen in Figure 3-8, EGCG caused a dose dependent increase in

Page 102: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

89

phosphorylated histone 2Ax. Quantification of the bands corroborates the data seen in the western

blot images. These results suggest that EGCG caused double strand breaks in cellular DNA. DNA

damage has been shown to induce apoptosis and necrosis [120], both of which were previously

seen to increase dose dependently by EGCG treatment. These findings are not only consistent

with previous studies [63], but support other data obtained in the study. No differences were

observed between diet groups at the different doses of EGCG, suggesting that EGCG treatment

affected mice on either diet in a similar manner.

Lipid peroxidation of fatty acids in cells has been shown to be initiated by ROS and

exacerbated by oxidative stress. This results in the formation of aldehydes among other

compounds which can be used as markers for oxidative stress in tissue [121]. Malondialdehyde,

one of the by-products of lipid peroxidation was measured in this study to determine the

magnitude of oxidative stress in liver tissue. We found that MDA levels increased significantly in

both obese and lean mice (Figure 3-9). MDA levels have been shown to increase in liver tissue

by known hepatotoxicant-acetaminophen, and this has been used as a marker for acetaminophen

induced toxicity [122]. Lambert et al have also showed that EGCG increased MDA levels in mice

treated with high doses of EGCG [63]. Our data confirms that EGCG caused significant oxidative

stress in mice fed an HF or LF diet. As seen with the ALT data, mice on a HF diet that received a

500 mg/kg dose of EGCG had significantly higher levels of MDA than LF fed mice. Obesity

related fatty livers have been shown to have increased levels of MDA because of the presence of

excess fatty acids, which increase ROS and levels of oxidative stress [78]. These effects as we

have seen are worsened by EGCG and can explain why HF mice have higher levels of MDA.

Similar to ALT data, at the 750 mg/kg the increase in not as significant, and is lower that of the

500 mg/kg dose. These results can be explained in much the same way, in that the death of obese

mice treated with 750 mg/kg EGCG did not make it to the end of the study, due to severe liver

damage, could not be included in the analysis, and as such were not represented.

Page 103: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

90

In summary EGCG induced hepatotoxicity in a dose dependent manner in both lean and

obese mice. Toxicity in the mice was evident in the decrease in survival rates in both lean and

obese mice. Hepatotoxicity was characterized by an increase in liver tissue injury seen in

increased ALT levels and tissue damage. EGCG treatment also increased oxidative stress in liver

tissue seen by depleted glutathione levels and increase phosphorylated histone 2Ax and

malondialdehyde levels. Survival data, liver weight data, GSH data, as well ALT and MDA data

all indicate that obese mice, particularly mice treated with 750 mg/kg EGCG were more

susceptible to EGCG treatment when compared to lean mice.

Page 104: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

Effect of High Dose Oral (-)-Epigallocatechin-3-gallate (EGCG) on Hepatic

Mitochondrial Antioxidants, Biogenesis and Function

Abstract

(-)-Epigallocatechin-3-gallate (EGCG) has been shown to induce hepatotoxicity in a dose

dependent manner in vivo. The underlying mechanism of these effects has been poorly studied. In

vitro, EGCG was shown to cause cytotoxicity by inducing mitochondrial reactive oxygen species

ROS and mitochondrial dysfunction. In this study we examined the effects of EGCG on factors

involved in mitochondrial biogenesis and function as well as mitochondrial antioxidant gene

expression and activity in an effort to shed light on the underlying mechanism of high dose oral

EGCG-mediated hepatotoxicity. Additionally, we explored the effects of obesity in potentially

exacerbating mitochondrial dysfunction. We found that EGCG significantly decreased mRNA

expression of mitochondrial superoxide dismutase in lean mice when treated with 750 mg/kg, but

not obese mice. EGCG treatment significantly decreased mRNA levels of catalase in both lean

and obese mice, and catalase activity in obese mice, while both lean and obese mice showed

significant decreases in protein expression of glutathione peroxidase. EGCG treatment

significantly decreased mRNA levels of glutathione-S-transferase zeta 1 and peroxiredoxin 3.

Metallothionein gene and protein expression was significantly upregulated by EGCG in both lean

and obese mice. EGCG treatment caused significant down regulation of antioxidant gene

regulators, Sirtuin 3 and forkhead box O3 and mitochondrial biogenesis regulator peroxisome

proliferator-activator receptor coactivator-1a. Mitochondrial transcription factors A, B1 and B2

were significantly reduced by EGCG, as well as complex I subunit Ndusf 8 and complex III

Page 105: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

92

subunit Uqcrc 1. Mitochondrial uncoupling protein 2 was significantly upregulated. These data

suggests that EGCG depletes mitochondrial antioxidant capacity by decreasing expression and

activity of antioxidant enzymes and their regulators, and inhibits mitochondrial biogenesis and

function by down regulating the regulator and transcription factors at the gene expression level.

Page 106: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

93

Introduction

Green tea polyphenol (-)-Epigallocatechin-3-gallate (EGCG) has been widely studied for

its numerous health benefits. EGCG has been shown to be effective in reducing fat accumulation

and modulating effects of metabolic syndrome in laboratory models; it has demonstrated

profound anti-inflammatory effects and shown to be effective in treatment and prevention of

cancer [9], [123].

Many of the cancer chemopreventative effects of EGCG have been attributed to its ability

to function as both an antioxidant and a pro-oxidant in vitro and in vivo [4], [9]. As an

antioxidant EGCG has been shown to scavenge reactive oxygen species (ROS) and chelate

transition metals such as iron in vitro, which are capable of triggering the Fenton reaction and

initiating lipid peroxidation [4], [8]. In its pro-oxidant capacity, EGCG has been shown to

undergo auto-oxidation and generate ROS, which can among other things induce apoptosis in

cancer cells [4], [97], [124].

Another property of EGCG that has been well-regarded is its anti-obesity effects. Several

studies have highlighted the role of EGCG in the modulation of obesity and its comorbidities

such as obesity related fatty liver disease (ORFLD). Grove et al showed that EGCG inhibited

digestive enzyme pancreatic lipase, and reduced body weight gain in high fat fed mice. Sae-Tan

et al showed that EGCG increased expression of genes related to fatty acid oxidation in high fat

fed mice. Bose et al showed that in high fat fed mice supplemented with 0.32% EGCG for 16

weeks, EGCG was able to ameliorate high fat-diet induced ORFLD [28], [29][25].

These beneficial properties of EGCG have led to its increased use in forms such as

dietary supplements, as it has generally been considered natural and safe. Case reports, however,

have since made an association between instances of liver injury and high concentrations of green

tea polyphenols found in these formulations [45], [125]. Cellular and in vivo studies have

Page 107: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

94

confirmed the hepatotoxic potential of EGCG. Galati et al showed the cytotoxic effects of EGCG

in isolated rat hepatocytes, where it collapsed mitochondrial membrane potential and generated

ROS [85]. Lambert et al showed the hepatotoxic effects of EGCG in mice treated with a single

daily dose of 1500 mg/kg i.g. or 2 daily doses of 750 mg/kg i.g, which increased tissue necrosis,

plasma alanine aminotransferase (ALT) and markers of oxidative stress [63].

In a recent study by Tao et al., the cytotoxic effects of EGCG in oral cancer cells were

shown to be mediated by the induction of mitochondrial ROS and mitochondrial dysfunction

[18]. Qanungo et al also showed that EGCG induced mitochondrial membrane depolarization and

caspase-dependent apoptosis in pancreatic cancer cells [126]. The mitochondria plays an

important role in the energy requirements of the cell, and through the electron transport chain

(ETC) it has been shown to be an endogenous source of ROS in the cell [127], [128]. As such,

damage to the mitochondrial has been implicated in a number of pathogeneses such as drug

induced liver injury (DILI) and non-alcoholic fatty liver disease (NAFLD) [91], [129].

Hepatotoxic drugs can cause mitochondrial dysfunction if a variety of ways, including

direct inhibition of mitochondrial respiration, damage to mitochondrial DNA and most

commonly transformation of the parent compound into reactive metabolites the can generate ROS

[91]. There are many antioxidant systems in the mitochondria to combat ROS, and inhibition of

any of these systems can increase oxidative stress and lead to injury and dysfunction. These

include the glutathione and thioredoxin redox systems and antioxidant enzymes such as

superoxide dismutase, catalase, glutathione peroxidase and peroxiredoxin [130]. Acetaminophen,

for example, has been shown to cause liver injury by generating toxic metabolites at high doses,

which covalently bind to DNA proteins and depleting GSH levels which increases ROS. This

leads to increased mitochondrial oxidative stress, opening of the mitochondrial permeability

transition pore and eventually mitochondrial dysfunction [131]–[134].

Page 108: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

95

Mitochondrial dysfunction has been implicated in the progression of NAFLD.

Mitochondrial dysfunction affects lipid homeostasis in the liver that can lead to an

overproduction of reactive oxygen species (ROS) that trigger lipid peroxidation, cytokine

overproduction and cell death [135], [136]. Rector et al, showed that hepatic mitochondrial

dysfunction as determined by reduced hepatic carnitine palmitoyl-CoA transferase-1 activity,

fatty acid oxidation, and cytochrome c protein in an obese mouse model preceded insulin

resistance and hepatic steatosis [137]. Liver tissue from individuals with non-alcoholic

steatohepatitis was analyzed using electron microscopy and it was found that 9 out of 10 subject

exhibited mitochondrial dysfunction as evidence by loss of mitochondrial cristae [138]. Thyfault

et al showed that rats bred for low aerobic capacity had reduced hepatic mitochondrial oxidative

capacity and showed increased susceptibility to hepatic steatosis[139].

In the previous chapter we demonstrated the hepatotoxic effects of EGCG. The

underlying mechanism associated with EGCG mediated hepatotoxicity remains unclear. The

mitochondria have been shown to be a target of EGCG induced ROS in inducing cytotoxicity in

cancer cells. The effects of EGCG mediated liver toxicity on factors involved in mitochondrial

antioxidant capacity, mitochondrial function and biogenesis in vivo, have not been studied.

Additionally, the effect of obesity on these factors in EGCG mediated toxicity remains unclear.

In this study, I investigate the effects of high dose EGCG on mitochondrial antioxidant

capacity, function and biogenesis, as well as the effect of obesity in potentially exacerbating the

hepatotoxic potential of EGCG.

Page 109: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

96

Materials and methods

Animal studies

The analyses reported in this Chapter used samples derived from C57BL6/J mice treated

in Chapter 3. In brief, male C57BL/6J mice (4 – 5 wks old, Jackson Laboratory, Jackson, MS)

were randomized into two groups and were on either a high fat (HF- 60% kcal from fat) diet or a

low fat (LF – 10 % kcal from fat ) diet. Following a 9 week period on the respective diets, mice

were fasted for 7 h (0700 – 1400 h) and then given a single dose of saline (0 mg/kg) or EGCG (

250, 500 or 750 mg/kg, i.g.) once daily for 2 days. On day 2, 1 hour post-gavage, mice were

anesthetized and blood was collected by cardiac puncture. Plasma was prepared by centrifugation

at 3200 g for 15 min and frozen at -80oC.

Gene expression studies

Primers for quantitative reverse transcriptase PCR q RT-PCR (Table 4-1) were

synthesized by the Genomics Core Facility at The Pennsylvania State University (University

Park, PA). RNA was isolated from liver tissue using Tri-reagent (Sigma Chemical Co. St. Louis,

MO) according to the manufacturer’s instructions. Isolated RNA was quantified using the

NanoDrop ND-1000 spectrophotometer. cDNA was reverse transcribed and amplified using

SYBR Green PCR Master Mix and an ABI Prism 7000 sequence detection system (Applied

Biosystems, Foster City, CA) . Relative gene expressions were normalized to the house-keeping

genes GAPDH for HF samples and 18S for LF samples. Data was represented by fold change in

Ct values.

Page 110: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

97

Table 4-1. Primer sequences for qPCR analysis of gene expression in liver tissue.

Gene

Abbreviation

Gene Name Forward Primer Reverse Primer

Cat Catalase AGCGACCAGATGAAGCAG

TG

TCCGCTCTCTGTCAAAGTG

TG

Gpx1 Glutathinone

Peroxidase 1

GATGGAGCCCATTCCTGAA

CC

CCCTGTACTTATCCAGGCA

GA

Sod2 Superoxide

dismutase 2

CCGAGGAGAAGTACCACG

AG

GCTTGATAGCCTCCAGCAA

C

Nrf1 Nuclear

Respiratory

factor 1

TATGGCGGAAGTAATGAA

AGACG

CAACGTAAGCTCTGCCTTG

TT

Pgc-1α Peroxisome

proliferator-

activator

receptor

coactivator-1a

ACAGCCGTAGGCCCAGGT

AC

GCCTTTCGTGCTCATAGGC

TT

Sirt3 Sirtuin 3 ATCCCGGACTTCAGATCCC

C

CAACATGAAAAAGGGCTT

GGG

Foxo3a forkhead box O3 ACAAACGGCTCACTCTGTC

CCAG

AGCTCTTGCCAGTTCCCTC

ATTCTG

Nrf2 nuclear factor

(erythroid-

derived 2)-like 2

CAACTCGGCGAAGAAAGA

AACA

AGGATACTGGGGATTCGTC

TG

Tfam Mitochondrial

Transcription

Factor A

CACCCAGATGCAAAACTTT

CAG

CTGCTCCTTTATACTTGCTC

ACAG

Page 111: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

98

Table 4-1 Continued

Tfb1m Mitochondrial

Transcription

Factor B1

ATAGAGCCCAAGATCAAG

CAG

TGTAACAGCCTTCCAGTGC

Tfb2m Mitochondrial

Transcription

Factor B2

ACCAAAACCCATCCCGTC TCTGTAAGGGCTCCAAATG

TG

Complex I

(Ndusf8)

Complex I

(Ndusf8)

GTTCATAGGGTCAGAGGTC

AAG

TCCATTAAGATGTCCTGTG

CG

Complex III

(Uqcrc1)

Complex III

(Uqcrc1)

ATCAAGGCACTGTCCAAG

G

TCATTTTCCTGCATCTCCCG

Mt-I Metallothionein

1

GCTGTCCTCTAAGCGTCAC

C

AGGAGCAGCAGCTCTTCTT

G

Mt-II Metallothionein

2

CAAACCGATCTCTCTCGTC

GAT

AGGAGCAGCAGCTTTTCTT

G

Mtf-1 Metallothionein

transcription

factor 1

CAGTCGGAATGTCCAGAA

ACG

CTGCACGTCACACTCAAAT

GG

Gapdh Glyceraldehyde

3-phosphate

dehydrogenase

TGAAGCAGGCATCTGAGG

G

CGAAGGTGGAAGAGTGGG

AG

18s 18S ribosomal

RNA

GTAACCCGTTGAACCCCAT

T

CCATCCAATCGGTAGTAGC

G

CoxIII Cytochrome c

oxidase subunit

III

CGTGAAGGAACCTACCAA

GG

ATTCCTGTTGGAGGTCAGC

A

Gstz1 Glutathione S

Transferase Zeta

1

TCAGGAGGTACGGGCTGTC TCAGGAGGTACGGGCTGTC

Ucp2 Uncoupling

protein 2

GAGCGGACCACTCCAGCG

TC

TCACCACATCCGTGGGCTG

G

Page 112: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

99

Western blot analysis

The expression of proteins of interest was determined by western blot. Protein was

extracted from liver samples using Tissue Protein Extraction Reagent (TPER) containing 1%

(v/v) phosphatase and protease inhibitor cocktails (Thermo-Scientific, Rockford, IL). After

homogenization and centrifugation the protein concentration in supernatant was determined by

the Bradford assay (Sigma-Aldrich, St.Louis, MO). Lysates were then combined with an equal

volume of laemmli sample buffer (Bio-Rad, Hercules, CA) and denatured at 90°C for 5 min.

Protein samples (75 μg protein) were resolved by SDS-polyacrylamide (15 %) gel

electrophoresis (PAGE) and transferred to a nitrocellulose membrane (Bio-Rad, Hercules, CA).

Membranes were probed with primary antibody for protein of interest (1:1000 dilution) (Cell

Signaling Technology, Danvers, MA) overnight at 4 °C. After incubation with a fluorescent-

conjugated secondary antibody (1:10, 000 dilution) (Li-Cor Co., Lincoln, NE), bands were

visualized using a Licor Odyssey Imaging System (LI-COR Corporate). Band density was

quantified using Odyssey Application Software Version 3.0. Beta actin was used as a protein

loading control.

Antioxidant enzyme activity

Glutathione peroxidase (GPx)

Glutathione peroxidase activity was determined using a commercially available assay kit

from Cayman Chemicals (Ann Arbor, MI). In brief, 50 mg of liver tissue was homogenized in

300 µL homogenization buffer (50 mM Tris-HCl, pH 7.5, 5 mM EDTA, and 1 mM DTT) and

centrifuged at 10, 000 g for 15 mins 4 °C. The supernatant was collected and assayed as per

Page 113: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

100

manufacturer’s instructions. Absorbance was measured at 340 nm and quantification was based

on a standard curve.

Superoxide dismutase (SOD)

Superoxide dismutase activity was determined using a previously reported method [140].

In brief, 100 mg of tissue was homogenized in 500 µL of homogenization buffer (20 mM Hepes,

1 mM EGTA, 210 mM, 70 mM sucrose; pH 7.2) and centrifuged at 1,500 g for 5 mins at 4 °C.

The supernatant was removed diluted 1:50. A volume of 0.1 mL of diluted sample (or standards)

was added to a reaction mixture (2.8 mL) containing: 0.05 M Na2CO3 (pH 10.2), 3mM EDTA,

1.5 mg/mL BSA, 0.75 mM NBT, and 3 mM Xanthine was added. The reaction was initiated with

0.1 mL xanthine oxidase (1:200 dilution) and incubated for 20 minutes at room temperature. The

reaction was terminated by addition of 6 mM CuCl2. The inhibition of NBT reduction was

determined spectrophotometrically at 560 nm.

