Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

157
Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids Prevent Mammary Tumor Development by Jiajie Liu A Thesis presented to The University of Guelph In partial fulfillment of requirements for the degree of Master of Science in Human Health and Nutritional Sciences Guelph, Ontario, Canada © Jiajie Liu, August, 2015

Transcript of Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

Page 1: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids Prevent

Mammary Tumor Development

by

Jiajie Liu

A Thesis

presented to

The University of Guelph

In partial fulfillment of requirements

for the degree of

Master of Science

in

Human Health and Nutritional Sciences

Guelph, Ontario, Canada

© Jiajie Liu, August, 2015

Page 2: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

ABSTRACT

PLANT- AND MARINE-DERIVED N-3 POLYUNSATURATED FATTY ACIDS

PREVENT MAMMARY TUMOR DEVELOPMENT

Jiajie Liu Advisor:

University of Guelph, 2015 Dr. David WL Ma

Marine-derived n-3 polyunsaturated fatty acids (PUFA) are shown to inhibit

mammary carcinogenesis. However, evidence regarding α-linolenic acid (ALA), a plant-

based and major n-3 PUFA in Western diet, remains equivocal. This study examined the

inhibitory potency of plant- versus marine-derived n-3 on mammary tumorigenesis. Female

MMTV-neu(ndl)YD5 mice were lifelong exposed to one of four oil diets: 1) 10% safflower

(n-6, control), 2) 10% flaxseed, or 7% safflower plus either 3) 3% flaxseed, or 4) 3%

menhaden. Compare to control, 10% flaxseed and 3% menhaden oil had similar inhibitory

effects, which significantly reduced terminal end buds, and decreased overall tumor

outcomes by regulating tumor protein expression (p<0.05). A significant dose-dependent

reduction on tumor outcomes were observed in mice fed 3% and 10% flaxseed oil (p<0.05).

However, 3% flaxseed had weaker inhibitory potency compared to 3% menhaden oil

(p<0.05). Overall, marine-based n-3 PUFA are more potent than plant-derived ALA in

mitigating mammary tumorigenesis.

Page 3: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

iii

ACKNOWLEDGEMENTS

First and foremost, I would like to thank my parents for supporting my all the way

through my undergraduate and master studies. Without your support, I cannot study

abroad and have different life experiences on the other side of the world. Thank you so

much for the encouragement and accompanying me through the good and bad days. None

of this would have been possible without your love and patience. I love you.

Second, I would like to express my deep and sincere gratitude to my supervisor,

Dr. David Ma for his advice, support and guidance throughout the entirety of this project.

I would like to thank him for giving me the incredible opportunity to work in his lab and

conduct this research project. It has been an unforgettable experience. To my committee

members, Dr. Lindsay Robinson and Dr. Krista Power, thank you for your counsel

throughout this process.

A big thank you to my wonderful lab mates, past and present, with whom I spent

many of my working hours over the past two years:

• Lyn Hillyer---Thank you so much for taking the Ma Lab under your motherly

wing, for managing to take care of all of us, from protocols to statistics to treats in

the lab. I am truly thankful for your extensive guidance and patience.

• Mike Leslie, Daniel Cohen, Elie Chamoun, Salma Abdelmagid, Chris Pinelli---

Thank you so much for the lab training, editing my report and making me laugh

all the time. You gave me lots of happy moments during these two years although

I still don't understand your jokes sometime. Love you all.

Last but not least, thank you to Dr. Chaowu, Dr. Tyler Avis and Dr. Veronic

Bezaire, who always made time to chat, advice, write reference letters and put me in

contact whenever research opportunities arise. It was a great privilege to get to know you

as mentors and friends.

My thesis work was funded by Canadian Institutes of Health Research and

Ontario Graduate Scholarship.

Page 4: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

iv

TABLE OF CONTENTS

Title Page

Abstract

Acknowledgements ........................................................................................................................ iii

Table of Contents ........................................................................................................................... iv

List of Tables ................................................................................................................................. ix

List of Figures ................................................................................................................................. x

List of Abbreviations ..................................................................................................................... xi

Chapter 1. Literature Review: Individual protective effect of n-3 Polyunsaturated

Fatty acids on Breast Cancer Development ............................................................ 1

1.1 Abstract .............................................................................................................................. 2

1.2 Introduction ........................................................................................................................ 2

1.3 The effects of n-3 PUFA in Human Breast Cancer studies ................................................ 4

1.4 PUFA – Potential Mechanisms of Action ....................................................................... 11

1.4.1 Influence on Cell Plasma Membrane Composition ............................................... 11

1.4.2 Inhibition of Arachidomic Acid Derived Eicosanoid Biosynthesis ....................... 12

1.4.3 Influence on Gene Expression and Signaling Transduction ................................... 13

1.5 The Effect of n-3 PUFA Mixtures on Breast Cancer Development ................................ 19

1.5.1 Animal Models ........................................................................................................ 19

1.5.1.1 Breast Cancer Studies in Xenograft Rodent Models ................................. 19

1.5.1.2 Breast Cancer Studies in Transgenic Rodent Models ................................ 20

1.5.1.3 Breast Cancer Studies in Chemically-Induced Rodent Models ................. 24

1.5.2 Cell Culture Studies ................................................................................................ 25

1.6 The effect of Individual n-3 PUFA on Breast Cancer Development ............................... 31

Page 5: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

v

1.6.1 ALA and Breast Cancer .......................................................................................... 31

1.6.1.1 Inefficient Conversion from ALA to EPA and DHA ................................. 31

1.6.1.2 Individual Effect of ALA on Breast Cancer............................................... 32

1.6.2 Individual Effect of EPA on Breast Cancer ............................................................. 34

1.6.3 Individual Effect of DHA on Breast Cancer ........................................................... 35

1.7 Plant-Derived n-3 (ALA) vs. Marine-Based n-3 (EPA, DHA) ........................................ 37

1.8 Conclusion ........................................................................................................................ 38

Chapter 2. Updated Mini Literature Review - Role of Flaxseed Oil in the Prevention

and Treatment of Breast Cancer ............................................................................ 43

2.1 Introduction ...................................................................................................................... 44

2.2 Role of Flaxseed in Mammary Gland Development and Breast Cancer Risk ................. 45

2.2.1 Animal Study............................................................................................................ 45

2.2.1.1 Early Life Exposure ..................................................................................... 45

2.2.1.2 Adulthood Exposure Once Breast Cancer is Established ............................ 47

2.2.2 Cell Culture Study .................................................................................................... 49

2.2.3 Human Observational Study .................................................................................... 50

2.2.4 Human Intervention Study ....................................................................................... 50

2.3 Role of Flaxseed Oil in Mammary Gland Development and Breast Cancer Risk ........... 51

2.3.1 Animal Study............................................................................................................ 51

2.3.1.1 Early Life Exposure ..................................................................................... 51

2.3.1.2 Adulthood Exposure Once Breast Cancer is Established ............................ 52

2.3.2 Cell Culture Study .................................................................................................... 55

2.3.3 Human Observational Study .................................................................................... 56

2.4 Conclusion and Future Directions .................................................................................... 56

Page 6: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

vi

Chapter 3: Rationale, Hypothesis, Objective and Experimental Design ............................... 63

3.1 Rationale .......................................................................................................................... 64

3.2 Mouse Model: MMTV-neu (ndl) YD5 ............................................................................ 64

3.3 Hypothesis ........................................................................................................................ 66

3.4 Objective .......................................................................................................................... 66

3.5 Experimental Design ........................................................................................................ 66

Chapter 4. Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids Prevent

Mammary Tumor Development ............................................................................. 68

4.1 Abstract ............................................................................................................................ 69

4.2 Introduction ...................................................................................................................... 70

4.3 Material and Methods....................................................................................................... 72

4.3.1 Animals and Diets ................................................................................................... 72

4.3.2 Food Intake, Body Weights, Puberty Onset ........................................................... 73

4.3.3 Early Mammary Gland Developmental Period (6-week-timepoint) ...................... 76

4.3.3.1 Euthanization and Tissue Collection .......................................................... 76

4.3.3.2 Mammary Gland Whole Mounts................................................................ 76

4.3.3.3 Counting of TEB structures ....................................................................... 77

4.3.4 Mammary Tumor Developmental Period (20-week-timepoint) ........................... 77

4.3.4.1 Tumor Palpation ........................................................................................ 77

4.3.4.2 Euthanization and Tissue Collection ......................................................... 78

4.3.4.3 Fatty Acid Anlysis ..................................................................................... 78

4.3.4.4 Western Blot Analysis ............................................................................... 80

4.3.4.5 ELISA........................................................................................................ 81

4.3.4.6 Immunohistochemistry .............................................................................. 81

Page 7: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

vii

4.3.5 Statistical Analysis ............................................................................................... 82

4.4 Results .............................................................................................................................. 83

4.4.1 Food Intake, Body Weight Gain and Puberty Onset ............................................... 83

4.4.2 TEB Structure in 6-week-old Mice ......................................................................... 85

4.4.3 Tumor Latency and Tumor Free Status................................................................... 88

4.4.4 Tumor Volume ........................................................................................................ 92

4.4.5 Tumor Multipicity ................................................................................................... 92

4.4.6 Final Total Tumor Weight....................................................................................... 93

4.4.7 Fatty Acid Composition .......................................................................................... 94

4.4.7.1 Fatty Acid Composition in Serum ................................................................ 94

4.4.7.2 Fatty Acid Composition in Mammary Gland ............................................... 94

4.4.7.2 Fatty Acid Composition in Mammary Tumors ............................................ 97

4.4.8 Tumor Protein Analysis ......................................................................................... 102

4.4.8.1 Her2 and pHer2 ........................................................................................... 102

4.4.8.2 Akt and pAkt ............................................................................................... 103

4.4.8.3 Ki67 and Cleaved-caspase-3 ....................................................................... 104

4.5 Discussion ....................................................................................................................... 107

4.5.1 Role of Plant- and Marine-derived n-3 PUFA in Mammary Gland

Development and Breast Cancer Risk .................................................................... 107

4.5.2 Role of Plant- and Marine-derived N-3 PUFA in Mammary Tumor

Development ........................................................................................................... 109

4.5.3 Potential Mechanisms of Anti-tumorigenic Actions .............................................. 111

4.5.3.1 Change Membrane Fatty Acid Composition ............................................. 111

4.5.3.2 Modulate Cellular Signaling Molecules .................................................... 113

4.5.4 Physiological Relevance to Human Intake............................................................. 116

Page 8: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

viii

4.6 Conclusion ...................................................................................................................... 117

Chpater 5. General Discussion and Future Directions .......................................................... 118

5.1 General Doscission ......................................................................................................... 119

5.2 Limitations ...................................................................................................................... 120

5.3 Future Directions ............................................................................................................ 121

5.4 Concluding Remarks ...................................................................................................... 122

Chapter 6. References and Appendix...................................................................................... 123

Page 9: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

ix

LIST OF TABLES

Table 1.1 N-3 PUFA and breast cancer risk: prospective cohort studies ....................................... 6

Table 1.2 N-3 PUFA and breast cancer risk: case-control studies ............................................. 7-8

Table 1.3 N-3 PUFA and breast cancer risk: xenograft rodent models ....................................... 22

Table 1.4 N-3 PUFA and breast cancer risk: transgenic rodent models ...................................... 23

Table 1.5 N-3 PUFA and breast cancer risk: chemically-induced rodent models .................. 27-28

Table 1.6 N-3 PUFA and breast cancer risk: cell culture studies ........................................... 29-30

Table 1.7 Individual role of ALA, EPA and DHA on BC ...................................................... 39-41

Table 1.8 Individual effect of ALA, EPA and DHA on different types of BC ........................... 42

Table 2.1 Early exposure to flaxseed and breast cancer risk ....................................................... 57

Table 2.2 Late life exposure to flaxseed and breast cancer risk ............................................. 58-59

Table 2.3 Late life exposure to flaxseed oil and breast cancer risk ........................................ 60-62

Table 4.1 Composition of purified diets ...................................................................................... 74

Table 4.2 Fatty acid composition of diets .................................................................................... 75

Table 4.3 Serum fatty acid composition of mice at 6 or 20 weeks of age ................................... 95

Table 4.4 Fatty acid composition of MG phospholipids ............................................................... 98

Table 4.5 Fatty acid composition of mammary tumor phospholipids ........................................ 101

Page 10: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

x

LIST OF FIGURES

Figure 1.1 Synthetic pathways of long-chain PUFA and eicosanoids .......................................... 14

Figure 1.2 Hypothetical scheme showing how n-3 PUFA modulates cell fuctions ..................... 15

Figure 4.1 Mice body weight changes throughout 3 to 20 weeks of age...................................... 84

Figure 4.2 Average pubertal onset ................................................................................................ 85

Figure 4.3 Representative stereoscopic wholemount images ....................................................... 86

Figure 4.4 Average total number and density of terminal end buds at 6 weeks of age ................ 87

Figure 4.5 Average tumor latency ................................................................................................ 88

Figure 4.6 The proportion of mice tumor free throughout 20-week study ................................... 89

Figure 4.7 Tumor volume. ............................................................................................................ 90

Figure 4.8 Tumor multiplicity....................................................................................................... 91

Figure 4.9 Final total tumor weight .............................................................................................. 93

Figure 4.10 Mammary gland fatty acid composition .................................................................... 96

Figure 4.11 Mammary Tumor fatty acid composition. ............................................................... 100

Figure 4.12 Effect of plant- versus marine-derived n-3 PUFA on tumor protein

expression of HER-2 and pHER-2 .......................................................................... 102

Figure 4.13 Effect of plant- versus marine-derived n-3 PUFA on tumor protein

expression of total Akt and pAkt

Ser 473 ..................................................................... 103

Figure 4.14 Effect of plant- versus marine-derived n-3 PUFA on tumor protein

expression of Ki-67 and cleaved-caspase-3 ............................................................. 104

Figure 4.15 The photomicrographs of representative immunohistochemical stained

mammary tumor section with Ki67 ......................................................................... 105

Figure 4.16 The photomicrographs of representative immunohistochemical stained

mammary tumor section with cleaved-caspase-3 .................................................... 106

Page 11: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

xi

LIST OF ABBREVIATIONS

AA: arachidonic acid

AB: alveolar buds

ALA: alpha-linolenic acid

Akt: protein kinase B

BC: breast cancer

BMI: body mass index

COX: cyclooxygenase

DHA: docosahexaenoic acid

DMBA: 7, 12-dimethylbenz(a)anthracene

EPA: eicosapentaenoic acid

FASN: fatty acid synthase

GC: gas chromatography

HER-2: human epidermal growth factor-2

LA: linoleic acid

5-LOX: 5-lipoxygenase

LTB4: leukotriene B4

MG: mammary gland

MMTV: mouse mammary tumor virus

MUFA: monounsaturated fatty acids

MNU: N-methyl-N-nitrosourea

NFĸB: nuclear factor kappa B

N-3 PUFA: omega-3 polyunsaturated fatty acids

N-6 PUFA: omega-6 polyunsaturated fatty acids

Page 12: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

xii

pAkt: phosphorylated- protein kinase B

PCR: polymerase chain reaction

PC: phosphatidylcholine

PE: phosphatidylethanolamine

PGE2: prostaglandin E2

PPARγ: peroxisome proliferator-activated receptor gamma

PUFA: polyunsaturated fatty acids

TEB: terminal end bud

TG: transgenic

WT: wild type

Page 13: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

1

Chapter One

Literature Review

Individual Protective Effect of n-3 Polyunsaturated Fatty Acids on

Breast Cancer Development

Published as an open access article in Nutrients 2014, 6:5184-5223- “The Role of n-3

Polyunsaturated Fatty Acids in the Prevention and Treatment of Breast Cancer” by Jiajie

Liu and David W.L. Ma

Page 14: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

2

1.1 Abstract

Breast cancer (BC) is the most common cancer among women worldwide.

Dietary fatty acids, especially n-3 polyunsaturated fatty acids (PUFA), are believed to

play a role in reducing BC risk. Evidence has shown that fish consumption or intake of

long-chain n-3 PUFA, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid

(DHA), are beneficial for inhibiting mammary carcinogenesis. The evidence regarding α-

linolenic acid (ALA), however, remains equivocal. It is essential to clarify the relation

between ALA and cancer since ALA is the principal source of n-3 PUFA in the Western

diet and the conversion of ALA to EPA and DHA is not efficient in humans. In addition,

the specific anticancer roles of individual n-3 PUFA, alone, have not yet been identified.

Therefore, the present review evaluates ALA, EPA and DHA consumed individually as

well as in n-3 PUFA mixtures. Also, their role in the prevention of BC and potential

anticancer mechanisms of action are examined. Overall, this review suggests that each n-

3 PUFA has promising anticancer effects and warrants further research.

1.2 Introduction

Breast cancer (BC) is a major health problem among women worldwide, and is

the second leading cause of death for women in Canada and the United States (1, 2). On

average, 65 Canadian women will be diagnosed with BC per day, with 1 in 9 females

expected to develop BC in their lifetime (1, 2). Both genetic and environmental factors

are believed to play a role in a woman’s risk of developing BC (3, 4). Most anticancer

drugs, developed to date, aim to kill cancer cells and decrease tumor burden but are

relatively ineffective against some phases of tumorigenesis (5, 6). Thus, alternate

strategies to prevent tumorigenesis are urgently required. In the past few decades,

epidemiological studies have suggested that a healthy diet and lifestyle are critical for the

prevention of BC. Dietary fatty acids are one of the most intensively studied dietary

factors (7-9).

Saturated fatty acids (SFA), monounsaturated fatty acids (MUFA) and trans fatty

acids (TFA) have been found to increase cancer risk; while specific polyunsaturated fatty

acids (PUFA) are indicated to have anticancer effects (9, 10). There are two major classes

Page 15: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

3

of PUFA: n-6 PUFA and n-3 PUFA. In mammals, n-6 and n-3 PUFA are both essential

fatty acids for health and must be consumed as part of the diet because they cannot be

endogenously synthesized (11). Linoleic acid (LA, 18:2n-6) and arachidonic acid (AA,

20:4n-6) are the two most common n-6 PUFA in typical Western diets; LA can be found

in some plant oils such as corn and safflower oils, and AA usually comes from dietary

animal sources or can be synthesized from LA (12, 13). α-linolenic acid (ALA, 18:3n-3)

is the precursor of then-3 PUFA family which can be further elongated and desaturated to

two important long chain n-3 PUFA, eicosapentaenoic acid (EPA, 20:5n-3) and

docosahexaenoic acid (DHA, 22:6n-3) (11). ALA is a plant-derived n-3 PUFA, which is

present in flaxseed, canola and soybean oils (14). The longer chain n-3 PUFA, EPA and

DHA can be obtained directly from marine sources such as seafood and fish oils, and are

widely known for their cardioprotective benefit (13). ALA is the major n-3 PUFA

consumed in the Western diet, whereas intakes of EPA and DHA are typically low. It is

estimated that the typical North American diet provides approximately 1.4 g of ALA and

0.1–0.2 g of EPA plus DHA per day (15). With regard to dietary reference intake of n-3

PUFA, the Institute of Medicine (IOM) recommends since 2005 a daily intake 1.1 g ALA

for women and 1.6 g ALA for men to prevent some chronic diseases, and up to ten

percent of this can be consumed as EPA and/or DHA (16). While current intakes meet

IOM recommendations, in a 2014 report by the Academy of Nutrition and Dietetics, 500

mg EPA plus DHA per day is required for the general healthy adult population (17).