Catalase Activity

Catalase activity was determined as described by [141]. Briefly, 50 mg of liver tissue

were minced in ice-cold 25 mM KH2P04-NaOH buffer, pH 7.0. The homogenates were then

diluted with buffer as appropriate. Purified catalase or homogenized tissues were incubated with

5.9 M methanol and 4.2 mM hydrogen peroxide in 250 mM KH2P04-NaOH buffer, pH 7.0 in a 96

well plate. The enzymatic reaction was initiated by addition of 100 µL of a catalase-containing

sample. The reaction mixture was incubated with continuous shaking for exactly 20 min at room

temperature. The enzymatic reaction was terminated by addition of 50 µL 7.8 M potassium

hydroxide solution to each tube. Immediately thereafter the tubes were treated with 100 µL each

Page 114: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

101

of 34.2 mM Purpald in 480 mM hydrochloric acid, and a second incubation with continuous

shaking was performed for 10 min at 20°C. To obtain a colored compound, the product of the

reaction between formaldehyde and Purpald was oxidized by adding 50 µL 65.2 mM potassium

periodate in 470 mM potassium hydroxide to each tube. Absorbance was measured at 540 nm and

quantified using a catalase standard curve.

Statistical Analysis

All experiments were repeated twice with at least three replicates for each treatment.

GraphPad Prism 5.0 (San Diego, CA) was used for statistical analysis. In this experiment two

outcomes were focused on; the effect of varying doses of EGCG and the difference between diets

at each dose of EGCG. To determine differences between control (0 mg/kg) and varying doses of

EGCG (250, 500, and 750 mg/kg), one-way ANOVA with Dunnett’s post-test was used to

compare differences in in enzyme activity, protein expression and gene expression for different

doses of EGCG, (* = P < 0.05, ** = P < 0.01, *** = P < 0.001). The Student’s t-test was used to

compare differences between diets (HF and LF) at each dose of EGCG, (# = P < 0.05, ## = P <

0.01, ### = P < 0.001). Data are expressed as mean ± SEM and significance was reached at

P<0.05.

Page 115: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

102

Results

Effect of EGCG on Antioxidant Response

Figure 4-1. Effect of EGCG on hepatic gene expression, protein expression and activity of

superoxide dismutase 2 (Sod2) in male C57BL/6J mice.

Mice were treated with 0, 250, 500 or 750 mg/kg b.w. EGCG once daily for 2 days. Gene

expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet. Protein

expression was normalized to β-actin. Each bar represents the mean of N = 8-18. Error bars

represent SEM. Data were analyzed by one-way ANOVA based on dose with Dunnett’s post-test

(* = P < 0.05, ** = P < 0.01, *** = P < 0.001) and t-test for diets.

Examination of the key components of the hepatic antioxidant defense system showed

that EGCG treatment significantly down regulated mitochondrial superoxide dismutase (Sod2) in

lean mice treated with 750 mg/kg dose of EGCG (Figure 4-1), but no significant changes were

observed in mice fed a HF diet. No changes in protein expression were seen for any of the

treatment groups based on band expression. Quantification of band intensity showed a significant

increase only for mice on a LF diet that received a 750 mg/kg dose of EGCG. EGCG treatment

Page 116: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

103

caused decreases in Sod activity in both lean and obese mice given a 750 mg/kg dose of EGCG

when compared to controls, but the decrease was not significant.

Figure 4-2. Effect of EGCG on hepatic gene expression, protein expression and activity of

catalase in male C57BL/6J mice.

Mice were treated with 0, 250, 500 or 750 mg/kg b.w. EGCG once daily for 2 days. Gene

expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet. Protein

expression was normalized to β-actin. Each bar represents the mean of N = 8-18. Error bars

represent SEM. Data were analyzed by one-way ANOVA based on dose with Dunnett’s post-test

(* = P < 0.05, ** = P < 0.01, *** = P < 0.001) and t-test for diets.

EGCG treatment caused an initial upregulation in catalase gene expression at the 250

mg/kg dose (Figure 4-2) followed by a significant dose dependent down regulation in catalase for

mice on both LF and HF diets. Quantification of protein expression levels showed that catalase

tended to decrease in all treatment groups when compared to controls, but the changes observed

were insignificant. Catalase activity in obese mice was significantly decreased in mice treated

with 250, 500, and 750 when compared to controls. No differences were seen in lean mice

following EGCG treatment.

Page 117: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

104

Figure 4-1. Effect of EGCG on hepatic gene expression, protein expression and activity of

glutathione peroxidase in male C57BL/6J mice.

Mice were treated with 0, 250, 500 or 750 mg/kg b.w. EGCG once daily for 2 days. Gene

expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet. Protein

expression was normalized to β-actin. Each bar represents the mean of N = 8-18. Error bars

represent SEM. Data were analyzed by one-way ANOVA based on dose with Dunnett’s post-test

(* = P < 0.05, ** = P < 0.01, *** = P < 0.001) and t-test for diets (# = P < 0.05, ## = P < 0.01,

### = P < 0.001).

EGCG treatment caused no observable changes in gene expression of glutathione

peroxidase 1 (Gpx1) (Figure 3-10 A), however, significant decreases in the protein expression of

Gpx1 can be seen for both obese and lean mice treated with a 750 mg/kg dose of EGCG. Obese

mice treated with 500 mg/kg EGCG also showed a significant decrease in Gpx1 protein

expression compared to controls (P < 0.05). At the 500 mg/kg dose obese mice had significantly

lower protein expression levels compared to lean mice. A similar pattern is seen at the 750 mg/kg

dose, but the difference was not significant. Total glutathione peroxidase activity showed an

initial but insignificant increase in Gpx1 activity for 250 mg/kg EGCG, followed by a dose

dependent decrease, in both lean and obese mice, but it was not insignificant.

Page 118: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

105

Figure 4-4. Effect of EGCG on hepatic gene expression of glutathione-S-transferase zeta 1 and

peroxiredoxin 3 in male C57BL/6J mice.

Mice were treated with 0, 250, 500 or 750 mg/kg b.w. EGCG once daily for 2 days. Gene

expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet. Each bar

represents the mean of N = 8-18. Error bars represent SEM. Data were analyzed by one-way

ANOVA based on dose with Dunnett’s post-test (* = P < 0.05, ** = P < 0.01, *** = P < 0.001)

and t-test for diets.

Other components of the cell’s antioxidant defense system, glutathione transferase zeta 1

(Gszt1) and peroxiredoxin 3 (Prx3) were measured to assess the effect of varying doses of EGCG.

An increase in Gszt1 gene expression was observed in mice given a 250 mg/kg dose of EGCG

(Figure 4-4), similar to results seen for catalase expression. However, Gszt1 was down regulated

at the 500 and 750 mg/kg doses in both lean and obese mice, and this was significant only for

mice on a LF diet. EGCG treatment caused a dose dependent decrease in expression of Prx3 in

mice on HF diet. This down regulation was significant for mice given a 750 mg/kg dose. A down

regulation of Prx3 was also seen in lean mice for both the 250 and 500 mg/kg doses of EGCG,

but these changes were not significant when compared to the controls. Interestingly, an increase

in Prx3 expression was seen for mice on LF diet given a 750 mg/kg dose, but this change was not

significant when compared to controls.

Page 119: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

106

Figure 4-5. Changes in metallothionein expression in male C57BL/6J mice.

Gene expression of metallothionein transcription factor 1 (MTF-1), metallothionein I (MT-1), and

metallothionein II (MT-2) and protein expression and quantification of MT-1 and MT-2 (MT I/II)

were measured. Mice were treated with 0, 250, 500 or 750 mg/kg b.w. EGCG once daily for 2

days. Gene expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF

diet. Protein expression was normalized to β-actin. Each bar represents the mean of N = 8-18.

Error bars represent SEM. Data were analyzed by one-way ANOVA based on dose with

Dunnett’s post-test (* = P < 0.05, ** = P < 0.01, *** = P < 0.001) and t-test for diets (# = P <

0.05, ## = P < 0.01, ### = P < 0.001).

Metallothioneins are low molecular weight sulphur-rich proteins that play a role in heavy

metal detoxification and metal homeostasis. EGCG treatment caused a significant upregulation in

gene expression of metallothionein transcription factor MTF-1 for both lean and obese mice

(Figure 4-5). For obese mice the upregulation appears to occur in a dose dependent manner, with

mice given a 750 mg/kg dose having the most significant upregulation (P < 0.05) when compared

to controls. Mice on a LF diet had the most significant upregulation of MTF-1 when they

Page 120: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

107

received 500 mg/kg EGCG. In general, lean mice exhibited higher expression levels of MTF-1

when compared to obese mice, with this difference being significantly highest in mice given 500

mg/kg EGCG.

The expression of MT-I was upregulated significantly by EGCG treatment at the 250 and

500 mg/kg doses for obese mice when compared to controls. Similar results were seen for lean

mice, but an increase in MT-I expression was most significant for mice on a 500 mg/kg EGCG.

Although there was upregulation of MT-I at the 750 mg/kg dose for both diets, this change was

not significant when compared to the controls. No significant differences were seen between the

diets at the various doses of EGCG.

The effect of EGCG on MT-II was similar to that seen for MTF-1. EGCG caused a dose

dependent upregulation of MT-II gene expression in both diet groups which was significant in

mice given 500 and 750 mg/kg EGCG. Expression of MT-II in lean mice is higher than obese

mice, but the difference between diets at all the doses was not significant. Protein expression of

metallothioneins supported the trends seen in MTF-1 and MT-II gene expression, for mice on LF

diet. EGCG treatment caused an increase in the expression of MTs which was significant in mice

given a 750 mg/kg dose (P < 0.05). Obese mice had significantly lower expression of MTs when

compared to controls at the 250 and 500 mg/kg dose. A significant difference in the protein

expression of MTs can also be seen between diets. Lean mice expressed significantly higher

protein levels of MTs when compared to HF fed mice when given 500 and 750 mg/kg EGCG (P

< 0.01).

Page 121: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

108

Effect of EGCG on regulators of antioxidant genes

Figure 4-6. Effect of EGCG on hepatic gene and protein expression of Sirtuin 3 (Sirt3) in male

C57BL/6J mice.

Mice were treated with 0, 250, 500 or 750 mg/kg b.w. EGCG once daily for 2 days. Gene

expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet. Protein

expression was normalized to β-actin. Each bar represents the mean of N = 8-18. Error bars

represent SEM. Data were analyzed by one-way ANOVA based on dose with Dunnett’s post-test

(* = P < 0.05, ** = P < 0.01, *** = P < 0.001) and t-test for diets.

Hepatic mRNA expression of the mitochondrial NAD-dependent deacetylase, sirtuin-3

(Sirt3), was significantly down-regulated in mice treated with 750 mg/kg dose of EGCG when

compared to controls (Figure 4-6). Although a decrease was seen at the 500 mg/kg dose for both

diets, this down regulation was not significant. Interestingly, an upregulation was observed at the

250 mg/kg dose of EGCG in both lean and obese mice. This increase in Sirt3 expression was

higher in obese mice compared to lean mice, but the difference was not significant. Western blot

analysis showed a dose dependent decrease in protein expression of Sirt3, which was significant

in obese mice. Quantification of the bands corroborated what we saw in the images. In mice

treated with 500 and 750 mg/kg EGCG, obese mice showed lower protein expression when

compared to lean mice, but the differences were not significant.

Page 122: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

109

Figure 4-7. Effect of EGCG on the hepatic gene expression of forkhead box O3 and nuclear

factor (erythroid-derived 2)-like 2 (Nrf 2) in male C57BL/6J mice.

Mice were treated with 0, 250, 500 or 750 mg/kg b.w. EGCG once daily for 2 days. Gene

expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet. Each bar

represents the mean of N = 8-18. Error bars represent SEM. Data were analyzed by one-way

ANOVA based on dose with Dunnett’s post-test (* = P < 0.05, ** = P < 0.01, *** = P < 0.001)

and t-test for diets (# = P < 0.05, ## = P < 0.01, ### = P < 0.001).

Other mitochondrial antioxidant regulators assessed were Foxo3a and Nrf2 (Figure 4-7).

Foxo3a mRNA expression was significantly dose dependently reduced by EGCG in both lean

and obese mice. No significant differences in Foxo3a expression was observed between HF and

LF mice at the different doses of EGCG, except in mice given a 500 mg/kg dose. A decrease in

Nrf2 expression can be seen in lean and obese treated with 750 mg/kg EGCG, but the differences

were not significant when compared to controls. No differences were observed in mice treated

with other doses of EGCG when compared to controls.

Page 123: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

110

Effect of EGCG on mitochondrial biogenesis

Figure 4-8. Effect of EGCG on the hepatic gene expression of nuclear respiratory factor 1 (Nrf1)

and peroxisome proliferator-activator receptor coactivator-1a (Pgc-1α) in male C57BL/6J mice.

Mice were treated with 0, 250, 500 or 750 mg/kg b.w. EGCG once daily for 2 days. Gene

expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet. Each bar

represents the mean of N = 8-18. Error bars represent SEM. Data were analyzed by one-way

ANOVA based on dose with Dunnett’s post-test (* = P < 0.05, ** = P < 0.01, *** = P < 0.001)

and t-test for diets.

Nuclear respiratory factor 1 is a key transcription factor in mitochondrial biogenesis. It

regulates mitochondrial transcription factor A. Hepatic mRNA levels of Nrf1 followed an

increasing trend as a function of EGCG dose up to 500 mg/kg, but decreased at 750 mg/kg dose

in both obese and lean mice (Figure 4-8). There was no significant difference in Nrf1 expression

between lean and obese mice. EGCG treatment caused a significant dose dependent decrease in

Pgc-1α expression in both lean and obese mice treated with 500 and 750 mg/kg EGCG.

Mitochondrial DNA copy number is an indicator of mitochondrial biogenesis. Obese

mice treated with EGCG showed a decreasing trend in mtDNA copy number from 0 to 500

mg/kg, but the change was not significant. Lean mice treated with 500 mg/kg EGCG showed a

significant decrease in mtDNA copy number when compared to controls. No difference in Mt

copy was seen at the other doses of EGCG when comparing lean and obese mice.

Page 124: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

111

0 250 500 7500.0

0.5

1.0

1.5HF

LF

Mt Copy Number

*

*

Dose (mg/kg)

Fo

ld C

han

ge

Figure 4-9. Effect of EGCG on mitochondrial copy number in male C57BL/6J mice.

Gene expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet.

Each bar represents the mean of N= 8-18. Error bars represent SEM. Data were analyzed by one-

way ANOVA based on dose with Dunnett’s post-test (* = P < 0.05, ** = P < 0.01, *** = P <

0.001) and t-test for diets.

Effect of EGCG on factor involved in mitochondrial function

Mitochondrial transcription factor A (Tfam), is a transcription factor that initiates

mitochondrial transcription during replication and is a regulator of MtDNA copy number. Tfam

was significantly down regulated in lean and obese mice given 750 mg/kg EGCG when compared

to controls (Figure 4-10). At the 250 and 500 mg/kg, EGCG appeared to increase the expression

of Tfam, but this change was not significant.

Page 125: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

112

Figure 4-10. Effect of EGCG on mitochondrial transcription factors A, (Tfam), B1 (Tfb1m) and

B2 (Tfb2m) in male C57BL/6J mice.

Gene expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet.

Each bar represents the mean of N= 8-18. Error bars represent SEM. Data were analyzed by one-

way ANOVA based on dose with Dunnett’s post-test (* = P < 0.05, ** = P < 0.01, *** = P <

0.001) and t-test for diets.

Mitochondrial transcription factors B1 and B2 (Figure 4-10) are dimethyltransferases that

methylate mitochondrial ribosomal RNA. They are also involved in mitochondrial transcription,

which is important for mitochondrial gene expression. EGCG treatment caused an increase in

Tfb1m mRNA levels at the 250 mg/kg dose level, although the effect was not statistically

significant. At the higher dose levels, EGCG reduced the expression of Tfb1m.The effect reached

statistical significance at 750 mg/kg. Tfb2m showed a similar expression pattern to that seen for

Tfb1m and Tfam. The down regulation in Tfb2m was significant at both the 500 and 750 mg/kg

doses of EGCG for obese mice (P < 0.01 and P < 0.001), and at 750 mg/kg EGCG for lean mice

(P < 0.05). Upregulation of Tfb2m by EGCG was also observed at the 250 mg/kg dose. No

difference in Tfb2m expression between diets was seen at the different doses of EGCG, except for

the 500 mg/kg dose, where LF mice had higher expression levels. This; however, was not

significant.

Page 126: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

113

Figure 4-11. Effect of EGCG on the hepatic gene expression of mitochondrial Complex I and

mitochondrial complex III in male C57BL/6J mice.

Gene expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet.

Each bar represents the mean of N = 8-18. Error bars represent SEM. Data were analyzed by one-

way ANOVA based on dose with Dunnett’s post-test (* = P < 0.05, ** = P < 0.01, *** = P <

0.001) and t-test for diets (# = P < 0.05, ## = P < 0.01, ### = P < 0.001).

Mitochondrial complex I and III are key parts of the mitochondrial electron transport

chain, which are encoded in MtDNA and are important sites of electron leakage. The subunits

analyzed were shown to be affected by EGCG in much the same way as other factors involved in

mitochondrial replication, transcription and function. Gene expression of Ndusf 8, a subunit of

mitochondrial complex I was significantly down regulated by EGCG treatment at the 500 and 750

mg/kg doses (Figure 4-11). At the 750 mg/kg dose, obese mice showed a significantly higher

expression level of Ndusf 8 when compared to lean mice at the 750 mg/kg dose of EGCG (P <

0.05). Like Ndusf 8, Uqcrc 1, a subunit of mitochondrial complex III also showed a decrease in

gene expression. For lean mice, down regulation of Uqcrc 1 was significant at 750 mg/kg EGCG,

and at 500 mg/kg for obese mice. Additionally, obese mice also exhibited significantly higher

levels of Uqcrc 1 expression when given 750 mg/kg EGCG, compared to lean mice (P < 0.05).

Page 127: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

114

0 250 500 7500

2

4

6

8

10HF

LF

Ucp 2

** *

Dose (mg/kg)

Fo

ld C

han

ge

#

#

Figure 4-12. Effect of EGCG on mitochondrial uncoupling protein 2 (Ucp2) in male C57BL/6J

mice.