Results from both in vivo and in vitro studies suggest that n-6 PUFA accelerate

tumorigenesis, in contrast, n-3 PUFA may have anticancer effects (18-22). Western diets

are typically deficient in n-3 PUFA and high in n-6 PUFA compared with traditional

Asian diets (8, 23, 24). Migration studies have shown that Asian women, who typically

have a lower rate of BC and higher fish consumption exhibited an increased incidence of

BC within one generation after migration to the Western countries (24, 25). Historically,

the intake of n-6 and n-3 PUFA has been estimated to be approximately equal. However,

in recent years, the content of Western diets has significantly increased in n-6 PUFA

resulting in an increase in the n-6/n-3 PUFA ratio (8). Excessive amounts of n-6 PUFA

and a very high n-6/n-3 ratio (16:1 or higher), as is currently found in Western diets, have

been suggested to promote the pathogenesis of many diseases such as cardiovascular

Page 16: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

4

disease, autoimmune diseases and some types of cancer; whereas increased levels of n-3

PUFA (a low n-6/n-3 ratio) have been shown to exert suppressive effects (8). However, it

remains to be resolved whether it is simply the reduced n-3 PUFA or the changing n-6/n-

3 ratio that is relevant to these outcomes.

Substantial evidence from cell culture and rodent studies indicate that increased

fish consumption or intake of n-3 PUFA inhibits BC cell proliferation and reduces BC

risk relative to n-6 PUFA (26-30). Nevertheless, there has been longstanding controversy

in epidemiological and observational studies regarding the potential anticancer effects

of n-3 PUFA due to the inability to show causality. Previous published reviews have

focused on the role of fish and marine n-3 fatty acids in BC prevention, whereas the

evidence for ALA and individual effects of long chain n-3 PUFA is lacking (3, 8, 11, 23,

31, 31, 31-34). Since a typical North American diet is mainly comprised of ALA as the

source of n-3 PUFA, it is necessary to elucidate the specific effects of ALA and cancer

risk. Therefore, the purpose of the present review is to evaluate the preventative role of

ALA, EPA and DHA in BC development when consumed individually, as well as in n-3

PUFA mixtures through dietary and supplemental forms. In addition, the potential

mechanisms by which they exert anticancer effects will also be discussed.

1.3 The Effects of n-3 PUFA in Human BC Studies

Dietary n-3 PUFA may influence breast cancer (BC) progression and prognosis.

In the past ten years, six prospective cohort studies (Table 1.1) and nine case-control

studies (Table 1.2) have examined the association between the consumption of either fish

or fish oil supplements and BC risk, showing a protective effect of n-3 PUFA. These

studies were conducted in many different geographic areas with mixed findings. In

general, Asian populations with a low total fat intake and high fish consumption,

associated n-3 PUFA intake with a reduced risk of BC (35-39). There was also a weak

association with reduced BC risk in US studies involving women whose diets had

higher n-6 PUFA content combined with fish oil supplements (18, 40, 41). European

studies were less consistent and somewhat contradictory (42-45).

Page 17: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

5

In the Japan Collaborative Cohort (JACC) study, a significant decrease in the risk

of BC was detected in women with the highest dietary intake of fish fat and the long-

chain n-3 PUFA (35). Similar results were observed in a large prospective study of

35,298 Singapore women, indicating an inverse association between dietary n-3 PUFA

from marine sources and BC risk (36). Relative to the lowest quartile of n-3 PUFA intake,

individuals in the top three quartiles exhibited a 26% reduction in BC risk (relative risk =

0.74; 95% confidence interval = 0.58–0.94). Additionally, a recent analysis from the

VITamins And Lifestyle (VITAL) cohort carried out in US indicated that the current use

of fish oil supplements was associated with a decreased risk of localized invasive ductal

carcinomas in postmenopausal women (40).

The association between dietary intakes of n-3 PUFA or fish oil supplements with

overall survival was also examined. Patterson et al. demonstrated that women with higher

intakes of EPA and DHA from food, but not from fish oil supplements, had a dose-

dependent reduction in all-cause mortality (46). They also showed a reduced risk of

additional BC events of approximately 25% when compared with the lowest tertile of

intake (tertile 3:hazard ratio = 0.72; 95% confidence interval = 0.57–0.90) (46). In

support of these self-reported intake studies, Zheng et al. performed a comprehensive

analysis of 21 independent prospective cohort studies and found that marine n-3 PUFA

were associated with a 14% risk reduction of BC, and the relative risk remained similar

whether marine n-3 PUFA was measured as dietary intake or as tissue biomarkers (7).

Further, a dose-response analysis indicated a 5% lower risk of BC per 0.1 g/day (0.95,

0.90 to 1.00, I2 = 52%) increment of dietary marine n-3 PUFA (7). Conversely, a French

study comprising over 56,000 women found no association between total n-3 or n-6

PUFA intake and BC risk (42). Also, a large study of postmenopausal women in

Denmark concluded that increased fish consumption was associated with elevated

incidence rates of BC, but this association was present only for development of estrogen

positive BC (43). These null studies were mostly conducted in European populations with

relatively low per capita intake of n-3 PUFA (32, 44).

Page 18: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

6

Table 1.1 n-3 PUFA and breast cancer risk: Prospective cohort studies.

Year Country Subjects Method of

Assessment n-3/n-6 PUFA Source BC Risk Ref

2005 Japan

26,291 women

40–79 years

129 BC cases FFQ

1

Animal and fish fat, vegetable

oil, SFA, MUFA and PUFA ↑ fish fat, EPA + DHA ↓ BC risk (35)

2003 Singapore

35,298 women

45–74 years

342 BC cases FFQ

Fish/shellfish, saturated,

monounsaturated and

polyunsaturated fat

↑ n-3 PUFA from fish/shellfish ↓ BC risk

↑ n-6 PUFA (low marine n-3) ↑ BC risk (36)

2010 US

35,016

postmenopausal

50–76 years

880 BC cases

FFQ Dietary fish oil supplement ↑ fish oil ↓ risk of invasive ductal

carcinomas (40)

2009 France

56,007 women

40–65 years

1650 BC case

FFQ

ALA and n-6 PUFA from fruit,

nuts and vegetable oils; Long

chain n-3 PUFA from meals

No association between total n-3 and BC risk

↑ ALA ↓BC risk

↑ long chain n-3 PUFA ↓ BC risk (at highest

quintile of n-6 PUFA)

(42)

2003 Denmark

23,693

postmenopausal

50–64 years

424 BC cases

FFQ Fish ↑ intake of fish ↑ ER+ BC incidence (43)

2011 China

72,571 women

40–70 years

712 BC cases

FFQ Fish, marine-derived

n-3 PUFA red meat ↑ n-6/n-3 PUFA ratio ↑ BC risk (37)

1 FFQ: food frequency questionnaire; ↑: increase; ↓: decrease

Page 19: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

7

Table 1.2 n-3 PUFA and breast cancer risk: Case-control studies.

Year Country Subjects

Characteristics

Method of

Assessment n-3/n-6 PUFA Source BC Risk Ref

2007 Japan 103 incident BC cases

309 controls

erythrocyte

membrane

FFQ

dietary food intake

including soy and meat

products, fish and other

seafood, vegetables

↑ dietary intake of n-3 fatty acids ↓ BC risk

↑ long chain n-3 PUFA in erythrocyte ↓ BC risk

↑ saturated fatty ↑ BC risk

(46)

2007 China 322 incident BC cases

1030 controls

erythrocyte

membrane ↑ total n-3 fatty acids and EPA ↓ BC risk (47)

2009 China

155 NPFC 1

185 PFC 2

241 BC, 1030 controls

erythrocyte

membrane

FFQ

dietary food intake

↑ EPA ↓ risk of NPFC

↓ progression of PFC to BC

↑ γ-linolenic acid ↑ risk of NPFC, PFC and BC

(38)

2002 US 73 BC patients

74 controls

breast

adipose tissue

↑ EPA and DHA ↓ n-6/n-3 PUFA ratio ↓ BC

risk

↑ n-6 PUFA ↑ BC risk

(18)

1 Benign proliferative fibrocystic conditions (PFC);

2 non-proliferative fibrocystic conditions (NPFC);

↑: increase; ↓: decrease.

Page 20: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

8

Table 1.2 n-3 PUFA and breast cancer risk: Case-control studies (continue)

Year Country Subjects

Characteristics

Method of

Assessment

n-3/n-6 PUFA

Source BC Risk Ref

2003 US 565 incident BC

554 controls FFQ

Daily fat

intake

↓ n-6/n-3 PUFA ratio ↓ BC risk (premenopausal)

↑EPA, DHA ↓ BC risk (21% and 18%,

respectively)

(41)

2009 Denmark 463 BC cases

1098 controls

Gluteal

adipose tissue

biopsy

Dietary food

intake

No association between total or individual marine

n-3 PUFA in adipose tissue and risk of BC (44)

2002 France

241 invasive BC cases

88 controls-benign

breast disease

Breast

adipose tissue ↑ ALA ↑ DHA ↓ n-6/n-3 PUFA ratio ↓ BC risk (45)

2012 Mexican 1000 incident BC cases

1074 controls

Interview

and FFQ

Dietary food

intake

↑ n-3 PUFA ↓ BC risk (obese women)

↑ n-6 PUFA ↑ BC risk (premenopausal) (19)

Page 21: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

9

In order to examine the relationship between n-3 PUFA exposure and BC risk,

several case-control studies have been conducted using different biomarkers (Table 1.2).

Kuriki et al. investigated the fatty acid compositions of erythrocyte membranes as a

biomarker and demonstrated that BC risk exhibited a significant inverse association with

dietary intake of n-3 PUFA derived from fish and high levels of long-chain n-3 PUFA in

erythrocyte membranes (47). Another assessment of erythrocyte fatty acid composition

found the inverse association significant only for EPA and total n-3 PUFA content (48).

Furthermore, Shannon et al. evaluated the role of n-3 and n-6 PUFA in the development

of benign proliferative fibrocystic conditions (PFC) and non-proliferative fibrocystic

conditions (NPFC) in the breast (38). They showed that women in the highest quartile of

erythrocyte EPA concentrations were 67% less likely to have NPFC alone or with BC,

and EPA significantly lowered the risk of progressing from PFC to BC by 43% (38).

However, γ-linolenic acid (n-6 PUFA) was found to be positively associated with nearly

all conditions (38). These results were consistent with an earlier meta-analysis showing

that total and individual n-3 PUFA, especially EPA and DHA, play a protective effect

against BC, while total SFA, MUFA, palmitic and oleic acids were associated with

increased BC risk (9).

In a Korean case control study, 358 patients with BC and 360 healthy controls

underwent dietary assessment by questionnaire and interview to determine their dietary

consumption of fish and n-3 PUFA derived from fish. Both pre- and postmenopausal

women in the highest quartile of fatty-fish intake had a lower incidence of BC (odds ratio

OR = 0.23, 95% CI = 0.13–0.42; p < 0.001), but the protective effect of EPA and/or DHA

intake was only observed for postmenopausal women (39). These findings were similarly

observed in a study of Mexican women where BC risk was lower in obese women (BMI

≥ 30) with high n-3 PUFA intake, not in women of normal weight (19). In contrast, a

case-cohort study of Danish women did not find any association between either total or

individual marine n-3 PUFA intake or BC risk (44). As the total levels of marine n-3

PUFA intake were low in Europe, this may account for observed discrepancies relative to

populations consuming marine-rich diets (32, 44, 45).

Page 22: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

10

Other population studies have investigated interactions between n-3 and n-6

PUFA. Quantifying consumption of both types of fatty acids is an essential step in

isolating n-3 PUFA specific effects. The study of Bagga et al. showed that excessive

intake of n-6 PUFA contributed to the high risk of BC in US, while a decreased risk of

BC development was accompanied with higher EPA and DHA consumption (18). A

similar inverse relationship was also observed in regard to the n-6/n-3 PUFA ratio. In

another US study, when the analysis was restricted to pre-menopausal women, the

consumption of the lowest ratio of n-6 to n-3 was associated with a 41% reduction of BC

risk, although it was not significant (41). This observation was also observed in studies

conducted in China and France, although there was no association between n-3 PUFA

intake and BC risk, low n-3 PUFA intake by women who had the highest n-6 PUFA was

correlated with elevated BC risk (37, 45). However, based on the ratio, it is not possible

to determine whether it is increased n-6 or decreased n-3 PUFA that is the causal driver

of BC risk. Nevertheless, these studies indicate the necessity of higher n-3 PUFA intakes

given that n-6 intakes are adequate in all populations, and thus heightening the potential

value of n-3 PUFA as effective agents against BC.

Diet intervention by n-3 PUFA supplements as a mean of decreasing BC risk in

women still needs to be tested clinically. To date, few human intervention studies have

assessed the effectiveness of n-3 PUFA in BC prevention and treatment. One randomized

clinical trial tested the combined effects of n-3 PUFA (EPA + DHA = 3.36 g/day, 2 year)

and Raloxifene (anti-estrogen) in reducing risk of BC in postmenopausal women (49).

Although the plasma n-6/n-3 ratio significantly decreased among subjects after n-3 PUFA

intervention compared with the subjects without intervention, n-3 PUFA administration

did not affect any selected biomarkers that associated with BC risk (49). While in another

human intervention study, Thompson et al. demonstrated that daily intake of 25 g

flaxseed (ALA = 57% of total fatty acids) can significantly reduce cell proliferation and

increase cell apoptosis in tumors of postmenopausal BC patients. These limited results

provide encouragement for future study of n-3 PUFA as an adjuvant therapy in BC (50).

Page 23: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

11

1.4 PUFA—Potential Mechanisms of Action

For more than 30 years, numerous studies have attempted to establish whether

there is a causal relationship between n-3 PUFA ingestion and a reduction in mammary

carcinogenesis. Mounting evidence shows that dietary n-3 PUFA may exert an anti-

carcinogenic action by altering the composition of cell membrane phospholipids,

inhibiting AA metabolism and decreasing AA derived eicosanoids, as well as modulating

the expression and function of numerous receptors, transcription factors and lipid derived

signaling molecules. However, studies of the effects of dietary n-3 PUFA on BC

progression and prognosis are limited in humans. Epidemiological studies do not allow

for the analysis of important cellular interactions and the specific molecular pathways

which are activated during the course of tumor initiation and cancer progression in the

mammary gland. Thus, the use of animal models (in vivo) and BC cell lines (in vitro)

provide crucial avenues for improving our understanding of the underlying biological

pathways involved in trigger BC development and potential therapeutic approaches for

BC treatment and prevention. Here we first introduce some potential mechanisms and

important downstream mediators that involved in the anticancer action of n-3 PUFA.

1.4.1 Influence on Cell Plasma Membrane Composition

Fatty acids play an important role in membrane biogenesis in the form of

glycerophospholipids, a major class of lipids found all cell membranes (51). Dietary

PUFA integrate into plasma membrane glycerophosholipids and influence the fatty acid

composition (52). The sn-1 position on the glycerol backbone of glycerophospholipids is

usually linked to saturated fatty acids, and the sn-2 position is linked to an n-6 PUFA

such as AA. Increased intake of dietary n-3 PUFA may replace n-6 with n-3 fatty acids at

the sn-2 position of glycerophospholipids (52). Since n-3 PUFA has a greater density

compared to n-6 PUFA, the aggregation of n-3 fatty acids tend to be closer to the lipid-

water interface of the membrane. This characteristic can significantly affect plasma

membrane fluidity and permeability (53). In addition, due to the high level unsaturation

of long chain n-3 PUFA, they have very poor affinity for cholesterol (52, 54). Membrane

Page 24: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

12

cholesterol serves as a spacer for the hydrocarbon chains of sphingolipids and maintains

the assembled microdomains of lipid rafts (55). Thus, cholesterol depletion leads to the

disorganization of lipid raft structure (54). Lipid rafts are important membrane domains

for cell signaling since it is enriched with many regulatory proteins and some growth

factor receptors(54, 55). As a result, the incorporation of n-3 PUFA, especially EPA and

DHA, can disturb formation of lipid rafts and suppress raft-associated cell signal

transduction (22, 56, 57).

1.4.2 Inhibition of Arachidonic Acid (AA) Derived Eicosanoid Biosynthesis

One of the key cellular functions of PUFA is related to their enzymatic

conversion into eicosanoids. Eicosanoids are short-lived, hormone-like lipids, typically

comprised of 20 carbon atoms, which play a critical role in platelet aggregation, cellular

growth and cell differentiation (31). The most salient mechanism by which n-3 PUFA

reduce tumor development is through inhibiting the synthesis of inflammatory

eicosanoids derived from AA (31). As indicated in Figure 1.1, firstly, both ALA and LA

are initially converted to their long-chain metabolites (EPA and AA, respectively)

through the same desaturation/elongation pathway; therefore, there exists potential

competition between these two families of fatty acids for desaturases and elongases. The

initial conversion of ALA to stearidonic acid (18:4n-3) is the rate limiting reaction of the

pathway. The affinity of delta 6-desaturase for ALA is greater than for LA (58). As a

result, a higher intake of ALA reduces the synthesis of AA from LA and thus, less AA is

available for synthesis of inflammatory eicosanoids (59). Secondly, increase consumption

of n-3 PUFA results in their incorporation into membrane phospholipids, where they

partially replace AA, therefore reducing the substrate for AA derived eicosanoids. Healy

et al. showed that dietary supplements with four different concentrations of fish oil

resulted in the incorporation of EPA and DHA into human inflammatory cells occurs in a

dose-response fashion at the expense of AA (60). Thirdly, both AA and EPA are

substrate for eicosanoid synthesis such as prostaglandins (PG) and leukotrienes (LT) (31).

AA can be metabolized by two major pathways including the cyclooxyenase (COX) and

Page 25: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

13

lipoxygenase (LOX) pathways. COX-2 catalyzes the rate limiting step in the formation of

2-series PGs. 5-LOX catalyzes the first step in oxygenation of AA to produce hydroxyl

derivatives and 4-series LTs. The overexpression of COX-2 has been detected in many

types of cancers and PGE2 has been shown to promote cell proliferation in mammary

tumor tissues (61). PGE2 stimulates the expression and activation of aromatase, the

enzyme that converts androgens to estrogens (62). Therefore, it is hypothesized that

estrogen levels can be lowered by decreasing n-6 PUFA intake. In contrast, EPA

metabolism results in 3-series PGs and 5-series LTs, with a slightly different structure

and anti-tumorigenic properties (58). N-3 PUFA supplementation has been shown to

lower production of PGE2 (by 60%) and LTB4 (by 75%) in human peripheral blood

mononuclear cells (63). Furthermore, n-3 PUFA has been suggested to suppress the

expression of COX-2 and 5-LOX. Feeding mice with high n-3 PUFA diets influences

mammary tumor development by down-regulating COX-2 and 5-LOX expression (20,

64). In addition to the inhibitory effects on the generation of inflammatory eicosanoids,

recent studies have identified a novel group of lipid mediators, termed resolvins, which

are formed from EPA and DHA (65, 66). These mediators appear to exert potent anti-

inflammatory actions and are considered as potential therapeutic interventions for some

chronic inflammatory diseases and cancer (67, 68).

In general, n-3 PUFA can not only block AA metabolism, but can also compete

with AA for eicosanoid synthesis. This anti-inflammatory effect of n-3 PUFA is of

interest since chronic inflammation has been linked to cancer initiation and progression

(69).

1.4.3 Influence on Gene Expression and Signaling Transduction

BC development is a multi-step process that requires the accumulation of several

genetic alterations in a single cell. Many studies have now demonstrated that the

regulation of intracellular signaling is a critical aspect of controlling cell functions in

various types of cancers. Neoplastic growth and progression are generally regarded as

dependent on a high rate of cell proliferation and a low rate of apoptosis. Dietary PUFA

Page 26: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

14

and their metabolites may exert some of their anti-cancer or tumor-promoting effects by

affecting gene expression or activating signal transduction molecules involved in the

control of cell proliferation, differentiation apoptosis and metastasis (Figure 1.2).

Figure 1.1 Synthetic pathways of long-chain PUFA and eicosanoids. α-linolenic acid

(ALA; 18:3n-3) and linoleic acid (LA; 18:2n-6) are essential PUFA obtained from the

diet, and involve in similar sequential desaturation and elongation steps, give rise to long

chain, more unsaturated PUFA eicosapentaenoic acid (EPA; 20:5n-3), docosahexaenoic

acid (DHA; 22:6n-3), and arachidonic acid (AA; 20:4n-6). Relevant intermediates in

these pathways include SDA (stearidonic acid), ETA (eicosatetraenoic acid), DPA

(docosapentaenoic acid), GLA (γ-linolenic acid), DGLA (dihomo-γ-linolenic acid) and

AdA (adrenic acid). Both AA and EPA are substrates for the synthesis of eicosanoid

products such as prostaglandins (PG) and leukotrienes (LT). The products of n-6 PUFA

tend to promote cell proliferation while the products of n-3 PUFA have anti-tumorigenic

properties. N-3 PUFA may lower the risk of BC by disrupting the biosynthesis of AA-

derived inflammatory eicosanoids.

Dietary n-3 PUFA

ALA (18:3 n-3)

SDA (18:4 n-3)

ETA (20:4 n-3)

EPA (20:5 n-3)

DPA (22:5 n-3)

DHA (22:6 n-3)

LA (18:2 n-6)

GLA (18:3 n-6)

DGLA (20:3 n-6)

AA (20:4 n-6)

AdA (22:4 n-6)

DPA (22:5 n-6)

Dietary n-6 PUFA

COX-2 ↑

5-LOX↑

∆6-desaturase

elongase

∆5-desaturase

elongase

∆6-desaturase

Prostaglandin-3-series

Leukotrienes-5-series Prostaglandin-2-series

Leukotrienes-4-series Resolvins (E-series)

Resolvins (D-series)

Page 27: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

15

Figure 1.2 Hypothetical scheme showing how n-3 PUFA modulates cell functions via

intracellular signaling molecules. Cell proliferation and cell apoptosis are the two

important fundamental processes integral to carcinogenesis. n-3 PUFA exerts anti-cancer

effects by reducing the expression of some growth factors including human epidermal

growth factor receptor-2 (HER-2), epidermal growth factor receptor (EGFR) and insulin-

like growth factor 1(IGF-1R); inhibiting cell proliferation by either activating PPARγ or

decreasing levels of fatty acid synthase (FAS) protein; and promoting cell apoptosis via

blocking PI3K/Akt pathways, downregulating phosphorylated Akt, inhibiting NF-κB

activity and lowering Bcl-2/Bax ratio.

1.4.3.1 EGFR and HER-2

Among numerous factors, carcinogenesis involves the activation of oncogenes

such as the epidermal growth factor receptor (EGFR) and human epidermal growth factor

receptor-2 (HER-2). EGFR is a receptor tyrosine kinase, which plays essential roles in

regulating a number of cellular processes including cell proliferation, survival and

migration (70). EGFR is usually activated in response to extracellular ligands (epidermal

growth factor [EGF)) by its phosphorylation [54]. Dysregulated EGFR activation is often

associated with overexpression of EGFR, which has been observed in several cancer

High n-3

Low n-6

HER-2↓

IGF-1R↓

EFGR↓

FASN↓

PPARγ↑

Bcl-2↓

Bax↑

pAkt↓

NF-kB↓

PI3K↓

Page 28: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

16

types including breast carcinomas (71). Marine n-3 PUFA were able to inhibit EGFR

activity, in particular, DHA was found to induce apoptosis in BC cells by down-

regulating EGFR expression (72). Similar to EGFR, HER-2 (HER-2/neu or erbB-2) is a

185-kD transmembrane receptor tyrosine kinase that is involved in human mammary

oncogenesis (73). The overexpression of HER-2 occurs in 25%–30% of human invasive

BCs, and is associated with a more aggressive phenotype and poor patient prognosis (12,

74). HER-2 functions as a co-receptor and forms homodimers or heterodimers with

EGFR or insulin-like growth factor 1(IGF-1R) to activate downstream target signaling

cascades that involve cell survival and proliferation, such as phosphatidylinositol 3-

kinase (PI3K)/Akt, mitogen-activated protein kinase (MAPK) and inhibition of apoptotic

pathways such as Bcl-2-associated death promoter protein (75, 76). It should be noted

that the HER-2 receptors also activate lipogenic pathways mediated by the fatty acid

synthase (FAS) protein (6), a key lipogenic enzyme catalyzing the terminal steps in the

de novo biogenesis of fatty acids in cancer pathogenesis (51). Dietary n-3 PUFA were

demonstrated to inhibit the early stages of HER-2/neu-mediated mammary

carcinogenesis in rats (77). Notably, ALA alone was able to reduce HER-2 protein

expression by 79% in MCF-7 cell lines (78). Both EGFR and HER-2 are regarded as

important therapeutic targets against BC, and n-3 PUFA may be a dietary treatment for

controlling the growth factor-mediated oncogenesis.

1.4.3.2 Peroxisome Proliferator-Activated Receptor Gamma (PPARγ)

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear

receptor superfamily and function as ligand-activated transcription factors (79). PPARγ is

a subset of the PPAR family, it is mainly expressed in adipose tissue, mammary gland,

colon and the immune system (80, 81). PPARγ regulates the expression of target genes

by binding to DNA sequence elements, termed PPAR response elements (PPREs).

PPREs have been identified in the regulatory regions of a variety of genes that are

involved in lipid metabolism and homeostasis, but recently have appeared to be involved

in cell proliferation, cell differentiation, and inflammatory responses (79, 82). PPARγ

ligands include naturally occurring compounds such as PUFA and eicosanoids, as well as

synthetic activators, such as the hypolipidemic drugs (83). Clay et al. indicated that

Page 29: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

17

induction of apoptosis is a biological response resulting from PPARγ activation in some

BC cells (84). n-3 PUFA are direct agonists for PPARγ, which have been shown to exert

anti-tumorigenic effects via the activation of PPARγ (82, 85). For instance, DHA was

found to attenuate MCF-7 cell proliferation by activation of PPARγ (86). In addition,

dietary supplementation with a low ratio of n-6/n-3 PUFA (1:14.6) was shown to increase

PPARγ protein content, which was paralleled with a reduction of tumor burden in rats

with induced mammary carcinogenesis (87). As a result, PPARγ activation is beneficial

for controlling BC, which suggests a potential role for PPARγ ligands in the treatment of

BC.

1.4.3.3 Bax/Bcl-2

Apoptosis is a form of cell death triggered during a variety of physiological

conditions and is tightly regulated by a number of gene products that promote or block

cell death at different stages (88). Bcl-2 is well-known as an important apoptosis-

regulator protein (89), normally blocking apoptosis and its overexpression contributes to

BC by prolonging cell survival (90). Bax is a pro-apoptotic member of the Bcl-2 family

of proteins. It is likely to have pore-forming activity to increase mitochondrial membrane

permeability, and can also form a homodimer with Bcl-2 to enhance the effects of

apoptotic stimuli (90, 91). Raisova et al. showed that the Bax/Bcl-2 ratio determines the

susceptibility of cells to apoptosis (92). Thus, a low Bax/Bcl-2 ratio is associated with

enhanced survival of BC cells and resistance to apoptosis, and vice versa. It has been

proposed that diets rich in n-3 PUFA, such as fish and canola oil, reduces the abundance

of Bcl-2 and up-regulates Bax expression to induce apoptosis, thereby reducing BC risk

(27, 93).

1.4.3.4 PI3K/Akt, NF-κB

Besides Bcl-2, the PI3K/Akt pathway also plays an important role in cell

apoptosis. Phosphatidylinositol 3-kinase (PI3K) is a heterodimeric lipid kinase that is

composed of a regulatory and catalytic subunit that is encoded by different genes (93).

The primary consequence of PI3K activation is the generation of the second messenger

PtdIns (3,4,5) P3 (PIP3) in the membrane, which in turn recruits and activates Akt, a

Page 30: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

18

downstream serine/threonine kinase (94). Akt activation is a dual regulatory mechanism

that requires translocation to the plasma membrane and phosphorylation at Thr308 and

Ser473 (52, 94, 95). Due to this mechanism, Akt functions as an anti-apoptotic signaling

molecule. Thus, upregulation of phosphorylated Akt is relevant to tumor cell growth and

resistance to cell apoptosis (6, 87, 96). HER-2 overexpression constitutively activates

survival and proliferation pathways by increasing activation of Akt, however, n-3 PUFA

was found to either modulate total Akt expression or interact with Akt to down-regulate its

phosphorylation (6, 97). Since Akt requires translocation to the plasma membrane for

activation, it is possible that tumor cell membrane enrichment of n-3 PUFA might affect the

phosphorylation of Akt that are recruited to the membrane for activation (96). In addition,

recent studies have demonstrated that PI3K/Akt promoted cell survival is mediated, in

part, through the activation of the nuclear factor kappa-B (NF-κB) transcription factor

(95, 98, 99). NF-κB is a key regulator of genes involved in cell proliferation, migration,

and angiogenesis (100, 101). In tumor cells, impaired regulation of NF-κB activation will

lead to deregulated expression of the anti-apoptotic genes under the control of NF-κB

(100). For instance, NF-κB has been shown to inhibit the activity of p53, a tumor

suppressor known to trigger apoptosis in cells with damaged DNA (102). As a result,

constitutive NF-κB expression may contribute to the development and progression of BC.

1.4.3.5 Cell Proliferation Marker: Ki-67 and PCNA

Cell proliferation is another fundamental process integral to carcinogenesis. Ki-67

is a nuclear protein, and being widely used as a prognostic or predictive marker in BC

and other malignant disease (103). The human Ki-67 protein is present during all active

phases of the cell cycle G(1), S, G(2), and mitosis, but is absent from resting cells G(0),

which makes Ki-67 an excellent marker for determining the growth fraction of a given

cell population (104). Ki-67 immunohistochemical staining has been used as an index of

tumor growth in numerous of cancer studies, especially prostate and breast carcinomas

(26, 87). Treatment with ALA-rich flaxseed oil markedly lowered tumor burden in rats

accompanied by reduced Ki-67 level(78, 105).

Similarly to Ki-67, Proliferating Cell Nuclear Antigen (PCNA) is also considered

a potential prognostic marker in BC (106). PCNA is a ring-like nuclear protein which

Page 31: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

19

functions as the sliding clamp of DNA polymerases (107, 108). Thus, it is involved in

DNA replication and repair machinery of the cell (109). Expression of PCNA is a valid

cell proliferation marker since the distribution of PCNA was found to occur during G1, S

and G2 phase, but reaches low immunohistochemically detectable levels in M-phase of

the cell cycle (106, 108, 109). It has been shown that supplementation with n-3 PUFA

reduces the percentage of proliferating tumor cells by decreasing the expression of PCNA

(110).

The overall effect of high n-3 PUFA intake on cellular signaling process either

inhibits cell proliferation or promotes cell apoptosis (Figure 1.2). The changes in gene

expression, transcription factor activity and signaling transduction will be highlighted in

the following sections.

1.5 The Effect of n-3 PUFA Mixtures on BC Development

1.5.1 Animal Models

The inhibitory effects of n-3 PUFA on tumor growth have been well documented

in rodent models of BC. Xenograft, transgenic and chemically induced methods are the

three main approaches employed in rodent models.

1.5.1.1 Breast Cancer Studies in Xenograft Rodent Models

Xenograft rodent models involve the transplantation of human BC cells into

immunocompromised mice (78, 111). The type and concentration of dietary PUFA have

profound influences on the growth rate of transplantable human BC in rodents (Table

1.3). In one study, MDA-MB-435 BC cells xenografted into athymic nude mice in order

to compare intake of LA alone with diets containing LA and various proportions of

EPA/DHA (20). The diet rich in n-6 PUFA stimulated the growth and migration of

human BC cells in mice, whereas diets supplemented with EPA or DHA exerted

suppressive effects (20). Similarly, Karmail et al. also found an inhibition of R3230AC

Page 32: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

20

mammary tumor growth in rat fed with Maxepa, a menhaden oil supplement that contains

approximately 18% EPA and 12% DHA (112). These observations were largely

attributed to the high incorporation of n-3 PUFA into the tumor phospholipids, which

further reduced pro-inflammatory eicosanoid synthesis from AA (20). More recently, a

diet supplemented with 3% w/w fish oil concentrate was shown to stimulate lipid

peroxidation in the tumor cells, and thereby slowed down the tumor growth rate in

athymic nude mice implanted with MDA-MB-231 (30). Similar effects were observed in

MCF-7 human breast cancer xenografts, although a higher percentage of fish oil (19%

w/w) was required to decrease tumor volume in the nude mice (113). These findings

supported an earlier study showed that n-6/n-3 PUFA consumed in a low ratio resulted in

prolonged tumor latency and reduced tumor growth rate in BALB/cAnN mice (114).

Therefore, diet enriched with n-3 PUFA can alter murine mammary tumorigenesis.

1.5.1.2 Breast Cancer Studies in Transgenic Rodent Models

In transgenic mouse models, tumor growth can be initiated in two different ways:

gain of function involving oncogenes responsible for cell proliferation or loss of function

involving cell apoptotic pathways (115, 116). For example, the mouse mammary tumor

virus (MMTV) promoter allows the cancer-causing virus to be activated and expressed in

mammary tissue, leading to the development of mammary tumors (117). Several studies

have employed MMTV-neu related models and provided strong evidence for protective

effects of n-3 PUFA towards HER-2 positive BC (Table 1.4).

One study examined mammary tumor development in MMTV-Her-2/neu mice

fed menhaden fish oil from 7 weeks of age onwards. The tumor incidence was

dramatically reduced as well as prolonged tumor latency (115). Further, Yee et al.

demonstrated that dietary n-3 PUFA downregulated COX-2 and Ki-67 to inhibit cell

proliferation, further reducing atypical hyperplasia to prevent HER-2/neu mammary

carcinogenesis at early stages (77). Moreover, we showed that lifelong n-3 PUFA

exposure can mitigate tumor development in mice expressing MMTV-neu(ndl)-YD5, a

more aggressive HER-2- positive BC model (116). In this study, MMTV-neu(ndl)-YD5

Page 33: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

21

mice were crossed with fat-1 mice, yielding mice that were capable of endogenous

synthesis of n-3 from n-6 PUFA and had the susceptibility to mammary tumor growth

(116). Thus, this study provided a direct evidence for a protective effect of n-3 PUFA via

both complementary genetic and conventional dietary approaches (116). In order to

understand the dose-dependent effect of n-3 PUFA on mammary gland tumor

development, Leslie et al. further demonstrated a dose-response relationship of 0%, 3%

and 9% (w/w) menhaden oil on tumor volume reduction in MMTV-neu-YD5 mice (21).

In line with the previous study, the observed dose-dependent effects of n-3 PUFA were

associated with a dose-dependent change in the fatty acid profile, reflecting a decreased

n-6/n-3 ratio in the mammary glands and an increase of EPA and DHA in tumor

phospholipid classes (21). This is consistent with an earlier human clinical trial which

showed dietary DHA and EPA supplementation caused a dose-dependent increase of n-3

PUFA in serum and breast adipose tissues of BC patients (118).

Supplementing mice with 24% (w/w) menhaden oil delayed mammary tumor

development by 15 weeks relative to mice fed the same amount of corn oil (119). This

result heightened the potential value of n-3 as an effective agent against BC, while the

timing of exposure to n-3 PUFA will also influence future cancer risk (34). Mammary

gland research has identified critical periods of development, including early windows

such as in utero, lactation and pubescence. The mammary gland undergoes rapid growth

during these periods, and exposure to environmental agents may influence the long-term

health of mammary tissue. Thus, supplement with n-3 PUFA in early life stages may

protect against later BC development. In support of this, Su et al. conducted a study

exposing rats to maternal high n-6 PUFA diet with or without fish oil supplementation

during the perinatal period via maternal intake or during puberty or adulthood (29). They

found that fish oil intake during the perinatal period had a greater effect in preventing

mammary tumors than fish oil supplementation in later life (29). The decreased maternal

serum estradiol levels in pregnant rats with fish oil supplementation was thought to play a

role in reducing susceptibility to later BC development in the female offspring (29).

Page 34: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

22

Table 1.3 n-3 PUFA and breast cancer risk: xenograft rodent models.

Animal Model n-3 PUFA Source Feeding

Period Main Findings Mechanism Ref

Athymic nu/nu mice

MDA-MB 231

3% w/w fish oil concentrate

(10.2 g/kg EPA, 7.2 g/kg

DHA, 3.0 g/kg ALA)

7-week

(fed after tumor

established)

↓ tumor growth rate

↑ effectiveness of

doxorubucin

↑ EPA incorporation into tumor

↑ lipid peroxidation in tumor (30)

Athymic nu/nu mice

(NCr-nu/nu)

MDA-MB 435

40 or 80 g/kg EPA, DHA 13-week

(fed before

transplantation)

↓ tumor growth, size

↓ tumor weight

↑ EPA, DHA in tumor

phospholipids

↓ LA, AA in tumor

phospholipids

↓ AA-derived eicosanoids

(20)

Inbred F44 rats

R3230AC

5% marine oil

supplementation

(18% EPA, 12% DHA)

4-week

(fed before

transplantation)

↓ tumor weight, volume ↑ EPA, DHA, AA incorporation

into tumor

↓ Prostaglandins 2 series

(112)

BALB/cAnN mice

Mouse BC cell

10% or 20% w/w menhaden

fish oil

7-week

(fed before

transplantation)

↑ tumor latency

↓ tumor growth rate NA (114)

Athymic nude mice

MCF-7

19% w/w menhaden oil

(1.9 g/kg ALA, 19.4 g/kg

EPA, 24.3 g/kg DHA)

6 or 8-week

(fed after tumor

established)

↓ tumor volume ↑ lipid peroxidation in tumor (113)

↑: increase; ↓: decrease; NA: not available

Page 35: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

23

Table 1.4 n-3 PUFA and breast cancer risk: transgenic rodent models.

Animal Model n-3 PUFA Source Feeding Period Main Findings Mechanism Ref

MMTV-HER-

2/neu

22.50 kcal% menhaden oil

(15 g/kg EPA, 10.8 g/kg

DHA)

28-week

(fed before

tumor

development)

↓ atypical ductal

hyperplasia

↓ cell proliferation

prevented HER-2/neu at

early stages

↓ Ki-67 expression

↓ COX-2 expression (77)

MMTV-HER-

2/neu

22.50 kcal% menhaden oil

(15 g/kg EPA, 10.8 g/kg

DHA)

52-week

(fed before

tumor

development)

↓ tumor incidence and

multiplicity

↑ tumor latency

↓ mammary gland dysplasia

NA (115)

MMTV-neu

(ndl)-YD5 × fat1

3% w/w menhaden oil (0.5

g/kg ALA, 4.1 g/kg EPA,

3 g/kg DHA)

20-week

(lifelong

treatment, fed

before tumor

development)

↓ tumor volume and

multiplicity

↑ EPA, DHA and overall n-3 in

mammary tissues

↓ n-6/n-3 ratio in tumor

phospholipids

(116)

MMTV-neu

(ndl)-YD5

3% w/w menhaden oil (0.5

g/kg ALA, 4.1 g/kg EPA,

3 g/kg DHA)

9% w/w menhaden oil (1.3

g/kg ALA, 12.4 g/kg EPA,

9 g/kg DHA)

20-week

(lifelong

treatment, fed

before tumor

development)

↓ tumor volume and

multiplicity

↑ tumor latency

(all in a dose-dependent

manner)

↑ EPA, DPA in mammary tissues

↑ EPA, DHA in tumor

phospholipids

↓ LA, AA, n-6/n-3 PUFA ratio in

both mammary and tumor tissues

in a dose-dependent manner

(21)

↑: increase; ↓: decrease; NA: not available.

Page 36: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

24

1.5.1.3 Breast Cancer Studies in Chemically-Induced Rodent Models

Evidence from chemical-induced BC studies consistently supports the anti-cancer

effect of n-3 PUFA. Induced mammary carcinoma in rats by the injection of carcinogenic

chemicals such as 7, 12-dimethylbenz (α) anthracene (DMBA) and N-methyl-N-

nitrosourea (MNU) have been widely used in various BC chemopreventive studies. When

provided in the diet through menhaden or fish oil concentrates, the incorporated n-3

PUFA, EPA and DHA in particular, lower tumor incidence as well as retard tumor

growth and metastasis of chemically-induced and transplantable mammary tumors

(Table 1.5). Olivo et al. compared the effects of low- or high-, n-3 or n-6 PUFA exposure

on mammary tumorigenesis in rats (110). Feeding rats a low-fat n-3 diet significantly

lowered the incidence of DMBA-induced mammary tumors compared to n-6 fed rats.