Gene expression was normalized to GAPDH for mice on HF diet and 18S for mice on LF diet.

Each bar represents the mean of N= 8-18. Error bars represent SEM. Data were analyzed by one-

way ANOVA based on dose with Dunnett’s post-test (* = P < 0.05, ** = P < 0.01, *** = P <

0.001) and t-test for diets (# = P < 0.05, ## = P < 0.01, ### = P < 0.001).

The effects of EGCG treatment on gene expression of mitochondrial uncoupling protein 2

(Ucp2), which separates oxidative phosphorylation from ATP synthesis in an attempt to reduce

ROS production, showed a does dependent increase in lean and obese mice (Figure 4-12).

Upregulation of Ucp2 was most significant for obese mice that received 500 and 750 mg/kg

EGCG. Additionally, obese mice showed significantly higher expression of Ucp 2 when given

250 and 500 mg/kg EGCG, compared to lean mice.

Page 128: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

115

Discussion

In the previous chapter, we examined the effect of obesity on EGCG induced liver

toxicity. We saw increases in markers of hepatotoxicity and oxidative stress. As stated previously,

work coming out of our lab showed that EGCG induces mitochondrial ROS and subsequent

dysfunction in cancer cells (Tao et al 2014). This suggests that the mitochondria may play a

significant role in EGCG toxicity. In the current study we investigated the dose dependent effect

of EGCG treatment on mitochondrial antioxidant responsive genes, mitochondrial biogenesis and

mitochondrial function in C57BL/6J mice.

Figure 4-11. Scheme of mitochondrial antioxidant pathways and gene regulators.

Page 129: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

116

Figure 4-13 shows the location of antioxidant genes and the regulatory transcription

factors in the mitochondria. We examined the changes in mRNA, protein expression, and/or

enzyme activity of these factors. Superoxide dismutase along with catalase and glutathione

peroxidase is essentially the cell’s first line of defense against reactive oxygen species. In this

study we determined the effect of EGCG treatment on gene expression, protein expression, and

enzyme activity of these three enzymes; as well as the effect of diet. Mitochondrial superoxide

dismutase (Sod2) has been shown to be down regulated in cases of hepatotoxicity or extreme

oxidative stress. Compounds such as cadmium and acetaminophen, known to cause

hepatotoxicity have been shown to cause a decrease in Sod2 expression [142], [143] and several

other key antioxidant genes were shown to be significantly down regulated in cancer cells treated

with EGCG, in which EGCG caused cytotoxicity (Tao et al. 2014).

The results of our study are consistent with the findings of these previous studies. Gene

expression of Sod2 decreased in a dose dependent manner for mice in both diet groups. No

difference in EGCG toxicity was seen for mice on either diet, from 0-500 mg/kg EGCG. At the

750 mg/kg dose of EGCG, a dose shown to cause hepatotoxicity, mice on LF diet showed

significantly lower expression of Sod2 when compared to obese mice. Protein expression

revealed something different from the gene expression data. For mice on HF diet there were no

significant changes in expression for any of the doses of EGCG. LF mice on the other hand

showed an increase in protein expression from 250 – 750 mg/kg EGCG. This increase in Sod2

expression we observed, maybe an effort by the cells to compensate for the increased oxidative

environment. Superoxide dismutase activity was suppressed most significantly at the 750 mg/kg

dose of EGCG for both diet groups. This could be as a result of changes to regulating factor Sirt3,

which regulates Sod activity, where a decrease in Sirt3 expression was observed in mice treated

with 750 mg/kg EGCG. The effect of EGCG on catalase gene expression was consistent with that

Page 130: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

117

of Sod2, except for an increase at the 250 mg/kg dose. This increase was significant for obese

mice and could be an adaptive mechanism by the cell to a change in the oxidative environment.

As the dose of EGCG increases there is a significant down regulation of catalase for mice in both

diet groups. This is consistent with data reported by Tao at el, where a down regulation of

catalase was observed in cancer cells treated with EGCG [18]. Other hepatotoxins such as furan

and methapyrilene have also been shown to down regulate catalase expression [144]. Protein

expression of catalase though not significant supported gene expression data and is consistent

with studies done using other hepatotoxins, namely acetaminophen and amiodarone, and showed

that EGCG causes a decrease in catalase expression [145].

Glutathione peroxidase 1 (Gpx1), like catalase, is a peroxide scavenger and is the most

abundant isozyme in the Gpx family [146]. In determining the effect of EGCG treatment on Gpx1

gene expression, protein expression and activity, we found no effect of EGCG at the gene level,

as there were no changes in expression levels from dose to dose. This contrasts with previous

work by Tao et al who had shown a down regulation of Gpx1 by EGCG treatment in cancer cells.

Additionally, acetaminophen has been shown to downregulate Gpx1 [144]. As was not observed

in gene expression, we found that protein expression of Gpx1 showed a significant decrease. This

data is consistent with other studies on the impact of hepatotoxins on Gpx1 protein expression.

Yamamoto et al., showed that acetaminophen, amiodarone and tetracycline all cause

hepatotoxicity and treatment with these compounds cause a decrease in Gpx1 expression [145].

We also observed that at the higher doses of EGCG, expression of Gpx1 was significantly lower

in obese mice compared to mice on LF diet. As was previously discussed, obesity causes

oxidative stress, which results in depleted levels of reduced glutathione (GSH). Gpx1 forms part

of the glutathione redox system where it used GSH to detoxify peroxides [147]. In our earlier

results we saw that the levels of GSH in obese mice were significantly lower than LF mice;

therefore this depletion of GSH can explain why Gpx1 expression is lower in HF mice. Further,

Page 131: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

118

studies have shown that obesity causes reduced Gpx1 expression [148]. A measure of GPx

activity showed a decrease in enzyme activity. This decrease however, was not significant and

there was no clear difference between diet groups.

Another component of the glutathione system is glutathione transferase which catalyzes

the conjugation of GSH to xenobiotic compounds during phase II metabolism (Pickett 1989). We

measured the gene expression of glutathione transferase zeta 1 (Gstz1) in this study, as it was

shown to be down regulated by EGCG treatment in cancer cells [18]. We too saw a down

regulation of Gstz1 by EGCG treatment in this study, which confirms an increase in oxidative

stress.

Mitochondrial localized peroxiredoxin 3 (Prx3) is a thiol containing endogenous

antioxidant that scavenges up to 90 % of the hydrogen peroxide produced in the mitochondria

[146]. Peroxiredoxins are generally overexpressed under oxidative stress conditions [149], which

makes them efficient peroxide scavengers; but they can become hyperoxidized under prolonged

exposure to hydrogen peroxide. Hyperoxidation can inactivate Prxs and lead to the accumulation

of the oxidized form [150]. Prx3 has been shown to be hyperoxidized more slowly than other

peroxiredoxins, but can be over oxidized under oxidative stress conditions [151], [152]. Our

results show a downregulation of Prx3 with increasing doses of EGCG. EGCG treatment was

shown to cause prolonged oxidative stress in cells, and as a result hyperoxidation could be a

possible explanation for the observed down regulation. Since Prx3 is regulated at the gene

expression level, another possible explanation for Prx3 gene down regulation could be

inactivation or down regulation of its transcription factors; Foxo3a, Nrf2 and Pgc1α, which we

observed to be down regulated by EGCG [153]. There were no differential effects of diet on

EGCG mediated hepatotoxicity.

Metallothioneins (MTs) are heavy metal binding proteins that traditionally have been

associated with detoxification of heavy metals [154]. Recent studies have shown that

Page 132: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

119

metallothioneins have other biological functions not related to metal detoxification and are also

induced by oxidative stress. MTs play a role in protecting the cell from ROS by scavenging

oxygen free radicals [154], [155]. MT-I and MT-II are the inducible isoforms of MTs and they

are regulated by the transcription factor MTF-1 in response to oxidative stress [156], [157]. MTs

complement the GSH/GSSG redox cycle and are significantly upregulated in instances of GSH

depletion and superoxide dismutase down-regulation [155]. We investigated gene and protein

expression of MTs to determine the effect of EGCG treatment in mice fed HF and LF diets. MTF-

1 increased significantly for both diet groups from 0 – 750 mg/kg EGCG. This increase was dose

dependent for mice on a HF diet and supports previous data that EGCG induces oxidative stress.

For mice on LF diet the upregulation of MTF-1 was highest a 500 and 750 mg/kg doses, again

supporting our previous findings. MT-I expression increased with increasing doses of EGCG,

except at the 750 mg/kg dose, where a decrease in expression was observed and not a continuous

increase as would be expected. Down regulation of MT-I was seen to occur in hepatocellular

carcinoma cells lines when the MT promoter was extensively methylated [158]. Methylation of

the MT promoter could provide an explanation for the down regulation of MT-I seen at the 750

mg/kg dose in both diets. MT-II expression followed a similar trend to that of MTF-1, where

EGCG caused a significant dose dependent increase in expression for both diets. Protein

expression of MTs supported the data seen for both MTF-1 and MT-II. Other hepatotoxicants

such as acetaminophen have also been shown to increase metallothionein expression [159]. All

of these data suggest that EGCG causes a dose dependent increase in oxidative stress, which

correlates to a dose dependent increase in MT gene and protein expression and is consistent with

previous findings and studies.

Analysis of the metallothionein data showed that mice on a high fat diet consistently

expressed lower levels of MTs at the different doses of EGCG, when compared to LF fed mice.

In a study conducted by Waller-Evans et al [160]an analysis of gene expression responses to HF

Page 133: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

120

feeding in C57BL/6J showed a downregulation of genes encoding for MT-I and MT-II. This

response was specific to this mouse strain. These results can help explain the differences between

diet groups that were observed in this study [160].

Mitochondrial NAD-dependent deacetylase sirtuin-3 (Sirt3) has been implicated in

regulation of mitochondrial function and antioxidant activity [161]. Sirt3 is activated by Pgc-1α

and through their combined efforts they modulate oxidative stress in the cell [129], [162], [163].

Sirt3 has been shown to regulate oxidative stress by activating mitochondrial antioxidant enzymes

such as Sod2 and Gpx1 and possibly Prx 3; all of which showed significant down regulation by

EGCG treatment [164]. Our data shows an initial up regulation of Sirt3, which can be attributed

to the cell adapting to a change in the oxidative environment. This was followed by a significant

down regulation of Sirt3 gene expression by EGCG treatment. Based on the fact that Sirt3 is

regulated by PGC-1α, which was also down regulated by EGCG, the argument can be made that

this contributed to the down regulation of Sirt3. Protein expression of Sirt3 from western blot

analysis showed a significant decrease in expression in both lean and obese mice. This data

supports what was seen at the gene level and in previous studies. For example, a study by Tao et

al., showed that Sirt3 protein expression was down regulated in EGCG mediated cytotoxicity in

cancer cells [165]. Cadmium, a known hepatotoxin was shown to decrease Sirt3 protein, which in

turn decreased Sod2 activity [166]. Since EGCG has been shown to cause liver toxicity at the

higher doses, this provides a possible explanation for the decrease in gene and protein expression

seen for Sirt3. The data also revealed that obese mice had lower protein expression of Sirt3 when

compared to lean mice, but the difference was not significant. One possible explanation for this is

that decreased levels of Sirt3 have been associated with a high fat diet [167]. This effect

combined with the toxic effect of EGCG may have made obese mice more susceptible, thus they

expressed lower levels of Sirt3.

Page 134: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

121

Forkhead box O3 transcription factor A (Foxo3a) is a transcription factor that forms part

of the cell’s antioxidant defense system by regulating antioxidant genes [168]. Foxo3a can

reduce the level of cellular oxidative stress by directly upregulating mRNA and protein levels of

Sod2, catalase and peroxiredoxin 3 [168], [169]. Foxo3a like Nrf1 requires coactivator Pgc-1α to

regulate oxidative stress [168]. Similar to the data seen for Pgc-1α a significant down regulation

of Foxo3a was seen in lean and obese mice treated with EGCG. This data supports what has been

seen based on the effect of EGCG on other transcription factors. Additionally, Foxo3a has been

shown to be regulated by Sirt3 and this provides an explanation to support the down regulation of

Foxo3a seen [164]. In obese mice given 500 mg/kg EGCG, a significantly higher expression of

Foxo3a was seen in lean mice fed the same dose. One possible explanation for this is the fact that

Foxo3a has been shown to be upregulated by a high fat diet [170]. In this case the effects of

EGCG treatment were seen, but compared to LF mice the effects were not as severe in obese

mice probably due to them having higher initial levels of Foxo3a.

Another important mitochondrial regulator is transcription factor nuclear factor

(erythroid-derived 2)-like 2 (Nrf2). Nrf2 has been implicated in the cell’s antioxidant defense

system with its ability to activate genes involved in detoxification of ROS through the antioxidant

response element (ARE) and is essential in cellular response to oxidative stress [171], [172].

Nrf2, like key the antioxidant enzyme that it regulates, glutathione-S-transferases, have been

shown to be down regulated in aggressive prostate cancer tissue [173]. Our analysis of Nrf2

expression after treatment in obese and lean mice showed that there was an upregulation from the

0 – 500 mg/kg dose of EGCG in both diet groups, followed by a down regulation at the toxic 750

mg/kg dose, also in both diet groups. Our results are consistent with data from previous studies.

In a study conducted by Wang et al., EGCG was shown to upregulate Nrf2 at non-lethal doses,

but down regulated Nrf2, as well as superoxide dismutase, catalase and glutathione peroxidase at

a toxic dose [174].

Page 135: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

122

We analyzed nuclear respiratory factor 1 (Nrf1) expression. Nrf1 is a transcription factor

located in the nucleus, and is involved in nuclear regulation of mitochondrial transcription,

translation and replication [175], [176]. Nrf1 along with co-activator Pgc-1α is an important

regulator of mitochondrial biogenesis [177], [178]. Our data showed an upregulation of Nrf1

from 0 mg/kg to 500 mg/kg which was significant at 500 mg/kg EGCG. Studies have shown that

Nrf1 is induced by compounds that cause damage to mitochondria in liver tissue, through ROS

generation, GSH depletion, and mtDNA damage [179]. EGCG has been shown to generate ROS

and deplete GSH [18], [105], so it is reasonable to assume that like these other compounds,

EGCG can induce Nrf1. As the dose of EGCG increased to 750 mg/kg, a known toxic dose, a

decrease in Nrf1 expression was seen in lean and obese mice. This could be explained by an

increase in cellular apoptosis and tissue necrosis seen at that dose. Also, Nrf1 is regulated by Pgc-

1α, and this too was down regulated, which could cause a down regulation of Nrf1. Additionally,

since Nrf1 is located in the nucleus, where an increase on DNA strand breaks were seen for 750

mg/kg EGCG, this could also explain the down regulation of Nrf1 seen here.

As was previously stated, Pgc-1α is a regulator of mitochondrial biogenesis [180][91].

Pgc-1α has also been shown to be a powerful regulator of ROS removal by increasing the

expression of numerous ROS-detoxifying enzymes thereby being effective in the cell’s

antioxidant capabilities [177]. These antioxidants include Sod2, Gpx and Prx3 just to name a few;

all of which were measured in our study and shown to be significantly down regulated. We saw a

significant decrease in Pgc-1α gene expression at 500 and 750 mg/kg EGCG in lean and obese

mice. Other hepatotoxins such as CCL4 have been shown to depress Pgc-1α in rats in which it

induced hepatic injury [181]. Pgc-1α has been shown to be associated with adaptive

thermogenesis, and is elevated in response to oxidative stress and under obese conditions [182],

[183]. At the 750 mg/kg dose of EGCG, though not significant, obese mice had even lower

expression levels than lean mice.

Page 136: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

123

The data obtained so far, based on the changes seen in expression of antioxidant

enzymes, regulators of antioxidant genes and mitochondrial biogenesis indicates that EGCG

induced changes in genes and proteins associated with or localized in the mitochondria. This is

consistent with previous work done by Tao et al., where EGCG was shown to induce

mitochondrial ROS and subsequent mitochondrial dysfunction [18]. We investigated the effect of

EGCG genes involved in mitochondrial function.

Mitochondrial DNA copy number is a marker of mitochondrial biogenesis and was

determined as the ratio between mitochondrial encoded gene, cytochrome c oxidase subunit 3

(CoxIII) and nuclear endogenous gene 18S [184], [185]. Mitochondrial DNA is highly susceptible

to oxidative damage as the mitochondria are an endogenous source of ROS in the cell due to the

electron transport chain (ETC) [186]. A decrease in mtDNA copy number is associated with

many pathologies such as liver disease and type II diabetes among others. Mitochondrial

dysfunction reduces the contents of mitochondria, which is expressed as a decreased mtDNA

copy number [185]. Our results show that mtDNA copy number decreased in all treatment groups

compared to control. This data is consistent with previous studies. Pieters et al., showed that

polycyclic aromatic hydrocarbons (PAHs), which are known to induce oxidative stress, were

shown to decrease mtDNA copy number, further supporting our findings, as EGCG also induces

mitochondrial oxidative stress [187]. Aging, which increases oxidative stress, also decreased

mtDNA copy in rat liver tissue [188]. The decrease in mtDNA copy number observed here can

also be attributed to the changes seen in transcriptional co-activator Pgc-1α. Pgc-1α, along with

nuclear respiratory factor 1 (Nrf1) and mitochondrial transcription factor A (Tfam), regulate

mitochondria biogenesis [177], [189]. EGCG treatment caused significant down regulation of

Pgc-1α, which could explain the observed decrease in mtDNA copy number. Many studies have

also shown that obesity decreases mtDNA copy number. However, no significant differences as a

direct consequence of obesity were observed in this study [190].

Page 137: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

124

We measured mitochondrial transcription factor A (Tfam) based on changes seen in Pgc-

1α, Nrf1 and mtDNA copy number. Tfam is a key activator of mitochondrial transcription as well

as a participant in mitochondrial genome replication and consequently mtDNA copy number

[191], [192]. Tfam is regulated by Nrf1 in collaboration with Pgc-1α, and is upregulated by

oxidative stress [189], [193], [194]. Similar to data obtained for Nrf1, we saw an initial

upregulation of Tfam (0 – 500 mg/kg EGCG) followed by a significant down regulation at the

750 mg/kg dose. The data obtained can be explained by the fact that Tfam is regulated by Nrf1

and the changes seen are consistent with those of Nrf1. It is also regulated by Pgc-1α, which was

significantly down regulated by EGCG. Obese mice given 250 mg/kg EGCG expressed higher

levels of Tfam then lean mice at that dose. This could be explained by the fact that as previously

indicated, Tfam is induced by oxidative stress, and also as previously stated, higher levels of

oxidative stress are associated with obese mice compared to lean mice, so obese mice can

potentially express higher levels of Tfam than lean mice.

In addition to Tfam we investigated other mitochondrial transcription factors;

mitochondrial transcription factor B1 (Tfb1m), and mitochondrial transcription factor B2 (Tfb2m).

Tfb1m and Tfb2m are dimethyltransferases encoded in the nucleus and essential for mitochondrial

DNA replication in the presence of TFAM and mitochondrial RNA polymerase (POLYRMT)

[192], [195]. Both transcription factors are regulated by Nrf1 and Pgc-1α [178]. Tfb1m has been

shown to regulate mitochondrial translation and loss of Tfb1m can result in mitochondrial

dysfunction [196]–[198]. Analysis of Tfb1m data showed an upregulation of the transcription

factor in lean and obese mice given 250 mg/kg EGCG. At this dose, though not significant obese

mice expressed higher levels of Tfb1m compared to lean mice. Like Tfam, Tfb1m is induced by

oxidative stress and obesity has been shown to facilitate an oxidative environment, so this could

explain the differences seen between the diets at this dose of EGCG. A down regulation of Tfb1m

Page 138: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

125

was observed at the other doses, and this could be as a result of the down regulation of

transcription factor Nrf1 and co-activator Pgc-1α, which are regulators of Tfb1m.

Whereas Tfb1m regulates mitochondrial translation, Tfb2m has been shown to regulate

mitochondrial transcription, making it have a more direct effect on mtDNA copy number [199]–

[201]. Gene expression of Tfb2m is consistent with data seen for Tfb1m and Tfb2m, and the

expression pattern can be explained in the same way. Obese mice expressed lower levels of

Tfb2m compared to lean mice, although this was not significant, one possible explanation for this

is that Tfb2m, because of its role in mitochondrial transcription has been associated with

metabolic functions, and shown to be essential [199]. Obesity is a metabolic disorder that has

been shown to increase oxidative stress and reduce mtDNA copy number [202], which could

explain why obese mice had lower expression of Tfb2m.

Complex I and III of the mitochondrial electron transport chain (ETC) are major sites of

ROS generation in the cell. They are exclusively encoded by mitochondrial DNA and as such are

makers for mtDNA oxidative damage [128], [131], [186]. Our study showed significant down

regulation of both complexes by EGCG treatment. This data is consistent with other studies that

show ROS generating compound, capsaicin decreases complex I and complex III [203]. This data

also supports our previous findings of the effect of EGCG on genes involved in mitochondrial

replication, transcription and function. The changes seen in complex I and III expression can be

attributed to down regulation of transcription factor Nrf1, which has been shown to regulate these

complexes, and was down regulated in the study [177]. Additionally, both complex I and III are

activated by Sirt3 [164]. In our study Sirt3 was also significantly down regulated by EGCG

treatment, and this could account for the down regulation seen in the complexes.

Mitochondrial uncoupling proteins separate oxidative phosphorylation from ATP

synthesis with energy dissipated as heat. UCPs facilitate the transfer of anions from the inner to

the outer mitochondrial membrane and the return transfer of protons from the outer to the inner

Page 139: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

126

mitochondrial membrane. Ucp2 has been implicated in the control of mitochondria-derived

reactive oxygen species. The uncoupling activity of Ucp2 increases the respiratory rate and thus

lowers the mitochondrial production of reactive oxygen species (ROS) [204]. In fact, increased

Ucp2 expression has been shown in tumor cells and have been associated with lower ROS levels;

indicating that cancer cells have adapted the over expression of Ucp2 as a mechanism to maintain

homeostasis, thus preventing mitochondrial superoxide generation and subsequent apoptosis

[205], [206]. Ucp2 has been shown to be highly inducible under conditions of oxidative stress

generated by agents such as H2O2, lipopolysaccharide, TNFα, free fatty acids, irradiation, as well

as high-fat diet both in vitro and in vivo. This suggests that a major function of Ucp2 is to lessen

mitochondrial production of ROS and like in cancer cells this too can be an adaptive response to

oxidative stress [143].

Ucp2 has also been shown to be upregulated by ROS in other tissue as well. Tsuboyama-

Kasaoka et al saw a significant upregulation of liver Ucp2 in C57BL/6J mice fed a high safflower

and a high fish oil diet for 5 months, compared to the high carbohydrate fed mice [207]. Our

results showed a dose dependent increase in Ucp2 in both lean and obese mice. These data are

consistent with previous studies showing that Ucp2 is expressed under conditions of oxidative

stress. EGCG treatment has been shown to increase Ucp2 expression in obese mice [30], [31].

Additionally, significant differences in Ucp2 gene expression levels were observed between lean

and mice treated with 250 and 500 mg/kg EGCG. Fulop et al., showed that Ucp2 is highly

induced in hepatocytes under obese conditions [208]. Rashid et al., also showed increased

expression of Ucp2 in 2 mice models of fatty liver [209]. Evaluation of Ucp2 expression in livers

of male obese mice showed an increase in both gene and protein expression of Ucp2, compared to

their lean counterparts where no expression was observed. The study also showed that the

increased Ucp2 expression corresponded with depleted ATP levels, which increased hepatocyte

sensitivity to necrosis [210].

Page 140: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

127

Based on these results, in can be seen that EGCG treatment lead to significant decreases

antioxidant response in liver tissue of male C57BL/6J. Changes were seen in gene, protein and

activity levels. A corresponding decrease in regulators of antioxidant genes was also observed

due to EGCG treatment. EGCG treatment also significantly upregulated markers of oxidative

stress which coincided with reduced antioxidant activity. EGCG resulted in a decrease in

mitochondrial biogenesis as evidenced by decreased mtDNA copy and gene expression of

mitochondrial transcription factors. Overall EGCG was able to induce mitochondrial dysfunction

by inhibiting the gene, protein and/or activity of many key targets.

Page 141: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

Conclusions and Recommendation for Future Work

Conclusions

Here, I tested the hypothesis that dietary pretreatment with EGCG markedly decreases

EGCG bioavailability and can potentially mitigate the toxic potential of EGCG, and that ORFLD

increases susceptibility to high dose EGCG-mediated hepatotoxicity. In addition, the underlying

mechanism by which EGCG mediates hepatotoxicity is through induction of mitochondrial

oxidative stress and increased mitochondrial dysfunction.

To determine the effect of dietary pretreatment with EGCG on the bioavailability and

hepatotoxicity of subsequent oral bolus dosing with EGCG in mice.

I found that dietary pretreatment with EGCG can significantly increase the gene

expression of antioxidant response genes in liver tissue, as well as reduce makers of liver injury

and oxidative stress. The decrease in oxidative stress and subsequent liver injury can be

interpreted as direct result of the increased antioxidant enzyme activity, which was able to

mitigate the toxic potential of high dose EGCG. My results also showed that dietary

pretreatment with EGCG was able to reduce the bioavailability of EGCG, which corresponded to

decreased plasma and tissue concentrations of EGCG; and consequently reduced the hepatotoxic

potential of subsequent high dose oral bolus EGCG. Essentially dietary pre-treatment was able to

exert a protective effect on subsequent high dose oral bolus EGCG. In terms of translation to

human consumption patterns, the results are analogous to comparing exposure of a non-tea

Page 142: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

129

consumer (NP) to a regular tea consumer (EP), and suggest that regular consumers of green tea

beverage may be at decreased risk of hepatotoxicity associated with use of green tea based dietary

supplements.

To determine the effect of pre-existing obesity and ORFLD on EGCG-mediated

hepatotoxicity in mice.

Obesity and obesity related fatty liver disease have been shown to adversely affect the

liver in many different ways, including but not limited to; increasing sensitivity to drug induced

liver injury, decreasing the antioxidant capacity of the liver and increasing hepatic oxidative

stress. These effects can severely disrupt liver function and consequently make the liver more

prone to damage from toxic compounds. I found that pre-existing obesity and ORFLD increased

susceptibility to EGCG mediated hepatotoxicity. The effects observed occurred in a dose

dependent manner, based on increasing dose of EGCG. Obese mice had lower survival rates and

exhibited higher levels of oxidative stress when compared to lean mice. However, the extent of

liver tissue damage was not significantly different between lean and obese mice. The effect of

high dose EGCG in lean mice was in some instances comparable to obese mice, when examined

in the context of overall tissue damage. No stark differences were seen when liver tissue from

mice in both diet groups treated with 500 and 750 mg/kg EGCG were evaluated for changes in

tissue necrosis, inflammation, hepatic lipidosis, hemorrhaging and apoptosis following EGCG

treatment. This observation could be attributed to the high carbohydrate content of the low fat

diet. Mice fed a high carbohydrate diet have been shown to accumulate lipid in the liver and

potentially exhibit signs of non-alcoholic fatty liver disease. EGCG treatment was able to

exacerbate tissue damage in lean mice treated with 500 and 750 mg/kg EGCG, causing the extent

of tissue damage to be similar to that seen in obese mice at those doses.

Page 143: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

130

To examine the effect of EGCG on hepatic markers of antioxidant response and

mitochondrial biogenesis/function in lean and obese mice.

Previous work in our lab showed that EGCG induced mitochondrial dysfunction in

cancer cells treated with varying doses of EGCG. This data suggested that the direct effects on

EGCG on the mitochondria observed, was a possible mechanisms by which EGCG can mediate

hepatotoxicity. I examined the gene expression, protein expression and/or activity of hepatic

markers of mitochondrial antioxidant response, biogenesis/function in a mouse model. I found

that EGCG treatment significantly reduced the antioxidant response and mitochondrial biogenesis

and function in the livers of lean and obese mice. Those effects occurred in a dose dependent

manner, with high doses of EGCG having the lowest gene expression, protein expression and/or

activity. EGCG treatment significantly reduced the gene and protein expression and activity of

endogenous antioxidants, as well as the gene expression of their regulators. EGCG treatment

significantly decreased the gene expression of transcription factors that regulate mitochondrial

biogenesis and genes involved in mitochondrial function in both lean and obese mice. These

results supported the hypothesis that the mitochondria are a target for EGCG induced

mitochondrial dysfunction in vivo, based on data obtained from previous in vitro studies, and

provide a mechanistic explanation for the observed hepatotoxic effects of EGCG.

Overall, the results of my study support the hypothesis that dietary pretreatment with

EGCG can mitigate the toxic effects of high oral bolus EGCG and that obesity increases

susceptibility to EGCG mediated liver toxicity. Through the inhibition of antioxidant response

and mitochondrial biogenesis/function EGCG reportedly increased mitochondrial oxidative stress

and induced mitochondrial dysfunction, which resulted in subsequent hepatotoxicity.

Page 144: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

131

The results of this work highlighted the potential of EGCG, a compound that is generally

regarded as safe and healthy when consumed in its more common form of a beverage to become

toxic when taken as a dietary supplement, where the doses are higher. These results can be

extrapolated to highlight the potential of many other compounds which are assumed to be safe

because they are natural, to be harmful when taken in more a concentrated form like that of a

dietary supplement as opposed to the more traditional forms, most commonly, beverages (teas).

Many dietary supplements are marketed with an emphasis on the fact that they are natural. As we

see in the results of this study, in the case of EGCG, this can be very misleading, and this could

be the case for many other supplements containing natural compounds. This exposes consumers

to risks that they are unaware of. It can also be seen from the data that more of a compound is not

necessarily more effective or safer. Again, dietary supplements expose persons to a higher dosage

form of a compound and based on how they are taken it is easy for persons to go from a safe dose

to a potentially toxic dose. From a public health perspective, this research sheds light on the need

to better evaluate compounds that are generally thought of as safe and used as dietary

supplements and to take into consideration lifestyle factors that affect the safety of these

compounds.

Page 145: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

132

Future Work

Effect of Other Lifestyle Factors on EGCG Mediated Liver Toxicity

In the present study I examined two aspects of the effect of lifestyle on the safety of

taking dietary supplement; diet and obesity. However many other lifestyle factors can potentially

increase the risks associated with taking supplements. It would be worthwhile to examine the

effects of other lifestyle factors, for example alcohol consumption, and how this impacts the

consumption of green tea supplements. Many people consume alcohol and take supplements.

Alcohol is a known hepatotoxin and the combined effect of alcohol and green tea supplement

warrants further investigation. A previous study in mice showed that green tea catechins can

exacerbate acetaminophen induced liver toxicity when taken together. [211]. The ability of green

tea catechins to potentiate the toxicity of acetaminophen indicates the ability of catechins to

interact with other compounds and impact their toxicity. Alcohol has also been shown to enhance

the toxicity of acetaminophen. Several studies report the toxic effect of consuming alcohol with

acetaminophen [212]. It has been established that alcohol induces cytochrome P450 enzymes,

which can metabolize acetaminophen into the toxic metabolite, NAPQI, which can bind to

cellular proteins and nucleic acids and induce toxicity. Additionally, alcohol has been shown to

reduce glutathione levels, which in turn can contribute to the increased toxicity of acetaminophen,

as the toxic metabolite is detoxified by glutathione conjugation [213]. Alcohol has also been

shown to affect the ability of the liver to metabolized compounds, by inhibiting the production of

UDP-glucuronic acid, another major detoxification pathway in the liver [214].

Based on this, it would be meaningful to investigate the potential of alcohol to impact

EGCG mediated liver toxicity. As was seen in previous results in this study, reduced glutathione

levels were associated with EGCG induced hepatotoxicity. With the ability of alcohol to also

Page 146: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

133

reduce glutathione levels, the possibility of high levels of oxidative stress is increased as a result

of these reduced GSH levels and can liver to further liver damage. EGCG was shown to be

extensively glucuronidated when metabolized in the liver. Alcohol was shown to reduce UDP-

glucuronic acid, which is needed for glucuronidation. This could lead to reduced EGCG

glucuronidation, increased bioavailability of EGCG, and possibly increased EGCG mediated liver

toxicity.

Determination of Hepatotoxicity of Green Tea Extract

In the present study we only focused on the effects of EGCG, as studies show that it is

the most active green tea polyphenol. With regards to dietary supplements, many contain green

tea extract as opposed to just EGCG. Now that we have established high dose EGCG mediated

liver toxicity, further studies on the potential toxicity of the whole extract should be conducted,

and the results compare it that of only EGCG. This would provide more insight into the safety of

green tea supplements.

One possible way to evaluate this, based on experiments conducted in this study, would

be perform in vivo studies, where mice are treated with only EGCG or with GTE containing the

same concentration of EGCG as the other treatment. Briefly, mice will be separated into 3

treatment groups. One group will receive a 750 mg/kg b.w. dose of EGCG, which in previous

studies was shown to induce hepatotoxicity. Another group will receive a dose of GTE solution

which has been prepared to contain 750 mg/kg EGCG. The last group will serve as controls and

not be treated with any EGCG. Mice will be treated with the respective solutions for 2-3 days.

Post treatment the plasma and liver tissue of the various treatment groups will be analyzed and

assessed for changes/differences in liver tissue damage, markers of liver toxicity and oxidative

Page 147: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

134

stress. The results of this study could potentially determine if supplements containing GTE are

toxic, and how much more or less toxic they compared to pure EGCG containing supplements.

Inhibition of Mitochondrial Proteins Involved in Mitochondrial Antioxidant Response and

Function by EGCG

Our data confirmed that EGCG treatment disrupted mitochondrial function and

corroborated previous data showing that EGCG induced mitochondrial dysfunction in vitro.

These results provided some evidence that the underlying mechanism of EGCG mediated liver

toxicity is mitochondrial dysfunction. However, further studies to generate more mechanistic data

are warranted. Following oxidation, EGCG had been shown to bind to cellular proteins like

cysteine and glutathione [4] [215]. As we have seen, glutathione is a key antioxidant in the cell

and plays an important role in the detoxification of compounds in the liver. Glutathione has been

shown to conjugate compounds like EGCG in order to eliminate them and can itself become

oxidized in order to remove reactive species. Reduced levels of glutathione have been observed

following high dose EGCG treatment, indicating the potential of EGCG to bind to this

antioxidant protein and deplete its levels.

EGCG has been shown to interact with a number of cytosolic and nuclear proteins in the

cell including enzymes, growth factors and their receptors, for MAP kinases, phosphatases, and

DNA methyltransferase. Some of these interactions have been determined to be inhibitory

interactions [216]. Inhibition of some of these proteins has been implicated in the

chemopreventative actions of EGCG and shown to affect signal transduction pathways,

transcription factors, DNA methylation, and mitochondrial function [217].

Having shown that mitochondrial dysfunction is the underlying mechanism of high dose

EGCG mediated hepatotoxicity, and that EGCG can interact with and inhibit several nuclear

Page 148: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

135

proteins, the potential of EGCG to bind to and directly inhibit key mitochondrial proteins should

be investigated. The results of these studies will provide further mechanistic data on the toxicity

of high dose EGCG. Tao et al showed the significance of mitochondria localized protein, Sirtuin

3 in mitochondrial response to EGCG treatment. The data showed that EGCG suppressed Sirtuin

3 mRNA and protein expression, as well as, SIRT3 activity in human oral squamous carcinoma

cells, where EGCG treatment had induced mitochondrial dysfunction [165]. Research into the

ability of EGCG to bind to and inhibit Sirtuin 3, needs to be investigated, as this protein has been

shown to be instrumental in mitochondrial response to oxidative stress, since it activates

mitochondrial antioxidant enzymes such as Sod2 and Gpx1 and Prx 3.