This was accompanied by reduced cell proliferation and elevated lipid peroxidation.

However, the high-fat n-3 diet resulted in a significant increase risk of mammary

tumorigenesis relative to rats fed with low fat n-6 diet (110). Further, an in vivo study

demonstrated that low-fat n-3 PUFA diets inhibited cell proliferation via activation of

PPARγ and/or down-regulation of COX-2 and PCNA expression; whereas high fat n-3

PUFA diets stimulated cell proliferation and were positively associated with levels of

phosphorylated Akt (110). Similar suppressive effects of n-3 PUFA were observed in

another DMBA-induced rat model (27). Supplementation of EPA and DHA (from

Maxepa) effectively suppressed cell proliferation, which was accompanied by down-

regulation of Ki-67 and HER-2/neu expressions. Meanwhile, EPA and DHA induced cell

apoptosis through modulating the expression of Bcl-2 and Bax in mammary tissue of

Sprague-Sawley rats (26-28). These findings suggest that gene-nutrient interactions are of

a critical importance in the development of BC.

It has been shown that the different dietary fatty acid composition and n-6/n-3

PUFA ratios can diversely influence the occurrence and progression of BC (8, 32, 64).

Wei et al. fed MNU-induced rats with various n-6/n-3 PUFA ratios and demonstrated the

1:1 ratio of n-6/n-3 PUFA in diet was more effective in the prevention of mammary

tumor development when compared with diets higher in n-6 PUFA (64). Replacement of

n-6 by n-3 PUFA in diets, reflected an increase in EPA and DHA in mammary tumor

Page 37: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

25

tissue, which subsequently downregulated the expression of lipid metabolic-related genes

and inhibited cell proliferation (64). In agreement with this study, Jiang et al. reported

that low n-6/n-3 PUFA ratio (1:14.6) caused 80% reduction in tumor burden and 30%

decrease in tumor multiplicity in the same rodent model compared to a high n-6/n-3

PUFA ratio (1:0.7). These observations were mainly mediated by downregulating NF-κB,

pAkt and IGF-IR, and increased activity of PPARγ (87).

1.5.2. Cell Culture Studies

It has been well established that n-3 PUFA can suppress the development of

cancers by inhibiting cellular proliferation and inducing apoptosis. Cell culture studies

investigating the effects of n-3 PUFA on murine and human BC cells provide important

insights into the mechanisms underlying this inhibitory effect. Although there are few

studies describing the individual effects of ALA, EPA and DHA in vitro, the available

data consistently show that n-3 PUFA have direct growth inhibitory effects on several BC

cells lines (Table 1.6).

MDA-MB-231 human BC cell line was used to investigate the effect of various

classes of fatty acids (n-3, n-6 and n-9 PUFA)(120). EPA and DHA exhibited a dose-

dependent inhibition of cell growth, whereas LA and oleic acid (OA) stimulated cell

growth at very low concentration (121). Some in vitro studies have not included the

essential n-6 PUFA (i.e., LA) in the growth medium, which would be present in vivo and is

required for mammary tumorgenesis in animals. These studies are not able to explain the

tumor growth inhibition by n-3 PUFA in the presence of abundant LA. Schley et al.

examined the inhibitory effects of n-3 PUFA on BC in vitro in the presence or absence of

LA (22, 96). It was demonstrated that EPA and DHA induced apoptosis and increased

DNA fragmentation in MDA-MB-231 cells when provided in combination with LA.

Similar effects were observed in the MCF-7 BC cell line (72, 86, 105, 122). Barascu et

al. revealed that EPA and DHA decreased MCF-7 cell growth and increased the fraction

of apoptotic cells in a concentration-dependent manner, and particularly, a higher

efficiency noted for DHA (86). Specifically, they demonstrated that n-3 PUFA inhibited

cell proliferation by lengthening the cell cycle between the G2/M transition (86). In

Page 38: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

26

accordance with this finding, a previous study demonstrated that DHA induced marked

G2-M and G1-S arrest of the MCF-7 cells (123). Furthermore, treatment with EPA and

DHA was also shown to induce cell differentiation and increase lipid peroxidation

product levels in MCF-7 cells (105, 123, 124).

Chajes et al. examined the effect of ALA, EPA, and DHA on the proliferation of

human estrogen-positive (ER+) and estrogen-negative (ER−) BC cell lines (124). EPA

and DHA displayed a significant inhibitory effect on the proliferation of all types of

tumor cells, whereas ALA significantly inhibited cell growth in (ER−) MDA-MB-231

and HBL-100 human breast tumor cells but not in (ER+) MCF-7 cells (124). The

efficiency of this inhibitory effect was found to be correlated with the generation of lipid

peroxidation products (124). Consistently, another in vitro study identified a dose-

dependent inhibitory effect of ALA, EPA, and DHA on MCF-7 cells; however, the cells

were dramatically inhibited by EPA and DHA, and moderately inhibited by ALA (122).

Rapid tumor cell proliferation is a critical feature for tumor aggressiveness.

Treatment with LA and OA increased MDA-MB-231 cell proliferation through

production of pro-inflammatory prostaglandins and leukotrienes (121). EPA and DHA

could mimic the effect of indomethacin, an inhibitor of both COX and LOX, which in

turn attenuated BC cell proliferation by inhibiting n-6 PUFA related eicosanoid synthesis

(125). In addition, treatment with EPA and DHA inhibited EGFR phosphorylation and

diminished EFGR levels in lipid rafts (22, 72). These observations were due to the

incorporation of n-3 PUFA into BC cell membrane, which subsequently altered

membrane structure, signal transduction and function of BC cells (72). Moreover, n-3

PUFA can exert anti-proliferative effects by limiting cell cycle progression, attributed to

an inhibition of CDK1-cyclin B1 complex, a master regulator required for the initiation

of mitosis (86). Furthermore, increasing evidence demonstrated that n-3 PUFA can

inhibit cell proliferation and increase cell differentiation by activating PPARγ, although

this effect was only observed with DHA treatment (105).

Page 39: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

27

Table 1.5 n-3 PUFA and breast cancer risk: chemically-induced rodent models

↑: increase; ↓: decrease; NA: not available.

Carcinogen n-3 PUFA Source Feeding Period Main Findings Mechanism Ref

MNU Fish oil 2%–10% w/w

n-3 PUFA in diet

18-week (at the same

time as MNU

administration)

10% w/w n-3 PUFA (1:1n-6/n-3 ):

↓ body weight, no tumor occurrence

5% w/w n-3 PUFA:

↓ tumor incidence and multiplicity

↑ EPA, DHA in

mammary

↓ FAS, COX-2, 5-LOX

(64)

MNU

Fish oil concentrate

Low n-6/n-3 = 1:14.6

High n-6/n-3 = 1:0.7

2-week (at the same

time as MNU

administration)

Low vs. high ratio n-6/n-3 PUFA diet:

↓ tumor incidence (21%),

↓ tumor multiplicity (30%), tumor burden

(80%)

↑ apoptotic index (129%)

↓ Ki-67

↑ Bax, Bax/Bcl2, PPARγ

↓ NF-κB p65, pAkt, IGF-

IR

(87)

MNU

EPA/DHA alone: 95 g/kg

EPA/DHA EPA + DHA:

47.5 g/kg EPA + 47.5 g/kg

DHA

20-week (at the same

time as MNU

administration)

DHA alone vs EPA + DHA vs EPA alone:

↓ tumor incidence: 23%, 73%, 65%

↓ tumor multiplicity: 0.23, 1.67, 1.59

DHA is more effectively than EPA

NA (120)

Page 40: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

28

Table 1.5 n-3 PUFA and breast cancer risk: chemically-induced rodent models (continue)

↑: increase; ↓: decrease; NA: not available.

Carcinogen n-3 PUFA Source Feeding Period Main Findings Mechanism Ref

DMBA

Maxepa (fish oil

concentrate):

90 mg EPA + 60 mg

DHA per day

24-week or 35-week

study (before

DMBA injection)

↓ DNA single-strand breaks

↓ cell proliferation ↓ Ki-67, Her-2/neu (26)

DMBA Maxepa: 90 mg EPA +

60 mg DHA per day

24-week study

35-week study

(before DMBA

injection)

↓ tumor incidence (23%), tumor

multiplicity (42%) ↑ cell apoptosis ↓cell

proliferation ↓ Bcl-2 ↑Bax ↑ p53 (27, 28)

DMBA

Fish oil (0.5%ALA,

16% EPA, 1.2% DPA,

8% DHA in fish oil) NA

↓ tumor incidence with fish oil

consumption: adulthood < puberty <

perinatal

↓ tumor multiplicity with fish oil

consumption: adulthood > puberty >

perinatal

↓ maternal serum

estradiol (29)

DMBA

Menhaden oil Low-fat

n-3 PUFA diet: 4.6 g/kg

EPA + 3.2 g/kg DHA

High fat n-3 PUFA diet:

9.1 g/kg EPA + 6.3 g/kg

DHA

20-day (before

DMBA injection)

Low n-3 diet: ↓ tumor incidence ↓ TEBs ↓

cell proliferation ↑ cell apoptosis;

High n-3 diets exert opposite effects

Low n-3 diet: ↓ COX-

2, PCNA ↑ PPARγ

↑ lipid peroxidation

High n-3 diet: ↑ pAkt

↑ lipid peroxidation

(110)

Page 41: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

29

Table 1.6 n-3 PUFA and breast cancer risk: cell culture studies.

Cell Type n-3 PUFA Source Main Finding Mechanism Ref

MDA-MB-

231

EPA/DHA alone: 75 μM

100 μM EPA+DHA

combination: 45 μM EPA+30

μM DHA or 60 μM EPA+40 μM

DHA

(in presence/absence of LA)

↓ cell viability, cell proliferation

↑ DNA fragmentation, cell apoptosis

DHA was more potent than EPA

↓ pAkt

↓ NF-κB and DNA binding

activity

(96)

MDA-MB-

231

0.5–2.5 μg/mL of EPA, DHA

(1.7–8.2 μM EPA, 1.5–7.6

μMDHA)

↓ tumor cells growth

(DHA > EPA, dose-dependent)

↓ LA composition in cell lipids

↓ AA-derived eicosanoid

synthesis (121)

MDA-MB-

231

EPA/DHA alone: 75 μM

100 μM EPA+DHA

combination: 45 μM EPA+30

μM DHA or 60 μM EPA+40 μM

DHA

(in presence/absence of LA)

↓cell growth (48%–62%)

↑ EPA, DHA, DPA and total n-3 in

lipid rafts

↓ EGFR levels ↑ pEGFR

(22)

MDA-MB-

231 MCF-7 EPA (230 μM), DHA (200 μM) ↓ cell viability ↑ cell apoptosis

↓ Bcl-2 ↑pro-caspase-8

↓ pEGFR

↓ EGFR (only DHA)

↓ AA ↑ EPA, DPA, DHA in total

cell lipids

(72)

↑: increase; ↓: decrease; NA: not available

Page 42: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

30

Table 1.6 n-3 PUFA and breast cancer risk: cell culture studies (continue)

Cell Type n-3 PUFA Source Main Finding Mechanism Ref

MDA-MB-231

MCF-7 3–100 μM of EPA, DHA

At 50 μM EPA, 30 μM DHA

↑ cell apoptosis ↓ cell growth

At 50 μM EPA, DHA

↑ G2/M duration (DHA was more potent than

EPA)

↓ phosphorylation of cyclin B1

↓ activity of CDK1-cyclin B1 (86)

MCF-7 100 μM of EPA, DHA

↓ cell growth (30% by EPA, 54% by DHA)

↑ cell differentiation (30% by EPA, 65% by

DHA)

No significant effects on cell apoptosis and cell

cycle DHA was more potent than EPA

↑ PPARγ (DHA only) (126)

MCF-7

MCF-10A 6–30 μM of ALA, EPA, DHA

All n-3 PUFA ↓ MCF-7 cell growth (EPA,

DHA > ALA, dose-dependent)

AA ↓ MCF-7 cell growth (similar as ALA)

NA (122)

ER+ and

ER− cells

20 μg/mL of ALA, EPA, DHA

(72 μM ALA,

66 μM EPA, 61 μM DHA)

EPA, DHA

↓ cell proliferation (all cell lines) ALA

↓ estrogen independent BC cell proliferation

↑ lipid peroxidation (124)

↑: increase; ↓: decrease; NA: not available

Page 43: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

31

Dysregulation of apoptosis is also a hallmark of cancer cells, and thus agents that

activate apoptosis are highly desired. Treatment with EPA and DHA inhibited

phosphorylation of Akt as well as the NF-κB DNA binding activity (96). This represents

a novel mechanism by which n-3 PUFA induce apoptosis in MDA-MB-231 cells (96).

Additionally, it has been shown that n-3 PUFA decreased expression of Bcl-2 and

increased activity of pro-caspase-8, an apoptosis effector enzyme (72). As a result, n-3

PUFA can inhibit BC development in vitro by both suppressing tumor cell proliferation

and inducing tumor cell death.

1.6. The Effect of Individual n-3 PUFA on BC Development

1.6.1 ALA and BC

1.6.1.1 Inefficient Conversion from ALA to EPA and DHA

α-Linolenic acid (18:3n-3; ALA) is the major n-3 PUFA in the Western diet.

Typical consumption of ALA in Europe, Australia and North America ranges between

0.6 and 1.7 g per day in men and 0.5–1.4 g per day in women (58). This is about 10-fold

lower than the consumption of n-6 PUFA. ALA, regarded as the precursor for long-chain

PUFA, can be converted to EPA (20:5n-3), DPA (22:5n-3) and DHA (22:6n-3) by the

pathway shown in Figure 1.1. Whether the essentiality of ALA in the diet primarily

reflects the activity of ALA itself or of long-chain PUFA synthesized from ALA is a

matter of debate. The concentration of ALA in plasma phospholipids, cells and tissues is

found to less than 0.5% of total fatty acids (127). Although the dietary intake of EPA and

DHA are approximately 10-fold lower than those of ALA in North America, the

concentrations of these long-chain PUFA in plasma, cell and tissue phospholipids are

greater than those of ALA (127). This apparent mismatch between dietary intakes and

levels of incorporation further suggests that the primary biological role of ALA is for

EPA and DHA synthesis. However, it is also possible that the low concentration of ALA

may be due to negative selection in the incorporation of ALA into blood and cell

membrane lipid pools (58). Since consumption of EPA and DHA show a strong inverse

Page 44: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

32

association with the risk of BC, this raises the question of whether conversion of ALA to

EPA and DHA in humans is a viable alternative to dietary sources of these long-chain

PUFA. The majorities of human studies estimate that ALA supplementation in human

adults generally lead to an increase in EPA and DPA, but have little or no effect on DHA

content (128-132). Conservatively, Pawlosky et al. estimated the overall efficiency of

conversion from ALA was 0.2% to EPA, 0.13% to DPA and 0.05% to DHA (129). This

inefficient conversion is due to the first rate limiting reaction catalyzed by Δ6-desaturase

(58). As a result, the extent of ALA conversion to EPA and DHA is inefficient and

limited. Thus, ALA may not be considered as an effective alternative source to fish for

providing EPA and DHA.

1.6.1.2 Individual Effect of ALA on Breast Cancer

Data derived from epidemiological and observational studies suggest that ALA

present in the Western diet has protective effects in BC (Tables 1.7 and 1.8). Two case

control studies compared the fatty acid composition in the adipose breast tissue from

women with invasive non-metastatic breast carcinoma and women with benign breast

disease (44, 133). Low ALA content in adipose tissue was found to be associated with an

increased risk of BC. This observation was consistent with a previous cohort study on

121 BC patients, which demonstrated a link between a low level of ALA in adipose

breast tissue and increased risk of metastatic development (134). In particular, the ratio of

n-6/n-3 PUFA was also shown to be positively correlated with BC in these patients,

highlighting the role of n-3 and n-6 PUFA balance in BC.

Dietary supplementation of ALA reduced the growth of established mammary

tumors in chemically-induced rats and xenograft rodent models. In OVX athymic mice

with high circulating estrogen level, flaxseed oil and its high ALA content, attenuated

(ER+) MCF-7 breast tumor growth by reducing cell proliferation and increasing

apoptotic index (78). This effect was probably due to the downregulation of tyrosine

kinase receptors such as EGFR and HER2, with a subsequent reduction in pAkt.

Compared to corn oil, consumption of ALA-rich flaxseed oil tends to modify the n-6/n-3

Page 45: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

33

PUFA ratio by increasing serum ALA, EPA and DHA concentrations. This provides

evidence that ALA absorption and conversion may contribute to the observed tumor

reducing effect in the flaxseed oil diet (78). Further, an in vitro study showed that pure

ALA inhibited MCF-7 cell proliferation by 33%, which was in accordance with the in

vivo reduction in palpable tumor growth (33%) (78). This suggests that ALA itself, rather

than the generated EPA and DHA, exerts this anti-tumorigenic effect (78). Another in

vivo study was designed to elucidate which component(s) of flaxseed (lignan or ALA)

was responsible for enhancing tamoxifens effect on reducing the growth of established

MCF-7 breast tumors at low circulating estrogen levels (105). ALA-rich flaxseed oil had

a stronger effect in reducing the palpable tumor size of tamoxifen-treated tumors

compared with lignan-treated mice (105). More importantly, ALA was found to

downregulate HER2 expression, and subsequently modulates growth factor-mediated

signaling pathways by repressing IGF-1R and Bcl-2 (105).

Remarkably, long-term changes in dietary ALA and LA ratio significantly affect

mammary tumor outcomes. Feeding BALB/c mice with ALA-rich linseed oil inhibited

the development of mammary tumors compared to mice on corn oil diets (135). In a

recent study, use of canola oil (10% ALA) instead of corn oil (1% ALA) in the diet of

MDA-MB-231 implanted mice reduced tumor growth rate (136). Progression to

apoptosis is associated with a balance between pro-apoptotic Bax and anti-apoptotic Bcl-

2. Mice exposed to canola oil had an increased ratio of Bax/Bcl-2, which was responsible

for the apoptosis of defective epithelial cells (93). Furthermore, the maternal diets have a

life-long influence on development of BC in the daughter. Substitution of corn oil with

canola oil in the maternal diet increased n-3 PUFA incorporation into mammary glands,

which in turn delayed occurrence of mammary tumors and increased tumor cell apoptosis

in offspring (93). Elsewhere, maternal replacement of dietary soybean oil with canola oil

significantly lowered the burden of MNU-induced mammary tumors, along with increased

survival rate in the offspring (137). As a result, maternal ALA supplementation brings

about a stable epigenetic imprint of genes that are involved in the development and

differentiation of the mammary gland, which are passed on to female offspring where they

exert a protective effect against BC.

Page 46: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

34

Notably, the protective role of ALA in BC was inconclusive in in vitro studies. In

BT-474 and SkBr-3 cancer cells that naturally amplify the HER-2 oncogene, exogenous

supplementation with ALA significantly suppressed HER-2 mRNA expression, thereby

reducing the probability of activation that leads to tumor growth (138). Moreover, ALA

co-exposure was reported to synergistically enhance trastuzumab (an anti-cancer therapy)

efficacy in HER2-overexpression BC cells (139). However, in a separate study, ALA

exerted minimal tumor-reducing effects in the presence of trastuzumab (140). In addition,

some studies had difficulties in characterizing the role of ALA in estrogen dependent and

independent BC cell lines (122, 124). This suggests a variable effect of ALA on cell

proliferation depending on the cell line assessed.

Overall, these findings suggest that diets rich in ALA can inhibit mammary tumor

development in animals and in vitro, however, it cannot be ruled out that some of the

effects are due in part to conversion of ALA to EPA and DHA, albeit limited.

1.6.2 Individual Effect of EPA on Breast Cancer

Fish oil is a mixture of EPA and DHA, which have been previously shown to

have protective effects against BC. However, whether EPA and DHA differentially or

similarly affect BC has not yet been determined. In support of the independent effect of

EPA on BC, several in vitro studies consistently demonstrated the ability of EPA to

induce apoptosis in human BC cells (Tables 1.7 and 1.8). Chiu et al. demonstrated that 40

μM EPA induced cell apoptosis through inhibition of anti-apoptotic regulator proteins,

such as Bcl-2 (141). Moreover, Akt was also shown to be susceptible to EPA (97).

Treatment with EPA lowered both total and phosphorylated Akt content in transfected

MCF-7 cells that overexpressing constitutively active Akt (97). Additionally, co-

treatment with EPA enhanced the growth inhibitory response to tamoxifen in MCF-7

cells, thus suggesting that EPA may be useful as a nutritional adjuvant in the treatment of

BC (97).

Diets rich in EPA have also been shown to inhibit the growth of spontaneous or

transplanted mammary carcinomas in animal models and human clinical trials. EPA was

Page 47: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

35

observed to slow tumor growth and reduced metastasis in mice implanted with KPL-1

human BC cells (142). In a separate study, when compared with LA diet, intake of EPA

significantly inhibited tumor cell proliferation and development of lung metastasis in

mice with induced mammary tumorigenesis (20, 121, 125). These inhibitory effects were

attributed to high incorporation of EPA into tumor phospholipids, and subsequently,

disrupting inflammatory eicosanoid biosynthesis from AA (20). More recently, EPA was

found to regulate cell proliferation in MCF-7 xenografts via an inhibitory G protein-

coupled receptor-mediated signal transduction pathway (143). Furthermore, a human

clinical study demonstrated a strong positive correlation between plasma EPA

concentrations and PPARγ mRNA levels in adipose tissue of obese subjects (144). Since

activators of PPARγ are known to inhibit cell proliferation and tumorigensis, up-

regulation of PPARγ gene expression by EPA might be a potential mechanism of action.

These findings suggest that EPA can independently act to inhibit the development and

progression of human BC.

1.6.3 Individual Effect of DHA on Breast Cancer

Several independent reports have shown that DHA can inhibit mammary

carcinoma development and progression (Tables 1.7 and 1.8). However, the specific

mechanisms underlying these protective effects of DHA remain to be determined.

HER-2 signaling is central to many processes involved in cellular proliferation

and survival (6). Treatment with DHA alone in vitro was shown to be effective in

disrupting lipid rafts in HER-2 overexpressing cells, inhibiting HER-2 activity and its

downstream signaling molecules (Akt and FAS, etc.), and consequently led to cell death

(6). Menendez et al. demonstrated that exogenous supplementation with DHA was able

to downregulate HER-2/neu oncogene expression in SK-Br3 and BT-474 human BC cells

(5). This supports the therapeutic potential of DHA supplementation in the treatment of

HER-2 positive BC. On the other hand, recent evidence suggests that DHA itself is

capable of decreasing EGFR localization in the lipid rafts of the MDA-MB-231 BC cell

line (56). Moreover, DHA supplementation was reported to significantly enhance the

Page 48: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

36

efficacy of EGFR inhibitors, which provides strong evidence for the potential

development of combination therapies targeting EGFR (56). The pre-exposure with DHA

has been shown to synergistically enhance the cytotoxicity of antimitotic drugs including

Taxane and Taxol against highly metastatic BC cells (5). This was attributed to the

incorporation of DHA into cellular lipids, and subsequently altered membrane fluidity

and function, thereby increasing drug intake (5). In agreement with this, a recent human

clinical trial demonstrated that addition of DHA into chemotherapy increased survival in

metastatic BC patients (145).

A number of earlier studies have suggested that the anti-cancer property of DHA

is attributable to its ability to inhibit cell growth and induce apoptosis. Kang et al.

demonstrated that DHA induced apoptosis in MCF-7 cells through a combination of

pathways (146). Mechanistically, it was largely due to increased lipid peroxidation,

followed by accumulation of reactive oxygen species in cancer cells and higher oxidative

stress, ultimately resulting in cell apoptosis (146). It was also possible that the elevated

intracellular levels of DHA stimulated activation of apoptosis effector enzyme, such as

caspase-8 and caspase-3, and thus inducing apoptotic cell death (146, 147). Furthermore,

Chiu et al. found that pure DHA inhibited growth of MCF-7 cells (148). Although DHA

did not affect pro-apoptotic Bax protein, it induced the downregulation of anti-apoptotic

Bcl-2 gene expression time-dependently, and thus increasing the Bax/Bcl-2 ratio (148).

DHA has also shown to suppress KPL-1 cell growth in vitro, accompanied by

downregulation of Bcl-2 (149). Since the ratio of Bax/Bcl-2 is positively associated with

apoptotic activity, the regulation of Bax and Bcl-2 can be considered an important step in

the apoptotic actions of DHA.

Animal studies also support the anti-cancer role of DHA in BC. Dietary

supplementation of pure DHA was showed to suppress tumor development in both

chemically-induced carcinoma and xenograft rodent models (150, 151). Low level DHA

administration markedly reduced tumor growth rates and tumor weight compared with

rats fed LA (150). These observed suppressive effects of DHA resulted from diminished

tumor eicosanoid concentrations and decreased cell proliferation (150). DHA has also

been shown to decrease mammary tumor incidence coinciding with a 60% increase in

Page 49: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

37

BRCA1 protein, a major tumor suppressor (151). In complement, DHA supplementation

was shown to increase BRCA1 at the transcriptional level (152). The correlation between

the in vitro and in vivo observations adds weight to DHA’s potential mechanism and

supports a beneficial role for DHA against BC.

Altogether, ALA, EPA and DHA have various effects against mammary tumor

development in vivo. In terms of in vitro studies, EPA and DHA individually exert an

inhibitory effect on the proliferation of almost all types of tumor cells; whereas ALA only

has effects on ER− and HER-2 positive BC cells in vitro (Table 1.8). As little as 50 μM

of EPA or 30 μM of DHA can dramatically reduce tumor cell viability, while 72 μM of

ALA only cause moderate inhibition on ER− cell proliferation. However, it is not

appropriate to compare the effective dosage of individual n-3 PUFA from different type

of studies, since the amount of tumor cells and the duration of treatment are different.

Additional experimental studies are needed to compare the efficacy of each individual n-

3 PUFA under the same conditions.

1.7 Plant-Derived n-3 (ALA) vs. Marine-Based n-3 (EPA, DHA)

The potential health benefits of n-3 PUFA have been examined in various types of

studies. However, the relative potency of plant-based n-3 versus marine n-3 PUFA, as

well as EPA versus DHA in inhibiting tumor growth remains unclear. There are only two

studies that have compared the efficacy between EPA and DHA (120). The first study

compared the ability of dietary EPA or DHA to suppress MNU-induced mammary

carcinogenesis in a rat model (120). Although treatment with DHA or EPA alone was not

as effective in combination, DHA was more potent in delaying tumor onset and reducing

tumor multiplicity relative to EPA (120). In accordance, several BC cell line studies

indicated that DHA was more effective in inhibiting MDA-MB-231 and MCF-7 cell

proliferation and invasion than EPA at the same concentration (86, 96, 121, 125, 153)

In terms of the efficacy of plant-based n-3 PUFA, there has not yet been a human

clinical study directly comparing ALA versus EPA or DHA in BC. To the best of our

Page 50: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

38

knowledge, only two cell culture studies have examined the effect of different types of n-

3 PUFA. ALA was less effective compared with EPA or DHA, but ALA had moderate

inhibitory effects in some BC cell lines (122). This may be due to the conversion of ALA

to EPA and DHA that would lower the amount of ALA incubated with cancer cells. In

addition, the lower incorporation of ALA into the cellular lipid pool may also be a

confounding factor.

1.8 Conclusions

In summary, the present review has assessed the anticancer effects of n-3 PUFA

when consumed or treated individually, as well as in n-3 PUFA mixtures. ALA, EPA and

DHA can differentially inhibit mammary tumor development by changing the cell

membrane fatty acid composition, suppressing AA-derived eicosanoid biosynthesis and

influencing signaling transcriptional pathways to inhibit cell proliferation and induce

apoptosis. This review also provided evidence for using n-3 PUFA as a nutritional

intervention in the treatment of BC to enhance conventional therapeutics, or potentially

lowering effective doses.

Overall, in order to provide definitive recommendations, additional human studies

are required. Long-term studies tracking fish or n-3 PUFA intake are needed to

demonstrate a role for n-3 PUFA in prevention. Also, additional clinical trials are needed

to evaluate the effect n-3 PUFA on BC outcomes. Nevertheless, evidence does not

indicate harm and all forms of n-3 PUFA may be included in a

healthy diet.

Page 51: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

39

Table 1.7 Individual role of ALA, EPA and DHA on BC.

n-3 Amount of Fatty Acid Effect Mechanism Ref

ALA

NA Moderate decrease BC risk NA (45)

~22.8 g of ALA per kg diet Reduced tumor cell proliferation Inhibited HER2, EGFR expression (78)

~22.8 g of ALA per kg diet Inhibited MCF-7 cell proliferation (78)

~11 g ALA per kg diet Reduced tumor incidence and burden Increased BAX/Bcl-2 ratio (93)

10.6 g ALA per kg diet Decreased tumor growth rate Inhibited HER2 expression (105)

72 μM ALA Moderate inhibited ER-negative cell

proliferation, not affect MCF-7 NA (124)

30 μM of ALA Slightly inhibited MCF-7 NA (122)

NA Inversely associated with BC risk NA (133)

NA Inversely correlated with metastasis

development NA (134)

55.9 g ALA per kg diet Reduced tumor growth and metastasis NA (135)

8 g ALA per kg diet Decreased tumor growth rate NA (136)

10 g ALA per kg diet Reduced tumor burden, increased survival rate NA (137)

2.5-40 μM of ALA Enhanced cytotoxic effects of Trastuzumab

(at 10 μM of ALA) Down-regulated HER2 (at 20 μM of ALA) (138)

10 μM of ALA Diminished proteolytic cleavage of the

extracellular domain of HER2 Inhibited HER-2 activity (139)

~21.2 g of ALA per kg diet Minimal inhibited tumor growth w/wo

Trastuzumab NA (140)

52.8 g of ALA per kg diet Inhibited mammary tumor development NA (141)

NA: not available

Page 52: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

40

Table 1.7 Individual role of ALA, EPA and DHA on BC (continue).

n-3 Amount of Fatty Acid Effect Mechanism Ref

EPA

40–80 g of EPA per kg diet Slowed down tumor growth, reduced tumor burden Decreased AA derived-eicosanoid (20)

3–100 μM of EPA Induced BC cell apoptosis (at 50 μM of EPA) NA (86)

40–200 μM of EPA Restored the growth inhibitory effect of Tamoxifen

(at 40 μM of EPA) Decreased pAkt (at 20 μM of EPA) (97)

20–80 g of EPA per kg diet Inhibited the development of lung metastasis NA (125)

100 μM of EPA Inhibited MCF-7 cell growth NA (126)

40 μM of EPA Induced apoptosis, inhibited cell proliferation, arrested

cell cycle at G0/G1 Down-regulated Bcl-2 expression (142)

95 g of EPA per kg diet Reduced KPL-1 cell proliferation rate and metastasis NA (143)

42 g of EPA per kg diet Suppressed cell proliferation in MCF-7 xenografts in rats NA (144)

50 μM of EPA Increased PPARγ at mRNA level NA (145)

0–200 μM of EPA Inhibited MCF-7 cell growth (at 60 μM of EPA) NA (146)

NA: not available

Page 53: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

41

Table 1.7 Individual role of ALA, EPA and DHA on BC (continue).

n-3 Amount of Fatty

Acid Effect Mechanism Ref

DHA

120 μM of DHA Decreased cancer cell viability, enhanced the cytotoxic

activity of taxanes Decreased the expression of Her-2/neu (5)

100 μM of DHA Disrupted lipid rafts, induced apoptosis in HER-2

overexpressing cells Decreased Akt activity and FANS (6)

100 μM of DHA Decreased MDA-MB-231 cell proliferation, enhanced

EGFR inhibitors

Altered EGFR phosphorylation and

localization (56)

0–200 μM of DHA Reduced MCF-7 cell viability and DNA synthesis

(at 25 μM of DHA)

Increased lipid peroxidation, caspase 8

activation (146)

20 or 100 μM of

DHA

Inhibited MDA-MB-231 cell proliferation, promoted

nuclear condensation

Increased caspase-3 activity

(at 100 μM of DHA) (147)

10–160 μM of DHA Inhibited MCF-7 cell growth and induced apoptosis

(at 40 μM of DHA)

Downregulated Bcl-2, increased Bax/Bcl-2

ratio (148)

270 μM of DHA 50% inhibitory KPL-1 cell growth after 72 h treatment Downregulated Bcl-2, increased Bax/Bcl-2

ratio (149)

40 g of DHA per kg

diet

Decreased tumor growth rate and final tumor weight,

increased apoptosis Reduced tumor PGE2, decreased Ki-67 (150)

32 g of DHA per kg

diet Reduced tumor incidence Increased BRCA1 at protein level (151)

30 μM of DHA 50% inhibitory MCF7 cell growth after 96 h treatment Increased BRCA1/2 at transcriptional level (152)

NA Increased response of the tumor to chemotherapies,

increased survival rate (154)

NA: not available

Page 54: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

42

Table 1.8 Individual effect of ALA, EPA and DHA on different types of BC.

BC Cell Type ALA EPA DHA

MDA-MB-231 (ER−)

MDA-MB 435 (ER−) NA

MCF-10A (ER−) —

HBL-100 (ER−)

MCF-7 (ER+) —

ZR-75 (ER+) —

T-47-D (ER+) —

SK-Br3 and BT-474

(HER-2/neu positive) NA

Have significant inhibitory effect on cell proliferation; — slightly

inhibit the cell growth; NA: not available.

Page 55: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

43

Chapter Two

Mini review update

Role of Flaxseed Oil in the Prevention and Treatment of Breast

Cancer

Page 56: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

44

2.1 Introduction

Flaxseed (Linum usitatissimum), also known as linseed, is widely cultivated

throughout the northern hemisphere for both human and animal consumption(155).

The composition of flaxseed (FS) varies by growth location, year, environmental

conditions and cultivar. Typically, FS contains approximately 30% fiber, 20% protein,

40% fat, 4% lignan and 6% moisture (33, 156). In the past several decades, FS has

been the focus of interest in the field of food supplements due to its potential health

benefits in reducing the risk of cardiovascular disease, type II diabetes and some types

of cancer, including BC (157-159).

Two components of FS that may play a role in breast cancer (BC) are the

lignan and oil. The oil is rich in α-linolenic acid (ALA, 18:3n-3)(14, 159). Although

many foods contain lignans, FS is unique in that it is the densest source of the

mammalian lignan precursors, namely secoisolariciresinol diglycoside (SDG), with

between 60 and 300mg lignans per 100g servings(160). SDG is known as a type of

phytoestrogen, which is suggested to have estrogenic or anti-estrogenic properties,

and thus has been studied in hormone-related diseases including BC (156, 161-163).

Flaxseed oil (FSO) is low in saturated (9%), moderate in monounsaturated

(18%), and rich in polyunsaturated fatty acid (73%) (156). Notably, the predominant

polyunsaturated fatty acid (PUFA) in FSO is ALA (approximately 57%), followed by

linoleic acid (16%) (155, 156). According to our previous review paper, both ALA

and its long-chain n-3 PUFA metabolites, eicosapentaenoic acid (EPA, 20:5n-3) and

docosahexaenoic acid (DHA, 22:6n-3), can differentially inhibit mammary

carcinogenesis (164). However, the conversion of ALA to EPA and DHA is highly

inefficient in humans. Several human clinical trials demonstrated that dietary

supplement with FSO significantly increased the levels of ALA and EPA in the breast

milk, plasma, and erythrocyte, but failed to increase DHA content (128, 165). Thus,

the health benefits and the anticancer effects of FSO may be related to ALA

specifically, the metabolites EPA, and DHA, or the overall fatty acid profile of the oil.

Substantial evidence from rodent studies suggests that dietary FS and its oil

components can prevent BC development and inhibit mammary tumorigenesis (78,

105, 159). The anti-tumorigenic properties of FS have received considerable attention

Page 57: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

45

in Western countries. Many women and newly diagnosed BC patients change their

diet by increasing consumption of FSO and FS associated food products (166, 167).

Nevertheless, the epidemiologic studies examining the associations of FS and ALA-

rich FSO with the risk of development of cancer, however, have been inconclusive.

Previous published reviews mainly focused on the anticancer effects of fish oil,

whereas the evidence regarding a protective role for FSO in BC development remains

equivocal. Since FS is widely consumed in Western countries and ALA is the

principle source of n-3 PUFA in the Western diets, there is growing interest in

determining the role of FSO in the prevention and treatment of BC. Therefore, the

purpose of this mini review is to give an update on the protective role of ALA-rich

FSO as well as whole ground FS on BC development. Additionally, the potential

mechanisms of these anti-tumorigenic properties will be highlighted.

2.2 Role of flaxseed in mammary gland development and breast cancer risk

2.2.1 Animal Study

Rodent models have allowed for the study of the role of FS exposure at

various stages of the life cycle and carcinogenesis pathway. Time of exposure to FS is

an important factor in modulating cancer risk at multiple stages, including 1) early life

exposure, such as exposure in utero, during gestation and lactation; 2) adulthood

exposure; 3) exposure once mammary tumor is established.

2.2.1.1 Early life exposure

Development of the mammary gland (MG) is unique and continues throughout

life. Findings from experimental rodent models reveal that exposure to dietary factors

during the early life periods when the MG is undergoing extensive modeling and re-

modeling, alter susceptibility to develop mammary tumors during adulthood (168,

169). Two important structures that relate to MG development and risk of BC are

terminal end buds (TEB) and alveolar buds (AB). TEBs are the large bulbous

structures in a rodent MG that give rise to malignant mammary tumors upon exposure

Page 58: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

46

to a chemical carcinogen (169). This was attributed to the presence of a cap of highly

proliferative stem cells on each bud, and thus TEB is related with high cell

proliferation (170, 171). Similar structures in a human breast called terminal ductal

lobular unit 1 (TDLU1), which appear to be the sites of BC initiation in most women

(169). The incidence of carcinomas in rodents is directly associated with the density

of TEBs in the MGs at the time of carcinogen administration (170-172). In response

to stimulation by endogenous hormone or some dietary components, such as n-3

PUFA and soy protein, the highly proliferative TEB structures can be differentiated to

AB structures, which are less susceptible to carcinogens (110, 169). Thus, the dietary

factors that promote the early differentiation of TEB to AB may be a potential

preventive agent for BC during adulthood, and research has been conducted to

determine whether FS is such an agent.

Findings from rodent studies that relate early life exposure to FS with BC risk

are variable (Table 2.1). Exposure of female rats to 10% (w/w) flaxseed in utero,

during suckling, from suckling to postnatal (PND) day 50, and throughout lifetime

significantly delayed puberty onset, reduced number of estrous cycles and enhanced

the differentiation of highly proliferative TEB structures to less proliferative ABs

(173-175). This enhanced differentiation was partially mediated via the modulation of

the epidermal growth factor receptor (EGFR) signaling pathway, which potentially

protects against mammary carcinogenesis at early adulthood (175). However, no

beneficial effects were observed when exposure to FS occurred post-weaning, which

suggested that gestation and lactation are the critical periods for inducing structural

changes in the MG (173). Furthermore, exposure to 10% (w/w) flaxseed during

suckling was shown to significantly inhibit dimethylbenz(α)anthracene (DMBA)

induced rat mammary tumorigenesis (176). This observation emphasized that

exposure to FS during early stages of MG development can reduce the risk of

mammary tumorigenesis in rats later in life. Moreover, SDG at a level present in a 10%

(w/w) FS diet, has a similar effect as FS, indicating the anti-tumorigenic effect of FS

may be partly dependent on its lignans (176). However, other studies concluded that

exposure to 10% (w/w) FS or 15% (w/w) defatted FS in utero and during suckling

failed to enhance MG morphogenesis, and subsequently increased DMBA-induced

mammary tumorigenesis among female offspring (177, 178). This increase was seen

both in mammary tumor incidence and multiplicity (177, 178). The reason for this

Page 59: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

47

discrepancy may be due to the source of FS (159, 177). Studies showing cancer-

promoting effect of FS used FS from the US and Finland, whilst those showing a

protective effect used the FS from Canada. The different growth locations may have

led to differences in the level of cadmium in the seed; cadmium is a heavy metal that

activates the estrogen receptor (ER), and thus has estrogenic effects (159, 179).