Results from the previous study shows that EGCG reduced mRNA and protein

expression, as well as activity of several mitochondria antioxidant enzymes. Decreases in levels

Sod2, Cat and Gpx1 were observed in the study. The effect of EGCG on these antioxidant

enzymes contributed to the observed increase in oxidative stress in the mice following EGCG

treatment. Investigating the interaction between EGCG and these antioxidant enzymes will

provide data on the ability of EGCG to induce oxidative stress and subsequent mitochondrial

dysfunction by directly inhibiting antioxidant enzymes in the mitochondria. A study by Pal et al

showed that EGCG had the highest binding affinity to and inhibition of pure and cellular catalase

in K562 cancer cells, which resulted in significant increases in cellular ROS and suppression of

cell viability [218]. However, the inhibition of mitochondrial superoxide dismutase (Sod2) and

glutathione peroxidase 1 (Gpx1) have not been very well studied. Additionally many of these

studies have been carried in vitro, in cancer cell lines. To get a clearer understanding of the direct

inhibiting effects of EGCG on these antioxidant enzymes as it relates to their in vivo activities

and EGCG mediated hepatotoxicity, binding and inhibition studies can be carried out on primary

hepatocytes of cells isolated from mice treated with varying doses of EGCG. Data from these

Page 149: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

136

studies will provide addition evidence for EGCG induced mitochondrial oxidative stress and

dysfunction.

Effect of EGCG Metabolites on EGCG Mediated Liver Toxicity

EGCG has been shown to undergo significant biotransformation in vivo (Figure1-3).

EGCG is readily methylated, sulfated, glucuronidated and undergoes ring-fission metabolism,

with glucuronidation being the predominant form of transformation in humans, rats, and mice [8].

EGCG has also been shown to form thiol conjugates following treatment with toxic doses of

EGCG. For example EGCG-cysteine conjugates were detected in urine of mice following

treatment with 200–400 mg/kg, i.p. or 1,500 mg/kg, i.g. EGCG [215]. EGCG biotransformation

has been shown to generally occur in liver microsomes and cytosol, as well as small intestines.

Although EGCG is greatly biotransformed, EGCG frequently appears in the unmetabolized form

in the plasma.

Recent studies have shown the EGCG and other polyphenols can also be metabolized by

gut microbiota, which can impact their bioavailability and subsequent activities[219][220]. It has

been shown that approximately 70% of green tea catechins pass from the small intestines to the

large intestines, where about 33% is in the parent form and undergoes microbial metabolism

[219]. The metabolites are then absorbed in the liver were they are metabolized by phase II

conjugating enzymes and then eliminated in the urine. The unconjugated metabolites are

eliminated in the feces. These microbial metabolites have also been shown to exert biological

activity. EGCG microbial metabolites have been shown to have antioxidant and anti-

inflammatory effects, and inhibit cell growth.

Page 150: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

137

With such a large amount of catechin being biotransformed by microbiota, and these

metabolites also having significant bioactivity, it raises the question of whether the effects of

EGCG are a direct result of the parent compound or the metabolites. The results of this study

focused on the toxicity of the EGCG parent compound, but did not take the metabolites into

consideration. In order to establish the toxicity of EGCG and not have these results be

confounded by the possible activity of microbial metabolites, EGCG studies should be repeated

and carried out in germ-free mice. Conducting studies in germ-free mice will eliminate the

microbially generated metabolites and determine toxicity of EGCG following liver and small

intestine metabolism where the parent compound has been shown to be active. The results of

these study can then be compared to the studies carried out in normal mice and be evaluated to

determine if EGCG metabolites impact the EGCG mediated liver toxicity.

Page 151: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

References

[1] M. Molinari, Kymberly D.S. Watt, T. Kruszyna, R. Nelson, M. Walsh, W.-Y. Huang, A.

Bjorn Nashan, and Kevork Peltekian, “Acute Liver Failure Induced by Green Tea

Extracts: Case Report and Review of the Literature,” Liver Transplant., vol. 13, no. 3, pp.

465–466, 2007.

[2] D. a Balentine, S. a Wiseman, and L. C. Bouwens, “The chemistry of tea flavonoids.,”

Crit. Rev. Food Sci. Nutr., vol. 37, no. 8, pp. 693–704, Dec. 1997.

[3] N. Khan and H. Mukhtar, “Tea and Health: Studies in Humans,” Curr Pharm Des, vol. 18,

no. 9, pp. 1199–1216, 2013.

[4] J. D. Lambert and R. J. Elias, “The antioxidant and pro-oxidant activities of green tea

polyphenols: a role in cancer prevention.,” Arch. Biochem. Biophys., vol. 501, no. 1, pp.

65–72, Sep. 2010.

[5] H. N. Graham, “Green tea composition, consumption, and polyphenol chemistry,” Prev.

Med. (Baltim)., vol. 21, no. 3, pp. 334–350, 1992.

[6] S. Bhagwat, D. B. Haytowitz, and J. M. Holden, “USDA Database for the Flavonoid

Content of Selected Foods Release 3 Prepared by USDA Database for the Flavonoid

Content of Selected Foods Release 3,” U.S. Dep. Argiculture, pp. 1–156, 2011.

[7] D. a Balentine, S. a Wiseman, and L. C. Bouwens, “The chemistry of tea flavonoids.,”

Crit. Rev. Food Sci. Nutr., vol. 37, no. 8, pp. 693–704, Dec. 1997.

[8] and S. S. C. S. Yang, J. D. Lambert, “Antioxidative and anti-carcinogenic activities of tea

polyphenols,” Arch Toxicol., vol. 83, no. 1, pp. 1–17, 2009.

[9] S. C. Forester and J. D. Lambert, “The role of antioxidant versus pro-oxidant effects of

green tea polyphenols in cancer prevention.,” Mol. Nutr. Food Res., vol. 55, no. 6, pp.

844–54, Jun. 2011.

[10] C. Rice-Evans, N. Miller, and G. Paganga, “Antioxidant properties of phenolic

compounds,” Trends Plant Sci., vol. 2, no. 4, pp. 152–159, 1997.

[11] C. YANG, J. LAMBERT, J. JU, G. LU, and S. SANG, “Tea and cancer prevention:

Molecular mechanisms and human relevance,” Toxicol. Appl. Pharmacol., vol. 224, no. 3,

pp. 265–273, 2007.

[12] J. V Higdon and B. Frei, “Tea catechins and polyphenols: health effects, metabolism, and

antioxidant functions.,” Crit. Rev. Food Sci. Nutr., vol. 43, no. 1, pp. 89–143, 2003.

[13] T. Nakagawa and T. Yokozawa, “Direct scavenging of nitric oxide and superoxide by

green tea,” Food Chem. Toxicol., vol. 40, no. 12, pp. 1745–1750, 2002.

[14] T. Yokozawa, E. J. Cho, Y. Hara, and K. Kitani, “Antioxidative activity of green tea

treated with radical initiator 2, 2’-azobis(2-amidinopropane) dihydrochloride.,” J. Agric.

Food Chem., vol. 48, no. 10, pp. 5068–5073, 2000.

[15] M. Kumamoto, T. Sonda, K. Nagayama, and M. Tabata, “Effects of pH and metal ions on

antioxidative activities of catechins.,” Biosci. Biotechnol. Biochem., vol. 65, no. January,

pp. 126–132, 2001.

[16] S. G. Khan, S. K. Katiyar, and R. Agarwal, “Enhancement of Antioxidant and Phase II

Enzymes by Oral Feeding of Green Tea Polyphenols in Drinking Water to SKH-1 Hairless

Mice : Possible Role in Cancer Chemoprevention Advances in Brief Enhancement of

Page 152: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

139

Antioxidant and Phase II Enzymes by Oral Feeding o,” pp. 4050–4052, 1992.

[17] J. Hong, H. Lu, X. Meng, H.-H. Colon, J. Ryu, Y. Hara, and C. S. Yang, “Stability ,

Cellular Uptake , Biotransformation , and Efflux of Tea Polyphenol ( − ) -

Epigallocatechin-3-Gallate in HT-29 Human Colon Adenocarcinoma Cells Stability ,

Cellular Uptake , Biotransformation , and Efflux of Tea Polyphenol,” pp. 7241–7246,

2002.

[18] L. Tao, S. C. Forester, and J. D. Lambert, “The role of the mitochondrial oxidative stress

in the cytotoxic effects of the green tea catechin, (-)-epigallocatechin-3-gallate, in oral

cells.,” Mol. Nutr. Food Res., vol. 58, no. 4, pp. 665–76, 2014.

[19] G. X. Li, Y. K. Chen, Z. Hou, H. Xiao, H. Jin, G. Lu, M. J. Lee, B. Liu, F. Guan, Z. Yang,

A. Yu, and C. S. Yang, “Pro-oxidative activities and dose-response relationship of (-)-

epigallocatechin-3-gallate in the inhibition of lung cancer cell growth: a comparative study

in vivo and in vitro,” Carcinogenesis, vol. 31, no. 5, pp. 902–910, 2010.

[20] J. D. Lambert, S.-J. Kwon, J. Hong, and C. S. Yang, “Salivary hydrogen peroxide

produced by holding or chewing green tea in the oral cavity.,” Free Radic. Res., vol. 41,

no. July, pp. 850–853, 2007.

[21] S. Wolfram, Y. Wang, and F. Thielecke, “Anti-obesity effects of green tea: from bedside

to bench.,” Mol. Nutr. Food Res., vol. 50, no. 2, pp. 176–87, Feb. 2006.

[22] I. Tokimitsu, “Effects of tea catechins on lipid metabolism and body fat accumulation.,”

Biofactors, vol. 22, pp. 141–143, 2004.

[23] T. Murase, a Nagasawa, J. Suzuki, T. Hase, and I. Tokimitsu, “Beneficial effects of tea

catechins on diet-induced obesity: stimulation of lipid catabolism in the liver.,” Int. J.

Obes. Relat. Metab. Disord., vol. 26, pp. 1459–1464, 2002.

[24] J. J. Choo, “Green tea reduces body fat accretion caused by high-fat diet in rats through

beta-adrenoceptor activation of thermogenesis in brown adipose tissue.,” J. Nutr.

Biochem., vol. 14, no. 11, pp. 671–6, 2003.

[25] M. Bose, J. D. Lambert, J. Ju, K. R. Reuhl, S. A. Shapses, and C. S. Yang, “The Major

Green Tea Polyphenol , Metabolic Syndrome , and Fatty Liver Disease in High-Fat – Fed

Mice 1 , 2,” no. April, pp. 1677–1683, 2008.

[26] S. Wolfram and R. Sandra, “TEAVIGO ( Epigallocatechin Gallate ) Supplementation

Prevents Obesity ...,” 2005.

[27] Y. K. Chen, C. Connie, and C. S. Yang, “Effects of Green Tea Polyphenol (-)-

Epigallocatechin-3-gallate on a Newly Developed High-fat/Western-style Diet-induced

Obesity and Metabolic Syndrome in Mice,” J Agric Food Chem, vol. 59, no. 6, pp. 11862–

11871, 2012.

[28] K. a Grove, S. Sae-tan, M. J. Kennett, and J. D. Lambert, “(-)-Epigallocatechin-3-gallate

inhibits pancreatic lipase and reduces body weight gain in high fat-fed obese mice.,”

Obesity (Silver Spring)., vol. 20, no. 11, pp. 2311–3, Nov. 2012.

[29] S. Sae-tan, K. A. Grove, M. J. Kennett, and J. D. Lambert, “(−)-Epigallocatechin-3-gallate

increases the expression of genes related to fat oxidation in the skeletal muscle of high fat-

fed mice,” Food Funct., vol. 2, no. 2, p. 111, 2011.

[30] S. Klaus, S. Pültz, C. Thöne-Reineke, and S. Wolfram, “Epigallocatechin gallate

attenuates diet-induced obesity in mice by decreasing energy absorption and increasing fat

oxidation.,” Int. J. Obes. (Lond)., vol. 29, no. 6, pp. 615–23, Jun. 2005.

[31] M.-S. Lee, C.-T. Kim, and Y. Kim, “Green Tea (&amp;ndash;)-Epigallocatechin-3-

Gallate Reduces Body Weight with Regulation of Multiple Genes Expression in Adipose

Tissue of Diet-Induced Obese Mice,” Ann. Nutr. Metab., vol. 54, no. 2, pp. 151–157,

2009.

[32] S. Sae-Tan, K. a Grove, M. J. Kennett, and J. D. Lambert, “(-)-Epigallocatechin-3-gallate

Page 153: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

140

increases the expression of genes related to fat oxidation in the skeletal muscle of high fat-

fed mice.,” Food Funct., vol. 2, no. 2, pp. 111–6, Feb. 2011.

[33] L. Meira, J. Bugni, and S. Green, “DNA damage induced by chronic inflammation

contributes to colon carcinogenesis in mice,” J. Clin. Invest., vol. 118, no. 7, pp. 2516–

2525, 2008.

[34] M. M.-Y. Chan, D. Fong, C.-T. Ho, and H.-I. Huang, “Inhibition of Inducible Nitric Oxide

Synthase Gene Expression and Enzyme Activity by Epigallocatechin Gallate, a Natural

Product from Green Tea,” Biochem. Pharmacol., vol. 54, no. 12, pp. 1281–1286, 1997.

[35] S. K. Katiyar, R. Agarwal, S. Ekker, G. S. Wood, and H. Mukhtar, “Protection against 12-

O-Tetradecanoylphorbol-13-Acetate-Caused Inflammation in SENCAR Mouse Ear Skin

by Polyphenolic Fraction Isolated from Green Tea,” Carcinogenesis, vol. 14, no. 3, pp.

361–365, 1993.

[36] S. K. Katiyar, M. S. Matsui, C. A. Elmets, and H. Mukhtar, “Polyphenolic antioxidant (-)-

epigallocatechin-3-gallate from green tea reduces UVB-induced inflammatory responses

and infiltration of leukocytes in human skin,” Photochem Photobiol, vol. 69, no. 2, pp.

148–153, 1999.

[37] S. K. Katiyar and H. Mukhtar, “Green tea polyphenol (-)-epigallocatechin-3-gallate

treatment to mouse skin prevents UVB-induced infiltration of leukocytes, depletion of

antigen-presenting cells, and oxidative stress.,” J. Leukoc. Biol., vol. 69, no. May, pp.

719–726, 2001.

[38] K. Kawai, N. H. Tsuno, J. Kitayama, Y. Okaji, K. Yazawa, M. Asakage, S. Sasaki, T.

Watanabe, K. Takahashi, and H. Nagawa, “Epigallocatechin gallate induces apoptosis of

monocytes,” J. Allergy Clin. Immunol., vol. 115, no. 1, pp. 186–191, 2005.

[39] M. Donà, I. Dell’Aica, F. Calabrese, R. Benelli, M. Morini, A. Albini, and S. Garbisa,

“Neutrophil restraint by green tea: inhibition of inflammation, associated angiogenesis,

and pulmonary fibrosis.,” J. Immunol., vol. 170, pp. 4335–4341, 2003.

[40] S. M. Haffner, “The Metabolic Syndrome: Inflammation, Diabetes Mellitus, and

Cardiovascular Disease,” Am. J. Cardiol., vol. 97, no. 2, pp. 3–11, 2006.

[41] E. Ford, “Prevalence of the Metabolic Syndrome Defined by the International Diabetes,”

vol. 28, no. 11, 2005.

[42] P. Paschos and K. Paletas, “Non alcoholic fatty liver disease and metabolic syndrome,”

Hippokratia, vol. 13, no. 1, pp. 9–19, 2009.

[43] F. Thielecke and M. Boschmann, “The potential role of green tea catechins in the

prevention of the metabolic syndrome – A review,” Phytochemistry, vol. 70, no. 1, pp. 11–

24, 2009.

[44] H. S. Chow, I. A. Hakim, D. R. Vining, J. A. Crowell, J. Ranger-moore, W. M. Chew, C.

A. Celaya, S. R. Rodney, Y. Hara, and D. S. Alberts, “Effects of Dosing Condition on the

Oral Bioavailability of Green Tea Catechins after Single-Dose Administration of

Polyphenon E in Healthy Individuals Effects of Dosing Condition on the Oral

Bioavailability of Green Tea Catechins after Single-Dose Adminis,” Clin. Cancer

Reserach, vol. 11, pp. 4627–4633, 2005.

[45] G. Mazzanti, F. Menniti-Ippolito, P. A. Moro, F. Cassetti, R. Raschetti, C. Santuccio, and

S. Mastrangelo, “Hepatotoxicity from green tea: a review of the literature and two

unpublished cases.,” Eur. J. Clin. Pharmacol., vol. 65, no. 4, pp. 331–41, Apr. 2009.

[46] R. J. Blendon, C. M. DesRoches, J. M. Benson, M. Brodie, and D. E. Altman,

“Americans’ views on the use and regulation of dietary supplements.,” Arch. Intern. Med.,

vol. 161, no. 6, pp. 805–810, 2001.

[47] J. Bradley, “Supplement Business Report 2015,” Nutr. Bus. J., 2015.

[48] A. C. Society, “Dietary Supplements : What Is Safe ? What you need to know first about

Page 154: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

141

dietary supplements,” pp. 1–16, 2007.

[49] T. C. Blumenthal M, Ferrier GKL, “Total Sales of Herbal Supplements in United States

Show Steady Growth,” Herb Gram, no. 71, pp. 64–66, 2006.

[50] K. a Grove and J. D. Lambert, “Human Intervention Studies Show That Tea ( Camellia

sinensis ) May Be Useful in the Prevention of Obesity 1 , 2,” J. Nutr., vol. 140, pp. 446–

453, 2010.

[51] J. D. Lambert, J. Hong, M. Lee, S. Sang, X. Meng, H. Lu, and C. S. Yang,

“Biotransformation and Bioavailability of Tea Polyphenols: Implications for Cancer

Prevention Research,” pp. 212–224, 2005.

[52] H. Lu, X. Meng, C. Li, S. Sang, C. Patten, S. Sheng, J. Hong, N. Bai, B. Winnik, C. T. Ho,

and C. S. Yang, “Glucuronides of tea catechins: enzymology of biosynthesis and

biological activities,” Drug Metab Dispos, vol. 31, no. 4, pp. 452–461, 2003.