2.2.1.2 Adulthood exposure once breast cancer is established

Many studies have investigated the late exposure effect of whole ground FS in

different rodent models of breast carcinogenesis (Table 2.2). Carcinogen-induced,

xenograft and transgenic mouse models are the three main approaches employed in

these animal studies.

A high fat diet has been shown to increase the proliferation of mammary

epithelial cells and mammary tumors in rodent models. Supplementation of a high-fat

diet with 5 or 10% (w/w) FS before DMBA administration reduced the risk for BC,

which was mediated by lowered the epithelial cell proliferation and nuclear aberration

in the rat MGs, particularly in the TEB structure (180). Nuclear aberrations are lethal

events that have been correlated with carcinogen exposure, and thus reductions in the

level of nuclear aberration indicate less carcinogen damage in MGs (180).

Interestingly, this inhibitory effect was more pronounced with FS flour (38% oil) than

with the defatted FS meal (2.8% oil), suggesting the oil components in FS may play a

role (180). Next, the same model was used to determine the effect of exposure of a 5%

(w/w) FS diet during initiation (before DMBA administration), promotion (after

DMBA administration), and throughout both of the two stages on mammary tumor

development after 25 weeks (181). Although there was no significant changes in

tumor incidence, the number of tumors was decreased in the groups fed 5% (w/w) FS

at the initiation stage and throughout the study (181). Moreover, the size of tumors

was significantly reduced in the group fed 5% (w/w) FS during the promotion stage.

These tumor-suppressive effects may partially be due to the high level of ALA in FS

oil, which altered the fatty acid composition of the rat MGs and tumor tissues (181).

Later on, Thompson et al. demonstrated that FS can reduce mammary tumor growth

at late stages of carcinogenesis (182). Feeding the mice with 2.5 and 5% (w/w) FS

Page 60: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

48

diet 13 weeks after DMBA administration significantly inhibited the growth of tumors

that established at the start of the dietary treatment, and also reduced the numbers of

newly developed tumors (182). In contrast to the DMBA-induced mammary

carcinogenesis, feeding FS at the 2.5 or 5% levels had no significant effect on MNU-

induced mammary tumor incidence, size or multiplicity when compared with the rat

fed a 20% soybean oil control diet (183). The lack of a significant effect of FS in

comparison to previous studies may be due to differences in the experimental design,

type and dose of carcinogen, and the protective effects of ALA present in the control

diet (1.6% ALA) (183).

The next series of experiments evaluated the effect of FS on established BC

using the xenograft rodent model. Dietary intake of 10% (w/w) FS inhibited the

growth of human ER-negative breast tumors (MDA-MB-435) and reduced metastasis

to distant organs, including the lymph node and lungs (184). This inhibitory effect

was modulated through downregulation protein expression of IGF-1 and EGFR.

Similar effects were also observed in the ER-positive MCF-7 human breast tumors (3).

In ovarietomized anthymic mice, 5% and 10% (w/w) FS exposure significantly

regressed established MCF-7 tumors in the presence of low or high estrogen level,

which mimics the hormone status of post- and premenopausal women, respectively

(3). This tumor suppression was concurrent with a reduction in cell proliferation (Ki-

67) and an induction of apoptosis, which was mediated by decreasing the expression

of estrogen sensitive gene products such as ERα and cyclin D1, and receptors of

tyrosine kinase growth factors such as human epidermal growth factor receptor-2

(HER2), EFGR and IGF-IR (185, 186). ER-α and ER-β are the two ER subtypes

identified in the mammary gland, and have distinct roles in mammary development.

ER-α is associated with increased cell proliferation and BC risk, while activation of

ER-β may prevent proliferative effects (186). Cyclin D1 is known as a major player in

cell cycle regulation, and has been shown to be regulated by estrogen acting through

the ER via the AP-1 DNA response element (187). As a result, the potential

mechanisms of the anti-tumorgenic effect of FS may be through inhibition of ER- and

growth factor-mediated signaling pathways.

Tamoxifen (TAM) and trastuzumab (TRAS, Herceptin™) are the most

commonly prescribed drugs for BC. It has been shown that dietary 10% (w/w) FS did

Page 61: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

49

not interfere with the effectiveness of TAM and TRAS, but strengthened the tumor

inhibitory effect of TAM under high or low estrogen concentration in both short- and

long-term treatment (105, 140, 188, 189). The 10% (w/w) FS used in these animal

studies is equivalent to human intake of about 25-50 g of FS per day, depending on

the person’s food consumption.

Furthermore, the Tg.NK transgenic mouse model that carries an activated

analogue, the c-neu oncogene, driven by the mouse mammary tumor virus (MMTV)

promoter was chosen as a relevant model for studying in vivo effects of FS on the

development of HER2-positive BC (190). Increasing levels of FS (60mg to 540mg/kg)

were incorporated into the diet and fed to female transgenic mice over a range of time

(190). Although post-weaning exposure to the FS diets had no effect on MG

differentiation, tumor incidence and number of large tumors (>6mm diameter) were

significantly lower in mice fed the highest FS diet (0.54% FS by weight) compared

with control after 23 weeks (190). However, none of the FS diets affected tumor

volume or multiplicity compared with control (190). This may be due to the dosage of

FS used in this study that was too low to have a significant inhibitory effect. Overall,

these findings support the role for dietary FS in the inhibition of mammary tumor

development in animal models.

2.2.2 Cell culture study

The protective role of FS has also been demonstrated in vitro. The cell growth

inhibitory effects of FS sprouts were examined over a wide range of doses and times

(191). FS sprout at 100 ug/ml resulted in a significant growth reduction of ER-

positive (MCF-7) and ER-negative (MDA-MB-231) cells in a time-dependent manner

(191). After 72 hours treatment, 100 ug/ml of FS sprouts significantly induced

apoptotic cell death, which was accompanied by an increase of p53 gene transcription

in both cell lines (191). p53 is known as a tumor suppressor gene involved in cell

apoptosis (192). Therefore, FS sprouts reduce BC cell growth by increasing p53-

mediated apoptosis.

Page 62: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

50

2.2.3 Human observational study

The initial case control study by the Ontario Women’s Diet and Health Study

was conducted to evaluate phytoestrogen intake and BC risk. Based on food

frequency questionnaire responses from 3370 women aged between 25-74 years, FS

associated products were accounted for 91% of the total lignan intake (193). Among

all the participants in this study, a daily intake of ≥ 5.35 mg lignans was associated

with a 19% reduction in BC incidence (odds ratio =0.81; 95% confidence interval

=0.65-0.99) (193). However, following stratification by BMI, this significant

association was observed only in overweight (BMI > 25) women. When stratified by

pre- or post-menopausal status, no statistically significant associations were observed

between lignan intake and BC risk (193). In 2013, Lowcock et al. reported a second

study, looking specifically at FS intake. Consumption of FS, and of flax bread was

associated with statistically significant 20-30% reductions in BC risk (194). This

relationship remained significant for postmenopausal women, but not among

premenopausal women (194).

The association between intake of sesame/flaxseed on mortality was

investigated in a large German cohort study of 2653 postmenopausal BC

patients(195). There was no significant association with increasing intake of

sesame/flaxseeds and BC-specific survival or overall survival (all PTrend>0.1).

Although the women in the study consuming lower level of sesame/flaxseeds

(0.3g/day) had a 25% reduction in BC-specific mortality, those with higher daily

intake (3.6g FS /day) had no survival advantage (195).

2.2.4 Human intervention study

To date, only one randomized double-blind placebo-controlled trial has

investigated the effect of FS on the biological markers of BC. In this study, muffins

containing 25g of FS were consumed by postmenopausal BC patients for 32 days

prior to surgery (50). Daily intake of FS significantly reduced tumor cell proliferation

and increased tumor cell apoptosis in postmenopausal patients, indicating by lowering

Ki-67 index and HER2 protein expression by 34% and 71%, respectively (50). In

addition to this intervention study, several other clinical trials have indirectly assessed

Page 63: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

51

the role of FS supplementation on BC risk by measuring changes in estrogen

metabolism (196-198). Since estrogen is involved in the development and progression

of BC, the ability to modulate estrogen metabolism and thereby affect tissue exposure

to biologically active estrogen may influence BC (196). 2-hydroxyestrone (2OHE1)

and 16α-hydroxyestrone (16αOHE1) are the two major metabolites of estrogen;

however, 2OHE1 has shown little biological activity with some anti-estrogenic

actions in vitro (199). Brooks et al. demonstrated that dietary supplementation with 25

g ground FS significantly alters the metabolism of estrogen in favor of the less

biologically active estrogen metabolite 2OHE1 in postmenopausal women (198).

Similar results were shown in a study with daily supplementation of 10g FS for 7

weeks (197). Furthermore, Hutchins et al. reported that daily consumption of 5 or 10

g ground FS for 7 weeks significantly reduced serum estrogen concentrations in

postmenopausal women (196). These findings consistently demonstrated the ability of

FS to modulate estrogen metabolism, which may be a potential mechanism for FS in

reducing the risk of BC.

Altogether, these data are supportive of findings from animal models and

confirm the protective role of FS in BC development.

2.3. Role of flaxseed oil in mammary gland development and breast cancer risk

2.3.1 Animal Study

2.3.1.1 Early life exposure

Few studies have specifically investigated the role of flaxseed oil (FSO) in the

development of MG and subsequent risk of BC. Gestation and lactation exposure to

1.82% (w/w) FSO (at levels present in 5% FS by weight) does not affect MG

differentiation as indicated by TEB and AB density (173). In a recent study, wild type

FVB/NHanHsd mice were exposed to a high fat diet containing 24% (w/w) corn oil or

FSO from preconception to 6 weeks of age (200). Exposure to FSO diet, but not corn

oil, resulted in differential expression of genes involved in energy metabolism,

immune response and inflammation (200). Since there were no detailed measurements

based on the histopathology of the MG or the fatty acid composition of the tissues, it

Page 64: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

52

is difficult to associate these gene changes in the MGs with BC risk. However, these

findings may at least suggest that intake of FSO during early life has a lasting effect

on the MG at the level of the gene expression (200).

2.3.1.2 Adulthood exposures once breast cancer is established

A considerable number of animal studies have shown that diet rich in ALA

can inhibit BC development (Table 2.3, some studies reviewed in Liu & Ma, 2014)

(164). Since FSO is known as the most abundant plant source of ALA, clarification of

FSO's involvement in BC development is essential. Cameron compared the effects of

diets rich in various oils (all at 6% by weight) on mammary tumor growth in DMBA-

treated C3H/Heston mice (201). ALA-rich FSO reduced tumor incidence compared

with n-6 PUFA rich corn and safflower oils. While in a separate study, feeding

Sprague-Dawley rats with 1.82% FSO, the level present in 5% FS, had no effect on

newly developed tumors, but significantly inhibited the growth of established tumors

(182). This inhibitory effect was correlated with an increased incorporation of total n-

3 PUFA, particularly ALA, and its elongation product EPA and DHA, resulting in a

significant rise in the ratio of n-3 to n-6 in mammary tumor tissue (182). Thus, FSO

may be more effective when tumors have already been established.

Studies using ovariectomized athymic mice with implanted tumors have been

valuable in determine the effect of FSO in reducing established tumors. One study

compared the effects of diets rich in various oils (from corn, fish and linseed) at 10%

level on the growth of implanted tumors derived from mouse mammary tumors (410

and 410.4) (135). Feeding BALB/c mice with ALA-rich linseed oil significantly

reduced the growth of 410.4 mammary tumors when compared to mice on corn oil

(135). Metastasis data paralleled the tumor growth rate (135). These observations

were negatively associated with the enhanced incorporation of n-3 PUFA into tumors;

where n-3 PUFA exert their physiological effects primary through changes in AA

metabolism, specifically decreasing pro-inflammatory eicosanoid synthesis, which

was indicated as reduced tumor prostaglandins E production (135).

Xenograft studies have also determined the effect of ALA-rich FSO diets on

the growth of human breast tumors. Dietary supplementation with 4% (w/w) FSO

Page 65: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

53

(levels equivalent in 10% FS diet) was shown to attenuate tumor growth and

metastasis in anthymic nude mice with established ER-negative human breast tumor

(MDA-MB-435) (202). Interestingly, when compared with mice fed with the same

amount of SDG present in 10% (w/w) FS diet, FSO was more potent in inhibiting

MDA-MB-435 tumor development. This in turn was related to a decrease in cell

proliferation and enhanced apoptosis, as indicated by lowered Ki67 and increased

apoptosis labeling index (202). Later on, Chen et al. used the same model to

determine the therapeutic effect of adjuvant FSO on the locoregional recurrence and

metastasis (203). 4% (w/w) FSO significantly reduced metastasis after the primary

tumor was surgically excised, and this was mediated by decreasing extracellular

vascular endothelial growth factor (VEGF) in breast tumor cells (204). However, FSO

had little effect on the tumor recurrence unless excision was made when the tumors

were small (< 0.9g) (203). Similar effects were also observed in ER-positive MCF-7

human breast tumors (refer to the review by Liu & Ma) (164). In support of this, a

recent study demonstrated dietary exposure to flaxseed cotyledon (FC), the primary

location of FSO in the seed (33.37g/kg diet), reduce the growth of MCF-7 breast

tumors at low circulating levels of estrogen (205). Notably, the regression of the

established tumor caused by the FC diet (56 %) was close to that caused by the 10%

(w/w) FS diet (62%) which was tested for 8 weeks under the same conditions,

suggesting that FC may potentially substitute for FS (206). These anti-tumorigenic

effects of FC was mainly mediated via downregulation of growth factor receptors,

such as EGFR, HER2 and IGF-1R, which further suppress the activation of protein

kinases MAPK and Akt downstream to the cascade of cyclin-dependent kinase (205).

BT-474, an ER-positive human BC cell line that exhibits amplified HER-2

was injected into athymic mice to investigate the interaction between FSO and TRAS,

since TRAS is a first line therapy for HER2-positive BC (159, 207). Although

feeding mice with 4 or 8% (w/w) of FSO alone did not cause significant effect on the

growth of HER2-overexpressing BT-474 tumors in athymic mice under high

concentrations of estrogen, it enhances the tumor-reducing effect of TRAS (159, 207).

Combining TRAS2.5 treatment (2.5 mg TRAS/kg body weight) with an FSO-rich diet

(4% by weight) for 4 weeks led to a greater reduction in tumor area compared to

treatment with this dose of TRAS alone (159). Tumor analysis showed that

FSO+TRAS2.5 treatment caused a greater reduction in cell proliferation and a greater

Page 66: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

54

increase in apoptosis compared to TRAS2.5 treatment alone further confirming the

reduction in tumor growth (159, 207). Importantly, combining low dose TRAS with

dietary FSO was just as effective as high dose TRAS, indicating that with FSO

consumption, only lower doses of the drug may be required to exert the same effect

(207). This may help delay the onset of TRAS resistance and relieve some of the side

effects of TRAS treatment, although more research is needed in this area. These

interactive effects are suggested to through the alteration in the membrane fatty acid

profile, which consequently led to a greater reduction of HER2 signaling as indicated

by lower levels of pHER2, pMAPK and pAkt expression (159). Additionally, FSO

was also shown to improve the responsiveness of TAM in reducing MCF-7 tumor

growth (206). Since TAM functions as a selective ER modulator, and is the most

commonly treatment for ER-positive BC, the interaction effect between FSO and

TAM was believe to mediate through ER signaling pathway, which may also block

the crosstalk between ER and growth factor receptors to inhibit the activation of

transcription (205).

The MMTV-c-neu transgenic mouse model has also been used to investigate

the role of FSO in the prevention of HER2-positive BC (208). The Tg.NK transgenic

mice were fed with 0.2 ml oil diets that contained increased proportion of FSO (0.05,

0.1, and 0.2 ml) mixed into corn oil for 30 weeks (208). Although the low dose FSO

wasn’t able to affect mammary tumor growth, there was a trend towards reduced

tumor incident and overall tumor weight with higher dose of FSO. This result

suggests that the ratio of n-6:n-3 plays an important role in mediating the effect of

FSO on HER2-overexpressing mammary tumourigenesis (208). The high level of

ALA in FSO may be beneficial in delaying the growth of mammary tumors if the n-6:

n-3 PUFA ratio is close to 1.

Taken together, these findings suggest that FSO is capable of reducing

tumorigenesis in animal models, modulating HER2 expression and growth factor

receptor signaling pathways. They also provide support for clinical trials to explore

the use of FSO as a dietary complementary aid for BC treatment with TRAS or TAM.

Page 67: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

55

2.3.2 Cell culture study

It has been shown that ALA-rich FSO can inhibit BC development in vitro by

both suppressing tumor cell proliferation and inducing apoptosis; however, this

protective effect may depend on the BC cell line being assessed (reviewed in Liu &

Ma, 2014) (164). BC can be divided into several molecular subtypes based on the

expression of cell ER, progesterone receptor (PR) and HER2 (209). Recently,

Wiggins et al. examine the effect of ALA on the growth of four BC cell lines with

varying ER, PR and HER2 status at both high and low circulation of estrogen (209).

As a result, ALA significantly reduced viable cell number (>50%) in all four cell lines

regardless of cancer subtype and estrogen status, at a dose as low as 50 µM treated for

96 h (209). Later on, those four cell lines were incubated with 75 µM ALA in

presence of estrogen to determine the potential mechanism of action of ALA (209).

Among the four cell lines, MCF-7 (ER+PR+low HER2) had the most changes in gene

expression after 24 h treatment, with the reduction in MAPK1 gene expression being

most notable. MAPK1codes a protein that is integral in the MAPK signaling cascade

leading to increased cell growth and crosstalk with other pathways such as PI3K/Akt

and estrogen signaling (98). This suggests the proposed mechanism of ALA is

through reducing the activation of MAPK cascade to decrease cell growth. Although

MDA-MB-468 (ER-PR-HER2-) has very little HER2 to begin with, there was a

significant reduction in HER2 expression after 24h incubation with ALA (209).

Interestingly, in BT474 (ER+PR+HER2+) cell line that overexpress HER2, no

significant changes in the expression of HER2 was observed; instead of it, there was

an unexpected decrease in BRCA1 (breast cancer 1, early onset), a tumor suppressor

gene (209). In the aggressive triple negative cell line MDA-MB-231 (ER-PR-low

HER2), there was a time-dependent reduction in the number of viable cell, and some

genes expressions that altered by ALA treatment were also dependent on time (209).

Overall, these results suggest the intake of ALA-rich FSO can reduce growth

of different BC cells and change the expression of a variety of genes; however, this

anticancer effect may depend on the molecular subtype of the BC cell lines.

Page 68: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

56

2.3.3 Human observational study

A large body of literature composed of human observational studies

examining the protective role of ALA in BC was inconclusive (has been reviewed in

Liu & Ma, 2014) (164). Some limitations are identified in these observational studied

which may impact the findings; they include 1) different measurements were used to

determine ALA exposure; 2) differences in sources of ALA; 3) the baseline

characteristics of the participants varied in different studies, such as age and

menopausal status. More importantly, no clinical studies have been conducted to

specifically examine the association between FSO consumption and BC risk.

Therefore, the protective role of FSO in decreasing BC risk still needs to be tested

clinically. To our knowledge, no human intervention trials have been completed to

investigate the effect of ALA-rich FSO on BC prevention or treatment.