[53] S. Sang, J. D. Lambert, J. Hong, S. Tian, M.-J. Lee, R. E. Stark, C.-T. Ho, and C. S. Yang,

“Synthesis and structure identification of thiol conjugates of (-)-epigallocatechin gallate

and their urinary levels in mice.,” Chem. Res. Toxicol., vol. 18, no. 11, pp. 1762–9, Nov.

2005.

[54] K. Nakagawa and T. Miyazawa, “Dose-dependent Incorporation of Tea catechins, (-)-

Epigallocatechin-3-gallate and (-)-Epigallocatechin, into Human plsama,” Biosci.

Biotechnol. Biochem., 1997.

[55] H. S. Chow, Y. Cai, I. A. Hakim, J. A. Crowell, F. Shahi, C. A. Brooks, R. T. Dorr, Y.

Hara, and D. S. Alberts, “Pharmacokinetics and Safety of Green Tea Polyphenols after

Multiple-Dose Administration of Epigallocatechin Gallate and Polyphenon E in Healthy

Individuals Pharmacokinetics and Safety of Green Tea Polyphenols after Multiple-Dose

Administration of Epigallo,” pp. 3312–3319, 2003.

[56] J. D. Lambert, M.-J. Lee, H. Lu, X. Meng, J. J. J. Hong, D. N. Seril, M. G. Sturgill, and C.

S. Yang, “Epigallocatechin-3-gallate is absorbed but extensively glucuronidated following

oral administration to mice.,” J. Nutr., vol. 133, no. 12, pp. 4172–7, Dec. 2003.

[57] A. Yashin, B. Nemzer, and Y. Yashin, “Bioavailability of Tea Components,” J. Food Res.,

vol. 1, no. 2, pp. 281–290, Apr. 2012.

[58] S. M. Henning, Y. Niu, N. H. Lee, G. D. Thames, R. R. Minutti, H. Wang, V. L. W. Go,

and D. Heber, “Bioavailability and antioxidant activity of tea flavanols after consumption

of green tea, black tea, or a green tea extract supplement.,” Am. J. Clin. Nutr., vol. 80, no.

6, pp. 1558–64, Dec. 2004.

[59] S. Bettuzzi, M. Brausi, F. Rizzi, G. Castagnetti, G. Peracchia, and A. Corti,

“Chemoprevention of human prostate cancer by oral administration of green tea catechins

in volunteers with high-grade prostate intraepithelial neoplasia: a preliminary report from

a one-year proof-of-principle study.,” Cancer Res., vol. 66, no. 2, pp. 1234–40, Jan. 2006.

[60] Bonkovsky, H.L., “Hepatotoxicity associated with supplements containing Chinese green

tea (Camellia sinensis),” Ann. Intern. Med., vol. 144, pp. 68–71, 2006.

[61] A. Federico, A. Tiso, and C. Loguercio, “A case of hepatotoxicity caused by green tea.,”

Free Radic. Biol. Med., vol. 43, no. 3, p. 474, 2007.

[62] M. Jimenez-Saenz and M. D. C. Martinez-Sanchez, “Acute hepatitis associated with the

use of green tea infusions.,” J. Hepatol., vol. 44, no. 3, pp. 616–7, 2006.

[63] J. D. Lambert, M. J. Kennett, S. Sang, K. R. Reuhl, J. Ju, and C. S. Yang, “Hepatotoxicity

of high oral dose (-)-epigallocatechin-3-gallate in mice.,” Food Chem. Toxicol. an Int. J.

Publ. Br. Ind. Biol. Res. Assoc., vol. 48, no. 1, pp. 409–416, 2010.

[64] I. M. Kapetanovic, J. a Crowell, R. Krishnaraj, a Zakharov, M. Lindeblad, and a

Lyubimov, “Exposure and toxicity of green tea polyphenols in fasted and non-fasted

dogs.,” Toxicology, vol. 260, no. 1–3, pp. 28–36, Jun. 2009.

Page 155: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

142

[65] G. Galati and P. J. O’Brien, “Potential toxicity of flavonoids and other dietary phenolics:

significance for their chemopreventive and anticancer properties,” Free Radic. Biol. Med.,

vol. 37, no. 3, pp. 287–303, 2004.

[66] M. Schmidt, H.-J. Schmitz, A. Baumgart, D. Guédon, M. I. Netsch, M.-H. Kreuter, C. B.

Schmidlin, and D. Schrenk, “Toxicity of green tea extracts and their constituents in rat

hepatocytes in primary culture.,” Food Chem. Toxicol., vol. 43, no. 2, pp. 307–14, 2005.

[67] R. L. Bailey, J. J. Gahche, P. E. Miller, P. R. Thomas, and J. T. Dwyer, “Why US adults

use dietary supplements.,” JAMA Intern. Med., vol. 173, no. 5, pp. 355–61, 2013.

[68] S. Kim, M. Lee, J. Hong, C. Li, T. J. Smith, G. Yang, D. N. Seril, and C. S. Yang,

“Plasma and Tissue Levels of Tea Catechins in Rats and Mice During Chronic

Consumption of Green Tea Polyphenols Plasma and Tissue Levels of Tea Catechins in

Rats and Mice During Chronic Consumption of Green Tea Polyphenols,” no. March 2014,

pp. 37–41, 2000.

[69] B. Tabakoff, N. Cornell, and P. L. Hoffman, “Alcohol tolerance.,” Ann. Emerg. Med., vol.

15, no. 9, pp. 1005–1012, 1986.

[70] D. P. Guh, W. Zhang, N. Bansback, Z. Amarsi, C. L. Birmingham, and A. H. Anis, “The

incidence of co-morbidities related to obesity and overweight: a systematic review and

meta-analysis.,” BMC Public Health, vol. 9, p. 88, Jan. 2009.

[71] A. Wree, A. Kahraman, G. Gerken, and A. Canbay, “Obesity affects the liver - the link

between adipocytes and hepatocytes.,” Digestion, vol. 83, no. 1–2, pp. 124–33, Jan. 2011.

[72] L. A. Adams and K. D. Lindor, “Nonalcoholic Fatty Liver Disease,” Ann. Epidemiol., vol.

17, no. 11, pp. 863–869, 2007.

[73] M. J. E. Brill, J. Diepstraten, A. Van Rongen, S. Van Kralingen, J. N. Van Den Anker, and

C. a J. Knibbe, “Impact of obesity on drug metabolism and elimination in adults and

children,” Clin. Pharmacokinet., vol. 51, no. 5, pp. 277–304, 2012.

[74] B. Fromenty, “Drug-induced liver injury in obesity.,” J. Hepatol., vol. 58, no. 4, pp. 824–

6, Apr. 2013.

[75] J. Aubert, K. Begriche, M. Delannoy, I. Morel, J. Pajaud, C. Ribault, S. Lepage, M. R.

McGill, C. Lucas-Clerc, B. Turlin, M.-A. Robin, H. Jaeschke, and B. Fromenty,

“Differences in early acetaminophen hepatotoxicity between obese ob/ob and db/db

mice.,” J. Pharmacol. Exp. Ther., vol. 342, no. 3, pp. 676–87, 2012.

[76] K. Begriche, J. Massart, M.-A. Robin, F. Bonnet, and B. Fromenty, “Mitochondrial

adaptations and dysfunctions in nonalcoholic fatty liver disease,” Hepatology, vol. 58, no.

4, pp. 1497–1507, 2013.

[77] B. K. Corcoran, G.B.; Wong, “Obesity as a Risk Factor in Drug-Induced Organ Injury :

Increased Liver and Kidney Damage by Acetaminophen in the Obese Overfed Rat1 by

hepatic,” 1987.

[78] J. D. Browning and J. D. Horton, “Molecular mediators of hepatic steatosis and liver

injury,” vol. 114, no. 2, 2004.

[79] a G. Dulloo, C. Duret, D. Rohrer, L. Girardier, N. Mensi, M. Fathi, P. Chantre, and J.

Vandermander, “Efficacy of a green tea extract rich in catechin polyphenols and caffeine

in increasing 24-h energy expenditure and fat oxidation in humans.,” Am. J. Clin. Nutr.,

vol. 70, no. 6, pp. 1040–5, Dec. 1999.

[80] J. D. Lambert, S. Sang, and C. S. Yang, “Possible controversy over dietary polyphenols:

benefits vs risks.,” Chem. Res. Toxicol., vol. 20, no. 4, pp. 583–5, Apr. 2007.

[81] G. Shen, C. Xu, R. Hu, M. R. Jain, S. Nair, W. Lin, C. S. Yang, J. Y. Chan, and a-N. T.

Kong, “Comparison of (-)-epigallocatechin-3-gallate elicited liver and small intestine gene

expression profiles between C57BL/6J mice and C57BL/6J/Nrf2 (-/-) mice.,” Pharm. Res.,

vol. 22, no. 11, pp. 1805–20, Nov. 2005.

Page 156: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

143

[82] L. Tang, M. Tang, L. Xu, H. Luo, T. Huang, J. Yu, L. Zhang, W. Gao, S. B. Cox, and J.-S.

Wang, “Modulation of aflatoxin biomarkers in human blood and urine by green tea

polyphenols intervention.,” Carcinogenesis, vol. 29, no. 2, pp. 411–7, Feb. 2008.

[83] R. a Isbrucker, J. a Edwards, E. Wolz, a Davidovich, and J. Bausch, “Safety studies on

epigallocatechin gallate (EGCG) preparations. Part 2: dermal, acute and short-term

toxicity studies.,” Food Chem. Toxicol., vol. 44, no. 5, pp. 636–50, May 2006.

[84] J. D. Lambert, M.-J. LEE, L. DIAMOND, J. JU, J. HONG, M. BOSE, H. L. NEWMARK,

and C. S. YANG, “Short Communication DOSE-DEPENDENT LEVELS OF

EPIGALLOCATECHIN-3-GALLATE IN HUMAN COLON ABSTRACT :,” Drug

Metab. Dispos., vol. 34, no. 1, pp. 8–11, 2006.

[85] G. Galati, A. Lin, A. M. Sultan, and P. J. O’Brien, “Cellular and in vivo hepatotoxicity

caused by green tea phenolic acids and catechins.,” Free Radic. Biol. Med., vol. 40, no. 4,

pp. 570–80, Feb. 2006.

[86] P. Tulayakul, K. S. Dong, J. Y. Li, N. Manabe, and S. Kumagai, “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, vol. 50, no. 3, pp. 339–348, 2007.

[87] K. R. Landis-piwowar, S. B. Wan, R. A. Wiegand, D. J. Kuhn, T. A. K. H. Chan, and Q.

P. Dou, “Methylation Suppresses the Proteasome-Inhibitory Function of Green Tea

Polyphenols,” no. September 2006, pp. 252–260, 2007.

[88] H. Lu, X. Meng, C. Li, S. Sang, C. Patten, S. Sheng, J. Hong, N. Bai, B. Winnik, C. Ho,

C. S. Yang, C. Biology, E. Mario, and F. Science, “GLUCURONIDES OF TEA

CATECHINS : ENZYMOLOGY OF BIOSYNTHESIS AND BIOLOGICAL

ACTIVITIES ABSTRACT :,” vol. 31, no. 4, pp. 452–461, 2003.

[89] L. Zhang, Y. Zheng, M. S. S. Chow, and Z. Zuo, “Investigation of intestinal absorption

and disposition of green tea catechins by Caco-2 monolayer model.,” Int. J. Pharm., vol.

287, no. 1–2, pp. 1–12, Dec. 2004.

[90] L. Badolo, B. Jensen, C. Säll, U. Norinder, P. Kallunki, and D. Montanari, “Evaluation of

309 molecules as inducers of CYP3A4, CYP2B6, CYP1A2, OATP1B1, OCT1, MDR1,

MRP2, MRP3 and BCRP in cryopreserved human hepatocytes in sandwich culture.,”

Xenobiotica., vol. 8254, no. January 2016, pp. 1–11, 2014.

[91] D. Pessayre, B. Fromenty, A. Berson, M.-A. Robin, P. Lettéron, R. Moreau, and A.

Mansouri, “Central role of mitochondria in drug-induced liver injury.,” Drug Metab. Rev.,

vol. 44, no. 1, pp. 34–87, Feb. 2012.

[92] a. Grieco, a. Forgione, L. Miele, V. Vero, a. V. Greco, a. Gasbarrini, and G. Gasbarrini,

“Fatty liver and drugs,” Eur. Rev. Med. Pharmacol. Sci., vol. 9, no. 5, pp. 261–263, 2005.

[93] P. Iozzo, M. Bucci, A. Roivainen, K. Någren, M. J. Järvisalo, J. Kiss, L. Guiducci, B.

Fielding, A. G. Naum, R. Borra, K. Virtanen, T. Savunen, P. a Salvadori, E. Ferrannini, J.

Knuuti, and P. Nuutila, “Fatty acid metabolism in the liver, measured by positron emission

tomography, is increased in obese individuals.,” Gastroenterology, vol. 139, no. 3, pp.

846–56, 856.e1–6, 2010.

[94] S. Yang, H. Zhu, Y. Li, H. Lin, K. Gabrielson, M. A. Trush, and A. M. Diehl,

“Mitochondrial adaptations to obesity related oxidant stress,” Arch. Biochem. Biophys.,

vol. 378, no. 2, pp. 259–268, 2000.

[95] C. S. Yang, X. Wang, G. Lu, and S. C. Picinich, “Cancer prevention by tea: animal

studies, molecular mechanisms and human relevance.,” Nat. Rev. Cancer, vol. 9, no. 6, pp.

429–39, Jun. 2009.

[96] C. S. Yang and J. G. Hong, “Prevention of Chronic Diseases by Tea: Possible

Mechanisms and Human Relevance,” Annu. Rev. Nutr. Vol 33, vol. 33, pp. 161–181,

2013.

Page 157: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

144

[97] Z. Hou, S. Sang, H. You, M. J. Lee, J. Hong, K. V. Chin, and C. S. Yang, “Mechanism of

action of (-)-epigallocatechin-3-gallate: Auto-oxidation- dependent inactivation of

epidermal growth factor receptor and direct effects on growth inhibition in human

esophageal cancer KYSE 150 cells,” Cancer Res., vol. 65, no. 17, pp. 8049–8056, 2005.

[98] L. Tao, S. C. Forester, and J. D. Lambert, “The role of the mitochondrial oxidative stress

in the cytotoxic effects of the green tea catechin, (-)-epigallocatechin-3-gallate, in oral

cells.,” Mol. Nutr. Food Res., vol. 58, no. 4, pp. 665–76, Apr. 2014.

[99] R. Hursel, W. Viechtbauer, and M. S. Westerterp-Plantenga, “The effects of green tea on

weight loss and weight maintenance: a meta-analysis,” Int. J. Obes., vol. 33, no. 9, pp.

956–961, 2009.

[100] O. J. Phung, W. L. Baker, L. J. Matthews, M. Lanosa, A. Thorne, and C. I. Coleman,

“Effect of green tea catechins with or without caffeine on anthropometric measures : a

systematic review and meta-analysis 1 – 3,” Am. J. Clin. Nutr., vol. 91, no. 1, pp. 73–81,

2010.

[101] T. M. Rains, S. Agarwal, and K. C. Maki, “Antiobesity effects of green tea catechins: a

mechanistic review,” J. Nutr. Biochem., vol. 22, no. 1, pp. 1–7, 2011.

[102] H. Wang, Y. Wen, Y. Du, X. Yan, H. Guo, J. A. Rycroft, N. Boon, E. M. R. Kovacs, and

D. J. Mela, “Effects of Catechin Enriched Green Tea on Body Composition,” Obesity, vol.

18, no. 4, pp. 773–779, 2010.

[103] B. Bayram, G. Runbach, J. Frank, and T. Esatbeyoglu, “Rapid Method for Glutathione

Quantitation Using High-Performance Liquid Chromatography with Coulometric

Electrochemical Detection,” J. Agric. Food Chem., vol. 62, pp. 402–408, 2014.

[104] N. Chalasani, J. a Marrero, J. Ahn, and K. Rajender Reddy, “ACG Clinical Guideline: The

Diagnosis and Management of Focal Liver Lesions.,” Am. J. Gastroenterol., no.

November 2013, pp. 1–20, 2014.

[105] K. D. James, S. C. Forester, and J. D. Lambert, “Dietary pretreatment with green tea

polyphenol, (−)-epigallocatechin-3-gallate reduces the bioavailability and hepatotoxicity

of subsequent oral bolus doses of (−)-epigallocatechin-3-gallate,” Food Chem. Toxicol.,

vol. 76, pp. 103–108, 2015.

[106] D. M. S. Ferreira, R. E. Castro, M. V. Machado, T. Evangelista, A. Silvestre, A. Costa, J.

Coutinho, F. Carepa, H. Cortez-Pinto, and C. M. P. Rodrigues, “Apoptosis and insulin

resistance in liver and peripheral tissues of morbidly obese patients is associated with

different stages of non-alcoholic fatty liver disease,” Diabetologia, vol. 54, no. 7, pp.

1788–1798, 2011.

[107] B. Sishi, B. Loos, B. Ellis, W. Smith, E. F. du Toit, and A.-M. Engelbrecht, “Diet-induced

obesity alters signalling pathways and induces atrophy and apoptosis in skeletal muscle in

a prediabetic rat model,” Exp. Physiol., vol. 96, no. 2, pp. 179–193, 2011.

[108] H. Zaher, J. T. Buters, J. M. Ward, M. K. Bruno, A. M. Lucas, S. T. Stern, S. D. Cohen,

and F. J. Gonzalez, “Protection against acetaminophen toxicity in CYP1A2 and CYP2E1

double-null mice,” Toxicol Appl Pharmacol, vol. 152, no. 1, pp. 193–199, 1998.

[109] L. W. York, S. Puthalapattu, and G. Y. Wu, “Nonalcoholic Fatty Liver Disease and Low-

Carbohydrate Diets,” Annu. Rev. Nutr., vol. 29, no. 1, pp. 365–379, 2009.

[110] L. C. Hudgins, M. Hellerstein, C. Seidman, R. Neese, J. Diakun, and J. Hirsch, “Human

fatty acid synthesis is stimulated by a eucaloric low fat, high carbohydrate diet.,” J. Clin.