2.4 Conclusion and future directions

In summary, the present review has assessed the anticancer effects of both FS

and its oil components. The current evidence from human observational and dietary

intervention studies suggests that FS and FSO are safe for consumption by healthy

individuals to potentially prevent BC and by BC patients to potentially reduce tumor

growth. ALA-rich FSO may be more effective in established mammary tumors. The

potential mechanisms by which FSO exert anti-tumorigenic actions are mainly

through inhibition of ER- and growth factor receptor-mediated signaling pathways.

This review also provided evidence for the potential use of dietary FSO as a

complementary agent in the treatment of BC to enhance conventional therapeutics, or

potentially lowering effective doses.

Further research is still needed in a clinical setting before definitive

recommendations can be made regarding the use of FS or FSO as an alternative,

adjuvant therapy for BC. For instance, long term studies tracking intake of FS and its

oil components intake are needed to demonstrate the relationship between FS

exposure and BC outcome. Also, additional large human intervention trails are

required to investigate the beneficial interactions of ALA-rich FSO with current BC

therapies.

Page 69: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

57

Table 2.1 Early exposure to flaxseed and breast cancer risk

Animal Dosage Feeding Period Main Findings Mechanism Ref

Sprague-

Dawley

female rats

5 or 10%

(w/w)

flaxseed

1) Gestation and

lactation; 2) after

weaning (PND 21-

50); 3) lifetime

(gestation to PND 50)

Gestation, lactation and lifetime exposure

Both 5 and 10% flaxseed ↓TEB density ↑AB density

5% flaxseed ↓puberty onset ↓estrous cycle

10% flaxseed ↑puberty onset ↓estrous cycle length

5% flaxseed anti-estrogenic effects

↑mammary gland atrophy

10%flaxseed estrogenic effects

↑mammary gland differentiation

(173)

Sprague-

Dawley

female rats

10%

(w/w)

flaxseed

1) Lactation only;

2) From lactation to

PND 50

During lactation only or continuously

10% flaxseed ↓TEB density ↑AB density

10% flaxseed

↑differentiation of TEB to AB (174)

Sprague-

Dawley

female rats

(DMBA)

10%

(w/w)

flaxseed

During suckling

(from birth to PND 21

day)

At PND 21

10% flaxseed ↑number of TEBs

↑cell proliferation of terminal ductal epithelium

At PND 49-51

10% flaxseed ↓number of TEBs

At PND 21

10% flaxseed ↓epithelial ER-β ↑EFGR

At PND 49-51

10% flaxseed ↓ EGFR and EGF

(175)

Sprague-

Dawley

female rats

(DMBA)

10%

(w/w)

flaxseed

During suckling

(from birth to PND 21

day)

10% flaxseed ↓tumor incidence

↓average tumor size and tumor number

↓total tumor load and final tumor weights

N/A (176)

Sprague-

Dawley

female rats

(DMBA)

5 or 10%

(w/w)

flaxseed

1) In utero; 2) during

lactation (PND 5-25)

5% flaxseed ↓ mammary tumor latency

10% flaxseed ↓ mammary tumor latency

↑ tumor multiplicity

Flaxseed exposure didn’t affect number of TEBs or the

cell proliferation within the epithelial structures.

10% flaxseed ↑lobular ER-α

↓ER-β in lobules and ducts (177)

Sprague-

Dawley

female rats

(DMBA)

15%

(w/w)

defatted

flax flour

In utero

15% defatted flax

No significant changes in mammary gland morphology

↑apoptotic cells in mammary gland at PND week 8

↑tumor incidence ↑tumor multiplicity

15% defatted flax ↓mRNA of BRCA1

(at PND 8-week) ↑mRNA of p53 (178)

Page 70: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

58

Table 2.2 Late life exposure to flaxseed and breast cancer risk (Carcinogen -induced rodent models)

Animal Dosage Feeding Period Main Findings Mechanism Ref

Sprague-

Dawley

female rats

5 and 10%

(w/w) flaxseed

flour

5 and 10%

(w/w) defatted

flaxseed meal

Start at 5-week-old, feed

for 4 weeks, before

DMBA administration

Flaxseed flour is more potent than defatted

flaxseed meal

↓epithelial cell proliferation

↓nuclear aberration in TEB

5% flaxseed flour exerted optimum effects

N/A (180)

Sprague-

Dawley

female rats

5% (w/w)

flaxseed flour

1) Initiation: post-

weaning to DMBA

administration

2) Promotion: after

DMBA administration

3) Initiation and

Promotion: after weaning

until end of 25 week

No significant difference in tumor incidence

5%FS during initiation or throughout the study

↓tumor multiplicity

5% FS during promotion ↓tumor size

↑ALA

↓DPA (n-6 PUFA)

No significant changes in

EPA and DHA content in

tumor or mammary

tissues

(180,

181)

Sprague-

Dawley

female rats

2.5 and 5%

flaxseed

13 weeks after DMBA

administration

Treat for 7 weeks

2.5% FS ↓new tumor incidence

2.5 and 5% FS ↓established tumor volume >50%

↓new tumor number and volume

2.5 and 5% FS ↓volume of established + new

tumor

N/A (182)

Sprague-

Dawley

female rats

2.5 and 5%

flaxseed

2 days after MNU

injection

Treat for 22 weeks

No significantly difference on tumor incidence,

volume, multiplicity and weight

↓tumor invasiveness and grade

↓plasma IGF-I in rats +/-

MNU (183)

Page 71: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

59

Table 2.2 Late life exposure to flaxseed and breast cancer risk (Xenograft rodent models, continue)

Animal Dosage Feeding Period Main Findings Mechanism Ref

Balb/c nu/nu mice

MCF-7

+/- estrogen

10% (w/w)

flaxseed +/- 5mg

tamoxifen

after tumor was

established

Treat for 6 weeks

Under both low and high estrogen level

↑tumor regression ↓tumor size

↑effectiveness tumor inhibitory effect of

TAM

Under both low and high estrogen

↓Ki-67 index in tumor

↑ tumor cell apoptosis

(3)

Ncr nu/nu mice

MDA-MB-435

10% (w/w)

flaxseed

after tumor was

established

Treat for 7 weeks

↓tumor growth rate, final tumor volume

and weight

↓lymph node metastasis incidence

↓number of lung metastatic tumor

↓ Ki67, IGF-I, EGFR in primary tumors (184)

Balb/c nu/nu mice

MCF-7

Low estrogen

10% (w/w)

flaxseed

after tumor was

established

Treat for 25 weeks

↓ tumor growth stimulating effect of soy

protein

↓ tumor volume and weight

↓ Ki67 cell proliferation index

↑ number of apoptotic tumor cells

↓pMAPK, IGF-1 and EGFR

↓ERα and cyclin D1

↑ERβ and HER2

(185,

186)

Balb/c nu/nu mice

MCF-7

Low estrogen

5 and 10% (w/w)

flaxseed +/- 5mg

tamoxifen

after tumor was

established

Treat for 16 weeks

5 and 10% FS ↑tumor regression

↓tumor volume and weight

10% FS ↑effectiveness of TAM in

tumor regression

5 or 10% FS combined with TAM

↓Ki-67, cyclin D1, ERα, HER2, IGF-1R (189)

Balb/c nu/nu mice

MCF-7

High estrogen

5 and 10% (w/w)

flaxseed +/- 5mg

tamoxifen

after tumor was

established

Treat for 8 weeks

5 and 10% FS alone ↓tumor growth (dose-

dependent)

FS combined with TAM ↓tumor growth

(5% FS is most effective)

FS alone ↓Ki-67, IGF-I, HER2, PgR

↑apoptotic cell

FS/TAM ↑apoptotic cell, ERα

↓Ki-67, IGF-I, PgR

(188)

Balb/c nu/nu mice

MCF-7

Low estrogen

10% (w/w)

flaxseed

after tumor was

established

Treat for 8 weeks

↓tumor size ↑tumor regression rate

↓Ki-67 index ↑apoptotic cells

↓Bcl-2, cyclin D1, ERα, ERβ, EGFR,

HER2, IGF-1R

(210)

Page 72: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

60

Table 2.3 Late life exposure to flaxseed oil and breast cancer risk

Animal Dose of FSO Feeding Period Main Findings Mechanism Ref

Carcinogen -induced rodent models

Sprague-

Dawley rats

DMBA

1.82% FSO:

18.2g/kg diet

(11.4g ALA/kg

diet)

7-week

(13 weeks after

DMBA

administration)

↓ established tumor volume (>50%)

no inhibitory effect on newly formed

tumor

↑ALA, EPA, DHA and total n-3

PUFA in tumors

↑ n-3: n-6PUFA

(182)

C3H/Heston

mice

DMBA

6% FSO: 60g/kg

diet

(28.2g ALA/kg

diet)

48-week

(1 week before

DMBA

administration)

↓ reduce tumor incidence (201)

Transgenic rodent models

TG.NK mice

with

MMTV/c-neu

0.05-0.2ml FSO

(59.4% ALA) 30-week

0.2ml FSO (when n3:n6 PUFA →1)

↓tumor incidence, overall tumor weight (208)

Page 73: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

61

Table 2.3 Late life exposure to flaxseed oil and breast cancer risk (Xenograft rodent models, continue)

Animal Dose of FSO Feeding Period Main Findings Mechanism Ref

Ovariectomized

anthymic mice with

high E2

MCF-7

4% FSO: 40g/kg diet

(22.8g ALA/kg diet)

8-week

(fed after tumor

established)

↓ tumor size and cell

proliferation

↑ tumor cell apoptosis

↑ serum ALA, EPA and DHA

↓ EGFR, HER-2 and Akt

protein

(78)

Ovariectomized

anthymic mice with

low E2

MCF-7

4% FSO: 38.5g/kg

diet + TAM

(21.2g ALA/kg diet)

8-week

(fed after tumor

established)

↓tumor size, cell proliferation

↑tumor cell apoptosis

↑effectiveness of TAM

↓HER-2, IGF-1R, Bcl-2 and

pMAPK

(105)

BALB/c mice

410 and 410.4 mouse

tumor cell

10% FSO: 100g/kg

diet

(55.9g ALA/kg diet)

3-8week

(fed before tumor

established)

no significant effect on 410

tumor growth

↓410.4 tumor growth and

metastasis

↑ALA, EPA and DHA in

tumors

↓tumor prostaglandin

production

(135)

Athymic mice

BT-474

(overexpressing HER-

2)

High estrogen

4% FSO: 40g/kg diet

+/- 2.5 mg/kg bw

TRAS

(22.8g ALA/kg diet)

4-week

(fed after tumor

established)

4% FSO alone has no effect on

tumor growth

FSO enhances the effectiveness

of TRAS

FSO+TRAS ↓tumor cell

proliferation

↑ tumor cell apoptosis

4% FSO ↑ALA, EPA and

DHA

↓n-6:n-3 PUFA ratio

FSO+TRAS ↓Ki-67

↓HER-2, pHER-2, pAkt,

MAPK protein expression

↓ mRNA of EFGR

(159)

Page 74: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

62

Table 2.3 Late life exposure to flaxseed oil and breast cancer risk (Xenograft rodent models, continue)

Animal Dose of FSO Feeding Period Main Findings Mechanism Ref

Athymic nude mice

MDA-MB-435

4% FSO: 36.53g/kg diet

(20.82g ALA/kg diet)

7-week

(fed after tumor

established)

↓tumor growth rate

↓distant lymph node metastasis (52%)

↓ Ki-67

↑ apoptosis index (202)

Athymic nude mice

MDA-MB-435

4% FSO: 36.53g/kg diet

(20.82g ALA/kg diet)

7-week

(fed after tumor

established)

↓lung and total metastases

No significant effect in tumor

recurrence

(203)

BALB/c athymic

nude mice with

MCF-7 with low

estrogen

8.2% FS cotyledon +/-

5mg TAM

= 33.37 FSO g/kg diet

(19g ALA/kg diet)

8-week

(fed after tumor

established)

↓ tumor size, tumor growth, cell

proliferation

↑ the effectiveness of TAM

↓Ki67

↓HER-2, pHER-2,

ERα, pMAPK, pAkt

↓mRNA of IGF-1R

(205)

Athymic mice

BT-474

(overexpressing

HER-2)

High estrogen

8% FSO: 80g/kg diet +

2.5 or 5mg/kg bw TRAS

(45.6g ALA/kg diet)

6-week

(fed after tumor

established)

↓ tumor size, tumor cell proliferation

↑ tumor cell apoptosis

↑ the effectiveness of TRAS

↓ Ki-67

(207)

Page 75: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

63

Chapter Three

Rationale, Hypothesis, Objective and Experimental Design

Page 76: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

64

3.1 Rationale

Breast cancer (BC) remains a major health concern as it is the leading cancer

killer of women worldwide (1, 2, 211). Growing evidence suggests that fish consumption

or intake of marine-derived long-chain n-3 polyunsaturated fatty acids (PUFA), such as

eicosapentaenoic acid (EPA, 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3), may

influence mammary gland (MG) development at early life stages and potentially reduce

the risk of BC (8, 11, 34, 164). However, substantive evidence for a biological role for

the precursor, alpha-linolenic acid (ALA, 18:3n-3), remains equivocal. ALA is a plant-

based n-3 PUFA, but the conversion of ALA to EPA and DHA is highly inefficient in

humans. Meanwhile, ALA is known as the principle source of n-3 PUFA in the Western

countries, where the intake of EPA and DHA are typically low (32, 44). Therefore, the

clarification of ALA’s involvement in BC is essential. To the best of our knowledge, no

study has directly compared the effects of plant- and marine-based n-3 PUFA on

mammary tumor development. This fundamental knowledge is lacking, and is highly

important for the development of dietary strategies to prevent BC in the Western

countries. Thus, the research objective was to 1) determine the relative inhibitory effects

of lifelong exposure to plant-derived versus marine-based n-3 PUFA on mammary tumor

development; and to 2) identify potential mechanisms by which they exert anticancer

effects.

3.2 Mouse Model: MMTV-neu (ndl) YD5

The creation of transgenic mouse models has provided a direct method of

observing tumor growth and greatly enhanced our understanding of molecular

mechanisms underlying BC development and progression. For instance, the mouse

mammary tumor virus (MMTV) mouse models have been informative models for human

BC despite morphological, hormonal and lifestyle differences between mice and humans.

MMTV is an oncoRNA virus of the Retroviridae family, which causes breast tumors

once activated (212). Efficient replication of MMTV occurs predominantly in the

alveolar epithelial cells of the MG, and the expression of MMTV is under the influence

Page 77: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

65

of steroid hormones (212). The MMTV induce premalignant lesions and malignant

tumors of the MG by acting as an insertional mutagen or by activating transcription of

nearby oncogenes (117, 213). ErbB2 (Neu/HER-2) is an epidermal growth factor receptor;

the term neu is used when referring to the murine gene or cDNA, whereas the human

homolog are designated as HER-2 (214). Overexpression of HER-2 is responsible for 25-

30% invasive human BC cases, which correlates with poor patient prognosis (73, 215,

216). In this study, transgenic mice used were engineered to overexpress the neu

protooncogene under the transcriptional control of MMTV enhancer. Oncogenic

activation of neu occurs as the result of a single point mutation in the transmembrane

domain, converting a valine residue to glutamic acid (217). Although comparable levels

of neu protein were detected in both normal and tumor tissues, catalytically active neu

was detected solely in tumors (214). Additionally, MMTV-neu breast tumors exhibit

sporadic mutations in neu resulting in its constitutive activation. Mutations commonly

encountered include an in-frame deletion within the extracellular domain of the neu

protein (212, 214). The neu deletion mutants (ndl) are able to promote the transforming

activity of neu transgene and lead to the development of mammary tumors faster than the

observed in the original MMTV-neu mice (212, 214). Furthermore, biochemical

characterization of these mammary tumors revealed the presence of neu receptor dimers

that were constitutively tyrosine phosphorylated (117). YD (tyrosine residue 1227) is one

of the neu autophosphorylation sites that can independently mediate the transforming

signals. Female MMTV transgenic mice expressing activated neu alleles of YD5 were

capable of inducing mammary tumors at an average age of 102 days (117).

Altogether, the MMTV-neu (ndl) YD5 is a highly aggressive BC model, which

can cause 50% of the MMTV transgenic mice to develop mammary tumors within 102

days of age (21, 116). Since the neu-oncogene corresponds to HER2 gene in humans, this

mouse model will be useful in detailed research and analysis of the signaling cascades

involved in HER-2 positive BC.

Page 78: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

66

3.3 Hypothesis

The overall hypothesis is that lifelong exposure to both marine- and plant-derived

n-3 PUFA can prevent mammary tumor development. Specifically, it is hypothesized that:

1) Lifelong exposure to n-3 PUFA can influence MG development early in life by

reducing the number of terminal end buds (TEB) in MMTV-neu (ndl)-YD5 mice

2) Lifelong exposure to n-3 PUFA have long-term preventive impact on

mammary tumor development by delaying tumor latency, reducing tumor volume and

multiplicity in MMTV-neu (ndl)-YD5 mice

3) Plant-derived n-3 PUFA can inhibit mammary tumor development in a dose-

dependent manner

3.4 Objective

To determine the effects of lifelong n-3 PUFA exposure on:

1) The number and density of TEBs in mammary glands of mice at 6 weeks

2) Mammary tumor development including tumor latency, tumor volume and tumor

multiplicity of mice over 20 weeks

3) Fatty acid composition of tumor tissues and adjacent MGs, as well as serum

4) Expression of proteins that involved in tumor cell proliferation and apoptosis

3.5 Experimental Design

Heterozygous MMTV-neu (ndl)-YD5 males were bred with wild-type female

FVB mice. From this cross, two genetically distinct offspring were obtained: wild-type

mice (WT) and heterozygous MMTV-neu (ndl)-YD5 (MMTV). The harem mice were

Page 79: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

67

fed one of the four diets, containing 1) 10% safflower oil, 2) 3% flaxseed oil plus 7%

safflower oil, 3) 10% flaxseed oil alone, or 4) 3% menhaden oil plus 7% safflower oil. 10%

safflower oil is an n-6 PUFA rich diet, which served as a positive control; while 3%

menhaden oil is rich in marine-derived n-3 PUFA and used as a negative control. The

amount of EPA and DHA present in 3% menhaden oil diet is equivalent to the level

present in traditional Japanese diets (1-2% of daily energy as marine-derived n-3 PUFA).

3% and 10% flaxseed oil contain different levels of ALA that are designed to test the

dose-response relationship between plant-derived n-3 PUFA exposure and BC outcome.

Female transgenic MMTV offspring were maintained on the same diets as their parents

for 6 or 20 weeks. At 6 weeks, mice were randomly chosen for euthanization, the number

and density of TEBs in the mouse mammary gland were measured. Starting at week 12,

the MGs of mice were palpated for new tumors; the tumor volume and multiplicity were

tracked for the duration of the 20-week-study. Mice were euthanized at 20 weeks to

collect tissues and organs. To determine the effect of diet on tissue fatty acid composition,

fatty acid analysis was conducted on tumor tissue, MGs and serum samples from 6- and

20-week-mice. Finally, the expressions of relevant protein targets in mouse tumors were

assessed to determine the impact of n-3 PUFA on cell proliferation and apoptosis.