Invest., vol. 97, no. 9, pp. 2081–91, 1996.

[111] N. M. Delzenne, N. A. Hernaux, and H. S. Taper, “A new model of acute liver steatosis

induced in rats by fasting followed by refeeding a high carbohydrate-fat free diet.

Biochemical and morphological analysis,” J. Hepatol. 1, vol. 1997, no. 7, pp. 880–885,

1997.

Page 158: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

145

[112] K. B. Scribner, D. B. Pawlak, and D. S. Ludwig, “Hepatic steatosis and increased

adiposity in mice consuming rapidly vs. slowly absorbed carbohydrate.,” Obesity (Silver

Spring)., vol. 15, no. 9, pp. 2190–9, 2007.

[113] M. Picard, O. S. Shirihai, B. J. Gentil, and Y. Burelle, “Mitochondrial morphology

transitions and functions: implications for retrograde signaling?,” AJP Regul. Integr.

Comp. Physiol., vol. 304, no. 6, pp. R393–R406, 2013.

[114] D. C. Chan, “Mitochondrial Fusion and Fission in Mammals,” Annu. Rev. Cell Dev. Biol.,

vol. 22, no. 1, pp. 79–99, 2006.

[115] J. Bereiter-Hahn, M. Vöth, S. Mai, and M. Jendrach, “Structural implications of

mitochondrial dynamics,” Biotechnol. J., vol. 3, pp. 765–780, 2008.

[116] T. Yu, R. J. Fox, L. S. Burwell, and Y. Yoon, “Regulation of mitochondrial fission and

apoptosis by the mitochondrial outer membrane protein hFis1.,” J. Cell Sci., vol. 118, no.

Pt 18, pp. 4141–51, 2005.

[117] R. J. Youle and D. P. Narendra, “Mechanisms of mitophagy,” Nat Rev Mol Cell Biol, vol.

12, no. 1, pp. 9–14, 2011.

[118] M. Jendrach, S. Mai, S. Pohl, M. Vöth, and J. Bereiter-Hahn, “Short- and long-term

alterations of mitochondrial morphology, dynamics and mtDNA after transient oxidative

stress,” Mitochondrion, vol. 8, no. 4, pp. 293–304, 2008.

[119] K. Begriche, J. Massart, M.-A. Robin, A. Borgne-Sanchez, and B. Fromenty, “Drug-

induced toxicity on mitochondria and lipid metabolism: mechanistic diversity and

deleterious consequences for the liver.,” J. Hepatol., vol. 54, no. 4, pp. 773–94, Apr. 2011.

[120] O. Surova and B. Zhivotovsky, “Various modes of cell death induced by DNA damage,”

Oncogene, vol. 32, no. 33, pp. 3789–3797, 2013.

[121] H. Esterbauer, R. J. Schaur, and H. Zollner, “Chemistry and Biochemistry of 4-

hydroxynonenal, malonaldehyde and related aldehydes,” Free Radic. Biol. Med., vol. 11,

no. 1, pp. 81–128, 1991.

[122] Y. Cigremis, H. Turel, K. Adiguzel, M. Akgoz, A. Kart, M. Karaman, and H. Ozen, “The

effects of acute acetaminophen toxicity on hepatic mRNA expression of SOD, CAT,

GSH-Px, and levels of peroxynitrite, nitric oxide, reduced glutathione, and

malondialdehyde in rabbit.,” Mol. Cell. Biochem., vol. 323, no. 1–2, pp. 31–8, 2009.

[123] S. Sudathip, K. A. Grove, and J. D. Lambert, “Laboratory Studies on Weight Control and

Prevention of Metabolic Syndrome by Green Tea,” vol. 64, no. 2, pp. 146–154, 2011.

[124] N. Ahmad and D. K. Feyes, “Green Tea Constituent Epigallocatechin-3-Gallate and

Induction of Apoptosis and Cell Cycle Arrest in Human Carcinoma Cells Agarwal , Hasan

Mukhtar * nolic compounds present in green tea Detection of Apoptosis by Quantification

of Apoptosis by Flow Cytomet,” pp. 1881–1886, 1881.

[125] G. Mazzanti, A. Di Sotto, and A. Vitalone, “Hepatotoxicity of green tea: an update,” Arch.

Toxicol., vol. 89, no. 8, pp. 1175–1191, 2015.

[126] S. Qanungo, “Epigallocatechin-3-gallate induces mitochondrial membrane depolarization

and caspase-dependent apoptosis in pancreatic cancer cells,” Carcinogenesis, vol. 26, no.

5, pp. 958–967, 2004.

[127] D. B. Zorov, M. Juhaszova, and S. J. Sollott, “Mitochondrial ROS-induced ROS release:

An update and review,” Biochim. Biophys. Acta - Bioenerg., vol. 1757, no. 5–6, pp. 509–

517, 2006.

[128] J. F. Turrens, “Mitochondrial formation of reactive oxygen species,” J. Physiol., vol. 552,

no. 2, pp. 335–344, 2003.

[129] F. Nassir and J. a Ibdah, “Role of mitochondria in nonalcoholic fatty liver disease.,” Int. J.

Mol. Sci., vol. 15, no. 5, pp. 8713–42, Jan. 2014.

[130] D. Jones and N. Kaplowitz, “Mechanisms of Pathogenesis in Drug Hepatotoxicity Putting

Page 159: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

146

the Stress on Mitochondria,” Mol. Interv., pp. 98–111, 2011.

[131] M. R. Mcgill, H. Yan, A. Ramachandran, G. J. Murray, E. Rollins, and H. Jaeschke,

“HepaRG CELLS: A HUMAN MODEL TO STUDY MECHANISMS OF

ACETAMINOPHEN HEPATOTOXICITY,” J Clin Invest, vol. 53, no. 3, pp. 974–982,

2012.

[132] H. Jaeschke, M. R. McGill, and A. Ramachandran, “Oxidant stress, mitochondria, and cell

death mechanisms in drug-induced liver injury: Lessons learned from acetaminophen

hepatotoxicity,” Drug Metab. Rev., vol. 44, no. 1, pp. 88–106, 2012.

[133] K. Kon, J.-S. Kim, H. Jaeschke, and J. J. Lemasters, “Mitochondrial permeability

transition in acetaminophen-induced necrosis and apoptosis of cultured mouse

hepatocytes.,” Hepatology, vol. 40, no. 5, pp. 1170–1179, 2004.

[134] M. R. Mcgill, M. R. Sharpe, C. D. Williams, M. Taha, S. C. Curry, and H. Jaeschke, “The

mechanism underlying acetaminophen- induced hepatotoxicity in humans and mice

involves mitochondrial damage and nuclear DNA fragmentation,” vol. 122, no. 4, 2012.

[135] K. Begriche, A. Igoudjil, D. Pessayre, and B. Fromenty, “Mitochondrial dysfunction in

NASH: Causes, consequences and possible means to prevent it,” Mitochondrion, vol. 6,

no. 1, pp. 1–38, 2006.

[136] S. K. Mantena, A. L. King, K. K. Andringa, H. B. Eccleston, and S. M. Bailey,

“Mitochondrial dysfunction and oxidative stress in the pathogenesis of alcohol- and

obesity-induced fatty liver diseases,” Free Radic. Biol. Med., vol. 44, no. 7, pp. 1259–

1272, 2008.

[137] R. S. Rector, J. P. Thyfault, G. M. Uptergrove, E. M. Morris, P. Naples, S. J. Borengasser,

C. R. Mikus, M. J. Laye, M. Harold, F. W. Booth, J. A. Ibdah, S. P. Naples, S. J.

Borengasser, C. R. Mikus, M. J. Laye, M. H. Laughlin, F. W. Booth, and J. A. Ibdah,

“Mitochondrial dysfunction precedes insulin resistance and hepatic steatosis and

contributes to the natural history of non- alcoholic fatty liver disease in an obese rodent

model,” J. Hepatol., vol. 52, no. 5, pp. 727–736, 2011.

[138] A. J. Sanyal, C. Campbell-Sargent, F. Mirshahi, W. B. Rizzo, M. J. Contos, R. K. Sterling,

V. A. Luketic, M. L. Shiffman, and J. N. Clore, “Nonalcoholic steatohepatitis: association

of insulin resistance and mitochondrial abnormalities.,” Gastroenterology, vol. 120, no. 5,

pp. 1183–92, 2001.

[139] J. P. Thyfault, R. S. Rector, G. M. Uptergrove, S. J. Borengasser, E. M. Morris, Y. Wei,

M. J. Laye, C. F. Burant, N. R. Qi, S. E. Ridenhour, L. G. Koch, S. L. Britton, and J. a

Ibdah, “Rats selectively bred for low aerobic capacity have reduced hepatic mitochondrial

oxidative capacity and susceptibility to hepatic steatosis and injury.,” J. Physiol., vol. 587,

no. Pt 8, pp. 1805–1816, 2009.

[140] S. Noori, K. Nasir, and T. Mahboob, “Effects of cocoa powder on oxidant/ antioxidant

status in liver, heart and kidney tissues of rats,” J. Anim. Plant Sci., vol. 19, no. 4, pp.

174–178, 2009.

[141] L. H. Johansson and L. A. Borg, “A spectrophotometric method for determination of

catalase activity in small tissue samples.,” Anal. Biochem., vol. 174, no. 1, pp. 331–6,

1988.

[142] a. R. Agarwal, L. Zhao, H. Sancheti, I. K. Sundar, I. Rahman, and E. Cadenas, “Short-

term cigarette smoke exposure induces reversible changes in energy metabolism and

cellular redox status independent of inflammatory response in mouse lung,” AJP Lung

Cell. Mol. Physiol., pp. 889–898, 2012.

[143] J. Pi, Y. Bai, K. W. Daniel, D. Liu, O. Lyght, D. Edelstein, M. Brownlee, B. E. Corkey,

and S. Collins, “Persistent oxidative stress due to absence of uncoupling protein 2

associated with impaired pancreatic β-cell function,” Endocrinology, vol. 150, no. 7, pp.

Page 160: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

147

3040–3048, 2009.

[144] Q. Huang, X. Jin, E. T. Gaillard, B. L. Knight, F. D. Pack, J. H. Stoltz, S. Jayadev, and K.

T. Blanchard, “Gene expression profiling reveals multiple toxicity endpoints induced by

hepatotoxicants.,” Mutat. Res., vol. 549, no. 1–2, pp. 147–67, 2004.

[145] T. Yamamoto, R. Kikkawa, H. Yamada, and I. Horii, “Identification of oxidative stress-

related proteins for predictive screening of hepatotoxicity using a proteomic approach.,” J.

Toxicol. Sci., vol. 30, no. 3, pp. 213–227, 2005.

[146] A. I. Rupérez, A. Gil, and C. M. Aguilera, “Genetics of oxidative stress in obesity,” Int. J.

Mol. Sci., vol. 15, no. 2, pp. 3118–3144, 2014.

[147] I. Y. Iskusnykh, T. N. Popova, A. a Agarkov, M. Â. a Pinheiro de Carvalho, and S. G.

Rjevskiy, “Expression of Glutathione Peroxidase and Glutathione Reductase and Level of

Free Radical Processes under Toxic Hepatitis in Rats.,” J. Toxicol., vol. 2013, p. 870628,

2013.

[148] S. Furukawa, T. Fujita, M. Shimabukuro, M. Iwaki, Y. Yamada, Y. Nakajima, O.

Nakayama, and M. Makishima, “Increased oxidative stress in obesity and its impact on

metabolic syndrome,” vol. 114, no. 12, 2004.

[149] S. H. Bae, S. H. Sung, H. E. Lee, H. T. Kang, S. K. Lee, S. Y. Oh, H. A. Woo, I. S. Kil,

and S. G. Rhee, “Peroxiredoxin III and sulfiredoxin together protect mice from pyrazole-

induced oxidative liver injury.,” Antioxid. Redox Signal., vol. 17, no. 10, pp. 1351–61,

2012.

[150] A. G. Cox, C. C. Winterbourn, and M. B. Hampton, “Mitochondrial peroxiredoxin

involvement in antioxidant defence and redox signalling,” Biochem. J., vol. 425, no. 2, pp.

313–325, 2010.

[151] C. Musicco, V. Capelli, V. Pesce, A. M. Timperio, M. Calvani, L. Mosconi, L. Zolla, P.

Cantatore, and M. N. Gadaleta, “Accumulation of overoxidized Peroxiredoxin III in aged

rat liver mitochondria.,” Biochim. Biophys. Acta, vol. 1787, no. 7, pp. 890–6, 2009.

[152] M. Chevallet, E. Wagner, S. Luche, A. van Dorsselaer, E. Leize-Wagner, and T.

Rabilloud, “Regeneration of Peroxiredoxins during Recovery after Oxidative Stress:

ONLY SOME OVEROXIDIZED PEROXIREDOXINS CAN BE REDUCED DURING

RECOVERY AFTER OXIDATIVE STRESS,” J. Biol. Chem., vol. 278, no. 39, pp.

37146–37153, 2003.

[153] F. Yin, H. Sancheti, and E. Cadenas, “Mitochondrial Thiols in the Regulation of Cell

Death Pathways,” Antioxid. Redox Signal., vol. 17, no. 12, pp. 1714–1727, 2012.

[154] M. Sato and M. Kondoh, “Recent studies on metallothionein: protection against toxicity of

heavy metals and oxygen free radicals.,” The Tohoku journal of experimental medicine,

vol. 196, no. 1. pp. 9–22, 2002.

[155] B. Ruttkay-Nedecky, L. Nejdl, J. Gumulec, O. Zitka, M. Masarik, T. Eckschlager, M.

Stiborova, V. Adam, and R. Kizek, “The Role of Metallothionein in Oxidative Stress,” Int.

J. Mol. Sci., vol. 14, no. 3, pp. 6044–6066, 2013.

[156] M. Vašák and G. Meloni, “Chemistry and biology of mammalian metallothioneins,” JBIC

J. Biol. Inorg. Chem., vol. 16, no. 7, pp. 1067–1078, 2011.

[157] G. K. Andrews, “Regulation of metallothionein gene expression by oxidative stress and

metal ions,” Biochem. Pharmacol., vol. 59, no. 99, pp. 95–104, 2000.

[158] J. Mao, H. Yu, C. Wang, L. Sun, W. Jiang, P. Zhang, Q. Xiao, D. Han, D. Han, H. Saiyin,

J. Zhu, T. Chen, L. R. Roberts, H. Huang, and L. Yu, “Metallothionein MT1M is a tumor

suppressor of human hepatocellular carcinomas.,” Carcinogenesis, vol. 33, no. 12, pp.

2568–77, 2012.

[159] C. Saito, H.-M. Yan, A. Artigues, M. T. Villar, A. Farhood, and H. Jaeschke, “Mechanism

of protection by metallothionein against acetaminophen hepatotoxicity,” Toxicol. Appl.

Page 161: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

148

Pharmacol., vol. 242, no. 2, pp. 182–190, 2010.

[160] H. Waller-Evans, C. Hue, J. Fearnside, A. R. Rothwell, H. E. Lockstone, S. Caldérari, S.

P. Wilder, J.-B. Cazier, J. Scott, and D. Gauguier, “Nutrigenomics of High Fat Diet

Induced Obesity in Mice Suggests Relationships between Susceptibility to Fatty Liver

Disease and the Proteasome,” PLoS One, vol. 8, no. 12, p. e82825, 2013.

[161] T. Shi, F. Wang, E. Stieren, and Q. Tong, “SIRT3, a mitochondrial sirtuin deacetylase,

regulates mitochondrial function and thermogenesis in brown adipocytes,” J. Biol. Chem.,

vol. 280, no. 14, pp. 13560–13567, 2005.

[162] X. Kong, R. Wang, Y. Xue, X. Liu, H. Zhang, Y. Chen, F. Fang, and Y. Chang, “Sirtuin 3,

a New Target of PGC-1α, Plays an Important Role in the Suppression of ROS and

Mitochondrial Biogenesis,” PLoS One, vol. 5, no. 7, p. e11707, 2010.

[163] M. B. Scher, A. Vaquero, and D. Reinberg, “SirT3 is a nuclear NAD+-dependent histone

deacetylase that translocates to the mitochondria upon cellular stress.,” Genes Dev., vol.

21, no. 8, pp. 920–8, 2007.

[164] A. S. Bause and M. C. Haigis, “SIRT3 regulation of mitochondrial oxidative stress,” Exp.

Gerontol., vol. 48, no. 7, pp. 634–639, 2013.

[165] L. Tao, J.-Y. Park, and J. D. Lambert, “Differential prooxidative effects of the green tea

polyphenol, (-)-epigallocatechin-3-gallate, in normal and oral cancer cells are related to

differences in sirtuin 3 signaling.,” Mol. Nutr. Food Res., pp. 1–9, 2014.

[166] H. Pi, S. Xu, R. J. Reiter, P. Guo, L. Zhang, Y. Li, M. Li, Z. Cao, L. Tian, J. Xie, R.

Zhang, M. He, Y. Lu, C. Liu, W. Duan, Z. Yu, and Z. Zhou, “SIRT3-SOD2-mROS-

dependent autophagy in cadmium-induced hepatotoxicity and salvage by melatonin.,”

Autophagy, vol. 11, no. 7, pp. 1037–51, 2015.

[167] M. Choudhury, K. R. Jonscher, and J. E. Friedman, “Reduced mitochondrial function in

obesity-associated fatty liver: SIRT3 takes on the fat,” Aging (Albany. NY)., vol. 3, no. 2,

pp. 175–178, 2011.

[168] E. Ambrogini, M. Almeida, M. Martin-Millan, J.-H. Paik, R. A. Depinho, L. Han, J.

Goellner, R. S. Weinstein, R. L. Jilka, C. A. O’Brien, and S. C. Manolagas, “FoxO-

mediated defense against oxidative stress in osteoblasts is indispensable for skeletal

homeostasis in mice.,” Cell Metab., vol. 11, no. 2, pp. 136–46, 2010.

[169] C. B. Chiribau, L. Cheng, C. Ioan, Y. Yu, R. E. Clempus, and D. Sorescu, “Expression in

Human Cardiac Fibroblasts FOXO3A Regulates Peroxiredoxin III Expression in Human

Cardiac Fibroblasts *,” vol. 283, no. 13, pp. 8211–8217, 2008.

[170] D. P. Relling, L. B. Esberg, C. X. Fang, W. T. Johnson, E. J. Murphy, E. C. Carlson, J. T.

Saari, and J. Ren, “High-fat diet-induced juvenile obesity leads to cardiomyocyte

dysfunction and upregulation of Foxo3a transcription factor independent of lipotoxicity

and apoptosis,” J Hypertens, vol. 24, no. 3, pp. 549–561, 2006.

[171] T. Nguyen, P. Nioi, and C. B. Pickett, “The Nrf2-Antioxidant Response Element

Signaling Pathway and Its Activation by Oxidative Stress,” J. Biol. Chem., vol. 284, no.

20, pp. 13291–13295, 2009.

[172] Q. Ma, “Role of Nrf2 in Oxidative Stress and Toxicity,” Gynecol Oncol., vol. 136, no. 3,

pp. 554–561, 2015.

[173] M. A. Schultz, A. B. Abdel-Mageed, and D. Mondal, “The nrf1 and nrf2 balance in

oxidative stress regulation and androgen signaling in prostate cancer cells.,” Cancers

(Basel)., vol. 2, no. 2, pp. 1354–78, 2010.

[174] D. Wang, Y. Wang, X. Wan, C. S. Yang, and J. Zhang, “Green tea polyphenol ( − ) -

epigallocatechin-3-gallate triggered hepatotoxicity in mice : Responses of major

antioxidant enzymes and the Nrf2 rescue pathway,” Toxicol. Appl. Pharmacol., vol. 283,

no. 1, pp. 65–74, 2015.

Page 162: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

149

[175] M. J. Evans and R. C. Scarpulla, “NRF-1: A trans-activator of nuclear-encoded respiratory

genes in animal cells,” Genes Dev., vol. 4, no. 6, pp. 1023–1034, 1990.

[176] S. S. Dhar, S. Ongwijitwat, and M. T. T. Wong-Riley, “Nuclear respiratory factor 1

regulates all ten nuclear-encoded subunits of cytochrome c oxidase in neurons.,” J. Biol.

Chem., vol. 283, no. 6, pp. 3120–9, 2008.

[177] E. D. Yoboue and A. Devin, “Reactive Oxygen Species-Mediated Control of

Mitochondrial Biogenesis,” Int. J. Cell Biol., vol. 2012, pp. 1–8, 2012.

[178] N. Gleyzer, K. Vercauteren, and R. C. Scarpulla, “Control of mitochondrial transcription

specificity factors (TFB1M and TFB2M) by nuclear respiratory factors (NRF-1 and NRF-

2) and PGC-1 family coactivators.,” Mol. Cell. Biol., vol. 25, no. 4, pp. 1354–1366, 2005.

[179] H. B. Suliman, M. S. Carraway, K. E. Welty-Wolf, a. R. Whorton, and C. a. Piantadosi,

“Lipopolysaccharide Stimulates Mitochondrial Biogenesis via Activation of Nuclear

Respiratory Factor-1,” J. Biol. Chem., vol. 278, no. 42, pp. 41510–41518, 2003.

[180] Z. Wu, P. Puigserver, U. Andersson, C. Zhang, G. Adelmant, V. Mootha, A. Troy, S.

Cinti, B. Lowell, R. C. Scarpulla, and B. M. Spiegelman, “Mechanisms controlling

mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1,”

Cell, vol. 98, no. 1, pp. 115–124, 1999.

[181] Y. Zhang, Y. He, H. Yu, F. Ma, J. Wu, and X. Zhang, “Liquiritigenin Protects Rats from

Carbon Tetrachloride Induced Hepatic Injury through PGC-1 α Pathway,” Evidence-Based

Complement. Altern. Med., vol. 2015, pp. 1–9, 2015.

[182] H. Liang and W. F. Ward, “PGC-1alpha: a key regulator of energy metabolism.,” Adv.

Physiol. Educ., vol. 30, no. 4, pp. 145–51, 2006.

[183] P. Puigserver and B. M. Spiegelman, “Peroxisome Proliferator-Activated Receptor-γ

Coactivator 1α (PGC-1α): Transcriptional Coactivator and Metabolic Regulator,” Endocr.

Rev., vol. 24, no. 1, pp. 78–90, 2003.

[184] M. P. Valdecantos, P. Pérez-Matute, P. González-Muniesa, P. L. Prieto-Hontoria, M. J.

Moreno-Aliaga, and J. a. Martínez, “Lipoic Acid Improves Mitochondrial Function in

Nonalcoholic Steatosis Through the Stimulation of Sirtuin 1 and Sirtuin 3,” Obesity, vol.

20, no. 10, pp. 1974–1983, 2012.

[185] L. L. Clay Montier, J. J. Deng, and Y. Bai, “Number matters: control of mammalian

mitochondrial DNA copy number.,” J. Genet. Genomics, vol. 36, no. 3, pp. 125–31, 2009.

[186] D. F. Stowe, A. K. S. Camara, E. Cadenas, S. Dikalov, and R. Rossignol, “Mitochondrial

Reactive Oxygen Species Production in Excitable Cells : Modulators of Mitochondrial and

Cell Function,” vol. 11, no. 6, 2009.

[187] N. Pieters, G. Koppen, K. Smeets, D. Napierska, M. Plusquin, S. De Prins, H. Van De

Weghe, V. Nelen, B. Cox, A. Cuypers, P. Hoet, G. Schoeters, and T. S. Nawrot,

“Decreased Mitochondrial DNA Content in Association with Exposure to Polycyclic

Aromatic Hydrocarbons in House Dust during Wintertime: From a Population Enquiry to

Cell Culture,” PLoS One, vol. 8, no. 5, p. e63208, 2013.

[188] R. Barazzoni, K. R. Short, and K. S. Nair, “Effects of aging on mitochondrial DNA copy

number and cytochrome c oxidase gene expression in rat skeletal muscle, liver, and

heart.,” J. Biol. Chem., vol. 275, no. 5, pp. 3343–3347, 2000.

[189] L. Huo and R. C. Scarpulla, “Mitochondrial DNA instability and peri-implantation

lethality associated with targeted disruption of nuclear respiratory factor 1 in mice.,” Mol.

Cell. Biol., vol. 21, no. 2, pp. 644–654, 2001.

[190] L. K. Heilbronn, K. G. Seng, N. Turner, L. V. Campbell, and D. J. Chisholm, “Markers of

mitochondrial biogenesis and metabolism are lower in overweight and obese insulin-

resistant subjects,” J. Clin. Endocrinol. Metab., vol. 92, no. 4, pp. 1467–1473, 2007.

[191] S. Miranda, R. Foncea, J. Guerrero, and F. Leighton, “Oxidative stress and upregulation of

Page 163: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

150

mitochondrial biogenesis genes in mitochondrial DNA-depleted HeLa cells.,” Biochem.

Biophys. Res. Commun., vol. 258, no. 1, pp. 44–49, 1999.

[192] M. I. Ekstrand, “Mitochondrial transcription factor A regulates mtDNA copy number in

mammals,” Hum. Mol. Genet., vol. 13, no. 9, pp. 935–944, 2004.

[193] J. V Virbasius and R. C. Scarpulla, “Activation of the human mitochondrial transcription

factor A gene by nuclear respiratory factors: a potential regulatory link between nuclear

and mitochondrial gene expression in organelle biogenesis.,” Proc. Natl. Acad. Sci. U. S.

A., vol. 91, no. 4, pp. 1309–13, 1994.

[194] C. a. Piantadosi and H. B. Suliman, “Mitochondrial transcription factor A induction by

redox activation of nuclear respiratory factor 1,” J. Biol. Chem., vol. 281, no. 1, pp. 324–

333, 2006.

[195] H. Kim, M. J. Kim, J. E. Jeong, J. Chung, H.-J. Lee, and H. Koh, “Mitochondrial

transcription factor B1 is required for mitochondrial function and oxidative stress

resistance in Drosophila,” Genes Genomics, vol. 32, no. 5, pp. 455–461, 2010.

[196] V. V Sharoyko, M. Abels, J. Sun, L. M. Nicholas, I. G. Mollet, J. A. Stamenkovic, I.

Gohring, S. Malmgren, P. Storm, J. Fadista, P. Spegel, M. D. Metodiev, N. G. Larsson, L.

Eliasson, N. Wierup, and H. Mulder, “Loss of TFB1M results in mitochondrial

dysfunction that leads to impaired insulin secretion and diabetes,” Hum. Mol. Genet., vol.

23, no. 21, pp. 5733–5749, 2014.

[197] M. D. Metodiev, N. Lesko, C. B. Park, Y. Cámara, Y. Shi, R. Wibom, K. Hultenby, C. M.

Gustafsson, and N.-G. Larsson, “Methylation of 12S rRNA Is Necessary for In Vivo

Stability of the Small Subunit of the Mammalian Mitochondrial Ribosome,” Cell Metab.,

vol. 9, no. 4, pp. 386–397, 2009.

[198] Y. Matsushima, R. Garesse, and L. S. Kaguni, “Drosophila mitochondrial transcription

factor B2 regulates mitochondrial DNA copy number and transcription in schneider

cells.,” J. Biol. Chem., vol. 279, no. 26, pp. 26900–5, 2004.

[199] C. Adán, Y. Matsushima, R. Hernández-Sierra, R. Marco-Ferreres, M. A. Fernández-

Moreno, E. González-Vioque, M. Calleja, J. J. Aragón, L. S. Kaguni, and R. Garesse,

“Mitochondrial transcription factor B2 is essential for metabolic function in Drosophila

melanogaster development.,” J. Biol. Chem., vol. 283, no. 18, pp. 12333–42, 2008.

[200] Y. Matsushima, C. Adan, R. Garesse, and L. S. Kaguni, “Drosophila Mitochondrial

Transcription Factor B1 Modulates Mitochondrial Translation but Not Transcription or

DNA Copy Number in Schneider Cells,” J. Biol. Chem., vol. 280, no. 17, pp. 16815–

16820, 2005.

[201] M. Falkenberg, M. Gaspari, A. Rantanen, A. Trifunovic, N.-G. Larsson, and C. M.

Gustafsson, “Mitochondrial transcription factors B1 and B2 activate transcription of

human mtDNA,” Nat. Genet., vol. 31, no. 3, pp. 289–294, 2002.

[202] A. N. Malik and A. Czajka, “Is mitochondrial DNA content a potential biomarker of

mitochondrial dysfunction?,” Mitochondrion, vol. 13, no. 5, pp. 481–492, 2013.

[203] K. C. Pramanik, S. R. Boreddy, and S. K. Srivastava, “Role of Mitochondrial Electron

Transport Chain Complexes in Capsaicin Mediated Oxidative Stress Leading to Apoptosis

in Pancreatic Cancer Cells,” PLoS One, vol. 6, no. 5, p. e20151, 2011.

[204] M. C. Saleh, M. B. Wheeler, and C. B. Chan, “Uncoupling protein-2: evidence for its

function as a metabolic regulator.,” Diabetologia, vol. 45, no. 2, pp. 174–87, 2002.

[205] M. V Carretero, L. Torres, U. Latasa, E. R. Garcia-Trevijano, J. Prieto, J. M. Mato, and M.

A. Avila, “Transformed but not normal hepatocytes express UCP2,” Febs Lett., vol. 439,

no. 1–2, pp. 55–58, 1998.

[206] M. Horimoto, M. B. Resnick, T. a Konkin, J. Routhier, J. R. Wands, and G. Baffy,

“Expression of uncoupling protein-2 in human colon cancer.,” Clin. Cancer Res., vol. 10,

Page 164: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

151

no. 18 Pt 1, pp. 6203–7, 2004.

[207] N. Tsuboyama-Kasaoka, M. Takahashi, H. Kim, and O. Ezaki, “Up-regulation of liver

uncoupling protein-2 mRNA by either fish oil feeding or fibrate administration in mice.,”

Biochem. Biophys. Res. Commun., vol. 257, no. 3, pp. 879–885, 1999.

[208] P. Fülöp, Z. Derdák, A. Sheets, E. Sabo, E. P. Berthiaume, M. B. Resnick, J. R. Wands, G.

Paragh, and G. Baffy, “Lack of UCP2 reduces Fas-mediated liver injury in ob/ob mice and

reveals importance of cell-specific UCP2 expression,” Hepatology, vol. 44, no. 3, pp.

592–601, 2006.

[209] a Rashid, T. C. Wu, C. C. Huang, C. H. Chen, H. Z. Lin, S. Q. Yang, F. Y. Lee, and a M.

Diehl, “Mitochondrial proteins that regulate apoptosis and necrosis are induced in mouse

fatty liver.,” Hepatology, vol. 29, no. 4, pp. 1131–1138, 1999.

[210] K. D. Chavin, S. Yang, H. Z. Lin, J. Chatham, V. P. Chacko, J. B. Hoek, E. Walajtys-

Rode, a Rashid, C. H. Chen, C. C. Huang, T. C. Wu, M. D. Lane, and a M. Diehl,

“Obesity induces expression of uncoupling protein-2 in hepatocytes and promotes liver

ATP depletion.,” J. Biol. Chem., vol. 274, no. 9, pp. 5692–700, 1999.

[211] W. F. Salminen, X. Yang, Q. Shi, J. Greenhaw, K. Davis, and A. a. Ali, “Green tea extract

can potentiate acetaminophen-induced hepatotoxicity in mice,” Food Chem. Toxicol., vol.

50, no. 5, pp. 1439–1446, 2012.

[212] S. M. Riordan, R. Williams, S. M. Riordan, and R. Williams, “Alcohol exposure and

paracetamol-induced hepatotoxicity,” Addict. Biol., vol. 7, pp. 191–206, 2002.

[213] M. Lautergurg, BH; Velez, “Glutathione deficiency in alcoholics : risk factor for

paracetamol hepatotoxicity,” Gut, vol. 29, no. April, pp. 1153–1157, 1988.

[214] R. Weathermon, D. Pharm, and D. W. Crabb, “Alcohol and Medication Interactions,”

Alcohol Res. Heal., vol. 23, pp. 40–54, 1999.

[215] S. Sang, J. D. Lambert, J. Hong, S. Tian, M. Lee, R. E. Stark, C. Ho, and C. S. Yang,

“Synthesis and Structure Identification of Thiol Conjugates of ( - ) -Epigallocatechin

Gallate and Their Urinary Levels in Mice †,” Chem. Res. Toxicol., no. 18, pp. 1762–1769,

2005.

[216] S. K. Patra, F. Rizzi, A. Silva, D. O. Rugina, and S. Bettuzzi, “MOLECULAR TARGETS

OF (–)-EPIGALLOCATECHIN-3-GALLATE (EGCG): SPECIFICITY AND

INTERACTION WITH MEMBRANE LIPID RAFTS,” J. Physiol. Pharmacol., no. 59,

pp. 217–235, 2008.

[217] H.-S. Kim, M. J. Quon, and J. Kim, “New insights into the mechanisms of polyphenols

beyond antioxidant properties; lessons from the green tea polyphenol, epigallocatechin 3-

gallate,” Redox Biol., vol. 2, pp. 187–195, 2014.

[218] S. Pal, S. K. Dey, and C. Saha, “Inhibition of Catalase by Tea Catechins in Free and

Cellular State : A Biophysical Approach,” vol. 9, no. 7, 2014.

[219] M. Monagas, M. Urpi-sarda, and S. Fernando, “Insights into the metabolism and microbial

biotransformation of dietary flavan-3-ols and the bioactivity of their metabolites,” Food

Funct., vol. 1, pp. 233–253, 2010.

[220] T. Ozdal, D. A. Sela, J. Xiao, D. Boyacioglu, and F. Chen, “The Reciprocal Interactions

between Polyphenols and Gut Microbiota and Effects on Bioaccessibility,” Nutrients, vol.

8, no. 78, pp. 1–36, 2016.

Page 165: EFFECT OF DIETARY PRETREATMENT AND OBESITY ON

VITA

Karma James

EDUCATION Doctor of Philosophy – Food Science, May, 2016

Pennsylvania State University, State College, PA Bachelor of Science – Chemistry, December, 2010 Grambling State University, Grambling, LA

EXPERIENCE

Research Assistant – Penn State, Dept. Food Science August 2011 – May 2016 Teaching Assistant – Penn State, Dept. Food Science Fall 2012 – Fall 2015

Research Intern – MIT, Dept. Chemistry June – August 2010 Research Intern – MIT, Dept. Chemistry June – August 2009

PUBLICATIONS AND PRESENTATIONS

1. James K, Forester SC, Lambert JD, (2015) Dietary pretreatment with green tea polyphenol, (−)-epigallocatechin-3-gallate reduces the bioavailability and hepatotoxicity of subsequent oral bolus doses of (−)-epigallocatechin-3-gallate. Food and Chemical Toxicology 76 (2015) 103–108.

2. James K, Lambert JD, (2015) Effect of Obesity on the Hepatotoxicity of High Dose Oral (-)-Epigallocatechin-3-gallate. Experimental Biology. Boston, MA; Gamma Sigma Delta Expo. University Park, PA; Graduate Research Exhibition. University Park, PA.

3. James K, Forester SC, Lambert JD, (2014) Effect of dietary (-)-epigallocatechin-3-gallate (EGCG) pretreatment on the hepatotoxicity of acute high dose EGCG. Experimental Biology. San Diego, CA; Gamma Sigma Delta Expo. University Park, PA; Graduate Research Exhibition. University Park, PA.

AWARDS AND HONORS

Feeding Tomorrow Kerry Graduate Student Award – Institute of Food Technologists, 2015 Donald Josephson and Stuart Patton Mentorship Award in Dairy and Food Science, 2015 College of Agricultural Sciences Graduate Student Competitive Grant Program, 2014 Robert D. and Jeanne L. McCarthy Memorial Graduate Teaching Award and Graduate Scholarship, 2014 3rd Place, Gamma Sigma Delta Graduate Poster Competition, 2014 Donald Josephson and Stuart Patton Mentorship Award in Dairy and Food Science, 2013 1st Place, Graduate Research Exhibition, 2013