Page 80: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

68

Chapter Four

Plant- and Marine-derived N-3 Polyunsaturated Fatty Acids Prevent

Mammary Tumor Development

Page 81: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

69

4.1 Abstract

Marine-derived n-3 polyunsaturated fatty acids (PUFA), such as eicosapentaenoic

acid (EPA) and docosahexaenoic acid (DHA), have been shown to inhibit mammary

carcinogenesis. However, evidence regarding α-linolenic acid (ALA), a plant-based and

major n-3 PUFA in the Western diet, remains equivocal. Therefore, this study examined

the relative inhibitory potency of plant versus marine based n-3 PUFA on mammary

tumor development in a relevant model of human breast cancer over expressing human

epidermal growth factor receptor 2 (HER-2/neu). Heterozygous MMTV-FVB males were

bred with FVB female mice and fed one of four diets: 1) 10% safflower oil, 2) 3%

flaxseed oil plus 7% safflower oil, 3) 10% flaxseed oil alone, or 4) 3% menhaden oil plus

7% safflower oil. Transgenic female offspring were maintained on the parental diet for 6

or 20 weeks. Compared to the 10% safflower oil, intake of 10% flaxseed and 3%

menhaden oil significantly decreased the number and density of terminal end buds in the

mammary glands of mice at 6 weeks of age (p <0.05). At 20 weeks, a significant (p<0.05)

dose-dependent reduction of tumor volume (24% and 78%) and multiplicity (15% and

43%) was observed in mice fed 3% or 10% flaxseed oil, respectively. Relative to 3%

menhaden oil, 10% flaxseed oil resulted in a similar reduction in tumor volume (78% and

72%) but led to a greater reduction in multiplicity (27% vs. 43%). However, 3% flaxseed

oil had weaker inhibitory potency compared to 3% menhaden oil (p<0.05). Based on the

fatty acid composition of these n-3 PUFA diets, plant-derived ALA was only 1/8 as

potent as marine-based EPA and DHA in inhibiting mammary development. 3%

menhaden and 10% flaxseed oil down-regulated tumor expression of proteins involved in

cell proliferation (i.e. HER-2, pHER-2, pAkt and Ki67; p<0.05); and increased the

expression of the cell apoptotic protein, cleaved-caspase-3 (p<0.05). However, 3%

flaxseed oil was less potent than 3% menhaden or 10% flaxseed oil. The dose-dependent

effect of flaxseed oil clearly demonstrates a role for ALA in cancer prevention. However,

it is not possible to determine whether this effect is directly due to ALA or the conversion

to EPA and DHA. Overall, this study demonstrates that while marine-based n-3 PUFA

are more potent than plant-derived ALA, lifelong exposure to n-3 PUFA, whether from

marine or plant source, can mitigate tumor outcomes.

Page 82: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

70

4.2 Introduction

Breast cancer (BC) is the most common cancer among women worldwide (1, 2).

It is the second leading cause of death for women in Canada and the United States (1, 2).

As estimated in 2014, 24,400 Canadian women will be diagnosed and 5,000 will die from

BC (211). This represents 26% of all newly diagnosed cancer and 14% of all cancer

deaths in Canadian women (211). BC is described based on receptor status, such as

human epidermal growth factor receptor 2 (HER-2) (73). Overexpression of HER-2

occurs in 25-30% of invasive human BC cases, and is associated with aggressive tumor

growth and poor prognoses (215, 216, 218). Though it has yet to be determined what

initially causes the onset of BC, research has found links between dietary habits and BC

risk. Dietary fatty acids in particular have garnered attention for their potential role in

modifying BC risk (32, 159, 164). Epidemiological studies have found significant

differences in BC incidence between populations consuming Asian diets and those

consuming Western diets (8, 25). Asian diets typically include a high intake of fish,

which are enriched in n-3 polyunsaturated fatty acids (n-3 PUFA), and associated with

lower incidence of BC compared to Westerners (8). The marine-derived n-3 PUFA, such

as eicosapentaenoic acid (EPA, 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3), are

the downstream metabolites of α-linolenic acid (ALA, 18:3n-3) (164). In contrast,

Westerners typically consume more n-6 PUFA, such as linoleic acid (LA, 18:2n-6) and

arachidonic acid (AA, 20:4n-6), and often lack recommended doses of n-3 PUFA (0.6%-

1.2% of total fat intake as ALA and 500 mg EPA+DHA per day) (17, 25). Furthermore,

Asians emigrating to Western cultures and adopting local dietary habits have reported

rises in BC incidence reaching rates similar to that of Western countries (24). Thus,

dietary fatty acids may play a critical role in the prevention of BC development.

Though a number of human observational studies have associated n-3 PUFA

intake with a reduction in BC risk, the totality of evidence and the specific mechanisms

responsible for the protective effects of n-3 PUFA remain inconclusive. Emerging

research suggests that early life exposure at the critical period when the mammary gland

(MG) is undergoing extensive modeling and remodeling, may alter the susceptibility to

develop BC during adulthood (29, 168, 219). MG development is unique in that it

Page 83: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

71

develops throughout life; the gland starts as a blank fat pad with an initial primary duct

that starts at the nipple and branches posteriorly throughout the fat pad over the course of

development (168, 220). In rodents, these ducts are led by club-shaped structures called

terminal end buds (TEB), and have been identified as the sites of tumor initiation in MGs,

likely due to the presence of a cap of highly proliferative stem cells on each bud (171,

221, 222). Therefore, MG development is a time of increased vulnerability to

carcinogenic exposures owing to the presence of TEBs. Similar structure in a human

breast is called terminal ductal lobular unit 1(TDLU 1), which appear to be the sites of

BC initiation in most women (169, 172). Exposure to n-3 PUFA during the early life

periods have been shown to decrease the TEB structures in mouse MG and reduce the

risk mammary tumorigenesis later in life (168, 173, 174, 177).

Experimental studies in rodent models have provided more consistent evidence

supporting an anticancer role of n-3 PUFA. The comparative nature of MG development

between humans and murine animals has allowed for numerous transgenic mouse models

of BC to be developed in order to recapitulate molecular pathways activated during

human mammary tumor development (223, 224). The mouse mammary tumor virus

(MMTV)-neu (ndl)-YD5 model is a highly aggressive BC model that overexpress HER-2

via the muring-equivalent neu-oncogene, and results in 50% of the MMTV transgenic

mice to develop mammary tumors within 100 days of age (117, 213, 225). Based on its

highly aggressive phenotype, the MMTV-neu (ndl)-YD5 mouse represents a relevant

model system for elucidating potential strategies aimed at prevention and/or treatment of

HER-2 positive BC (21, 116). Previously, we have demonstrated that lifelong exposure to

n-3 PUFA can mitigate mammary tumor development in mice expressing MMTV-neu

(ndl)-YD5 (116). Intake of a 3% (w/w) menhaden fish oil based diet reduced tumor

volume and multiplicity compared to mice receiving a 10% (w/w) n-6 PUFA diet (116).

Furthermore, a dose-response relationship of 0%, 3% and 9% (w/w) menhaden oil on

tumor size reduction was observed in MMTV-neu (ndl)-YD5 mice, and it was negatively

correlated with a dose-dependent incorporation of EPA and DHA into MG and tumor

phospholipid classes (21). As a result, the incorporation of n-3 PUFA into cellular and

tumor lipids is hypothesized to be an important mechanism by which n-3 PUFA elicit

their anti-tumorigenic effects, although other mechanisms may also be included.

Page 84: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

72

Previous published studies have mainly focused on the protective role of fish

consumption or intake of marine-derived n-3 PUFA, such as EPA and DHA in BC

development (11, 31, 164); whereas the evidence regarding the biological role of the

precursor, ALA, a plant-based n-3 PUFA, remains equivocal. Flaxseed oil (FSO) is one

of the richest plant sources of ALA, where ALA comprises approximately 57% of total

fatty acids (155, 156). Research studies have demonstrated the anti-tumorigenic

properties of flaxseed were mainly attributed to its oil components (78, 159, 182, 205,

209). More importantly, ALA is known as the principal source of n-3 PUFA in Western

diets, but its endogenous conversion to EPA and DHA is not efficient in humans (58,

129). Therefore, the clarification of ALA’s involvement in BC is essential. To the best of

our knowledge no experimental study has directly compared the effects of plant- and

marine-based n-3 PUFA on mammary tumor development. This would help to determine

the relative effectiveness of ALA versus EPA/DHA. Also, it is the fundamental

knowledge for the development of dietary strategies to prevent BC in Western countries.

Therefore, the present study was designed to determine the relative inhibitory potency

and potential mechanisms of lifelong exposure to plant-derived versus marine-based n-3

PUFA on mammary tumor development in female MMTV-neu (ndl)-YD5 mice. The

results from this study may provide guidance regarding the potential use of ALA-rich

foods, such as flaxseed, in the management of BC, and whether FSO can be used as an

alternative dietary supplement in the Western diet when intakes of fish oil, or EPA and

DHA are typically low.

4.3 Material and Methods

4.3.1 Animals and diets

MMTV-neu (ndl)-YD5 mice, on an FVB background, were obtained from an in-

house breeding colony derived from animals originally obtained as a generous gift from

Dr. William Muller (McGill University). Mice were housed in ventilated cages in a

temperature- and humidity- controlled environment and were exposed to a 12 hour light-

dark cycle. Harems consisted of one male heterozygous MMTV-neu (ndl)-YD5 mouse

Page 85: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

73

and three female FVB mice yielding progeny with a wild type or a heterozygous (MMTV)

genotype. All mice had ad libitum access to food and double-distilled water. Harems

were randomly assigned to one of four modified AIN93G diets (Research Diet Inc.): 1)

10% safflower oil (enrich in n-6 PUFA, 0% n-3 PUFA diet), or 2) 3% flaxseed oil+7%

safflower oil (3% plant-based n-3 PUFA diet), or 3) 10% flaxseed oil alone (10% plant-

based n-3 PUFA diet), or 4) 3% menhaden oil+7% safflower oil (3% marine-based n-3

PUFA diet). All diets were isocaloric and provided for 20 kcal% protein, 58 kcal%

carbohydrate and 22 kcal% fat. Diet ingredients and fatty acid composition of the diets

are listed in Tables 4.1 and 4.2, respectively. The mice consuming the 10% safflower oil

(n-6 PUFA diet) were used as a positive control to reflect an n-6 PUFA enriched Western

style diet. The mice fed 3% menhaden fish oil served as a negative control as it is

supplemented with a similar level of EPA and DHA as consumed by Asian countries. 3%

and 10% plant-based n-3 PUFA diets containing different amounts (%w/w) of flaxseed

oil are used to examine the dose-dependent relationship between ALA and BC.

All offspring were weaned and genotyped at 3 weeks of age as described

previously (116). Female transgenic (MMTV) offspring were kept and maintained on

their parental diet while all the males were euthanized post-weaning. Therefore, female

transgenic offspring received the same experimental diet throughout life, from in utero

until termination at 6 or 20 weeks of age. All experimental procedures were approved by

the institutional animal care committee (University of Guelph).

4.3.2 Food intake, body weights, puberty onset

Starting at 3 weeks of age, female mice were checked daily for vaginal opening, a

marker of puberty onset. Food intake and body weights were measured weekly.

Page 86: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

74

Table 4.1 Composition of purified oil diets

Macronutrient

10% safflower 3% flaxseed 10% flaxseed 3% menhaden

g% kcal% g% kcal% g% kcal% g% kcal%

Protein 21 20 21 20 21 20 21 20

Carbohydrate 60 58 60 58 60 58 60 58

Fat 10 22 10 22 10 22 10 22

Total 100 100 100 100

Kcal/g 4.1 4.1 4.1 4.1

Ingredient

gm/Kg

Diet Kcal

gm/Kg

Diet Kcal

gm/Kg

Diet Kcal

gm/Kg

Diet Kcal

Casein 200 800 200 800 200 800 200 800

L-Cystine 3 12 3 12 3 12 3 12

Corn starch 336.7 1347 336.7 1347 336.7 1347 336.7 1347

Maltodextrin 10 132 528 131 528 131 528 132 528

Sucrose 100 400 100 400 100 400 100 400

Cellulose, BW200 50 0 50 0 50 0 50 0

Soybean oil 0 0 0 0 0 0 0 0

Safflower oil 97 873 67.9 611 0 0 67.9 611

Flaxseed oil 0 0 29.1 262 97 873 0 0

Menhaden oil 0 0 0 0 0 0 29.1 262

t-Butylhyfroquinone 0.02 0 0.02 0 0.02 0 0.02 0

Mineral mix 35 0 35 0 35 0 35 0

Vitamin mix 10 40 10 40 10 40 10 40

Choline Bitatrate 2.5 0 2.5 0 2.5 0 2.5 0

Total 966.3 4000 966.3 4000 966.3 4000 966.3 4000

Composition of AIN-93G modified diets with 10% safflower oil, 7% safflower oil+3% flaxseed

oil, 10% flaxseed oil, and 7% safflower oil+3% menhaden oil as provided by manufacture,

Research Diets.

Page 87: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

75

Table 4.2 Fatty acid composition of diets

Fatty acid 10% safflower oil 3% flaxseed oil 10% flaxseed oil 3% menhaden oil

12:0 0.0 0.0 0.0 0.1

14:0 0.2 0.2 0.1 2.7

15:0 0.0 0.0 0.0 0.2

16:0 6.5 6.3 5.8 10.2

16:1c9 0.1 0.1 0.1 0.1

18:0 2.7 2.8 3.2 3.1

18:1c9 16.3 16.0 15.0 14.3

18:1c11 0.7 0.7 0.8 1.4

18:2n6 71.5 55.4 16.5 56.1

18:3n6 0.3 0.2 0.0 0.1

18:3n3 0.2 17.4 57.6 0.6

18:4n3 0.1 0.0 0.0 0.8

20:0 0.4 0.4 0.1 0.4

20:1c11 0.0 0.0 0.1 0.4

20:2n6 0.0 0.0 0.0 0.1

20:3n6 0.0 0.0 0.0 0.1

20:4n6 0.0 0.0 0.0 0.5

20:3n3 0.0 0.0 0.1 0.1

20:5n3 0.0 0.0 0.0 3.9

22:0 0.3 0.0 0.1 0.0

22:1n9 0.0 0.0 0.0 0.1

22:2n6 0.0 0.0 0.0 0.1

22:4n6 0.0 0.0 0.0 0.1

22:3n3 0.0 0.0 0.0 0.0

22:5n6 0.0 0.0 0.0 0.2

22:5n3 0.0 0.0 0.0 0.8

24:0 0.2 0.0 0.0 0.0

22:6n3 0.0 0.0 0.0 3.4

24:1 0.0 0.0 0.0 0.0

Total n-6 71.8 55.7 16.7 57.3

Total n-3 0.4 17.6 57.8 9.5

Total SFA 10.5 9.8 9.5 16.8

Total MUFA 17.3 16.9 16.0 16.3

Total PUFA 72.2 73.3 74.4 66.9

Fatty acid composition (%) of n-6 and n-3 PUFA diets. Lipids were extracted from three

individual pellets from each diet and analyzed by gas chromatography. SFA: saturated fatty acids;

MUFA: monosaturated fatty acids; PUFA: polyunsaturated fatty acid.

Page 88: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

76

4.3.3 Early mammary gland developmental period (6-week-timepoint)

4.3.3.1 Euthanization and tissue collection

At 42 days postnatal, mice were randomly chosen from each diet group (n=12-16)

to assess the effect of dietary n-3 PUFA exposure on MG development. Mice at 6 weeks

of age were selected as previously reported that the number of TEBs in the mouse MGs

typically reaches their maximum during this period. A vaginal smear was taken by

flushing the vagina with 30 µL of a phosphate buffer saline solution. The solution was

then placed on a glass slide and viewed under a Nikon Eclipse TS100 microscope for

estrus cycle classification. Mice were classified as being in one of four estrous cycle

stages including proestrus, estrus, metaestrus and diestrus. If in proestrus, estrus or

metaestrus, mice were euthanized via carbon dioxide asphyxiation followed by cervical

dislocation. If the mice were in diestrus, estrous check was then repeated the next day,

and euthanization was delayed in order to control for the impact of hormone fluctuations

on cell proliferation profiles. This was done for a maximum of two days past the set

euthanization date, at which point estrous stage was checked and recorded, and mice

were euthanized regardless of stage, in order to maintain consistency in MG

measurements.

At necropsy, blood was collected by cardiac puncture, allowed to sit for 30 min

before separating into cells and serum by centrifuge (10000 xg for 5 min), and stored in

an -80˚C freezer. The mouse pelt with MGs attached was removed for wholemounting

analysis. The 5th

MGs were excised, weighed and snap-frozen in liquid nitrogen for fatty

acid analysis.

4.3.3.2 Mammary gland whole mounts

The skin pelt was stretched on corkboard and fixed in 10% formaldehyde. After

48-hour fixation, the 4th

left abdominal MG was dissected from the skin pelt and de-fatted

in acetone for two days (refreshed daily). This was followed by a series of rehydration

steps in decreasing concentrations of 99%, 95% and 70% ethanol, and distilled water for

Page 89: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

77

1 h each and then stained in carmine alum overnight. The following day the glands were

destained in a 2% hydrochloric acid/70% ethanol solution for 1 h, then stepwise

dehydration through an increasing series of graded ethanol (70%, 95%, and 99% each for

1 h) and transferred into xylene overnight. The whole mount MGs were preserved in

heat-sealed polyethylene packages containing 2ml methyl salicylate.

4.3.3.3 Counting of TEB structures

The whole mount mammary glands were coded so that the investigator was

blinded to the identity of the glands. The most undifferentiated TEBs are mostly

concentrated in the periphery of the MGs. The distal portions of MGs were viewed under

a stereoscope at a 10× magnification (10× ocular lens and 1.0× objective lens, Zeiss

Stemi 2000-c) to enumerate number of TEBs and other measures including total fat pad

area, ductal tree area, and length of ductal infiltration. Density of TEBs per MG was

calculated using the formula (number of TEBs) / (ductal tree area). Mammary gland

morphology measures were evaluated blindly by 2 investigators.

4.3.4 Mammary tumor developmental period (20-week-timepoint)

4.3.4.1 Tumor palpation

The remaining female mice continued on the same experimental diet until the 20

weeks of age (n=12 per genotype per diet group). Starting at 10 weeks of age, mice were

palpated for mammary tumor formation. When a new tumor was detected, measurements

of tumor size and multiplicity were recorded 3 times per week for the duration of the

study. Tumors were measured along the sagittal (length) and transverse (width) planes

with the use of digital calipers, and the tumor volume was calculated as follows: [(length)

× (width)2]/2.

Page 90: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

78

4.3.4.2 Euthanization and tissue collection

At 20 weeks of age, all the mice were checked for estrous cycle and euthanized by

CO2 asphyxiation as described above. For ethical reasons mice that lost more than 20% of

their body weight or possessed tumors exceeded either 17mm in length/width or more

than 5000mm3 in volume were euthanized prior to the 20-week time point.

At necropsy, blood and serum samples were first collected as described above.

For tumor-bearing mice, MG and tumors were photographed, and final tumor weight and

volume measurements were recorded. One of the excised primary tumors (per mouse)

was immediately preserved in 10% formaldehyde for immunohistochemistry analysis.

The remainder of the tumors tissues, the 4th

and 5th

MGs were cleaned of surrounding

tissue, removed, weighed and snap frozen in liquid nitrogen for fatty acid and molecular

analysis.

4.3.4.3 Fatty acid analysis

Lipid extraction. Lipids were extracted from serum, MGs and tumor tissues via

the Foch Method (226). In brief, 50ul serum sample from either 6- or 20-week-old mice

was first vortexed with 0.1M KCl and 4ml of CHCl3: MeOH (2:1), and then incubated

overnight at 4˚C to facilitate lipid exaction. Lipids from MGs and tumor tissue were

extracted in a similar manner. The entire 4th

MG from 6-week old mice or the tumor

adjacent MG from 20-week-old mice, was homogenized in 0.1M KCl. For tumor samples,

only 0.1g of the tissue was homogenized in 0.1M KCl. Homogenates were then added to

10ml of CHCl3: MeOH (2:1), vortexed and chilled overnight.

The following day, samples were centrifuged (1460 rpm, 10 min) and the

chloroform layer was then collected, dried down under a gentle stream of nitrogen and

reconstituted to 10mg/ml. Serum samples were examined by total lipid analysis, while

MGs and tumor tissues were analyzed by phospholipid-class TLC in the following

manner. Phospholipid class analysis of MGs and tumors provides more detailed

information regarding potential mechanisms of action.

Page 91: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

79

Total lipid analysis (saponification). 0.5 M KOH prepared in methanol was added

into the serum sample. A C17:0 FFA standard was transferred to each sample using a

Hamilton syringe. Samples were vortexed and saponified at 100˚C for 1 hour. Samples

were checked every 10 min to avoid evaporation.

Phospholipid-class TLC. Lipid samples extracted from MGs and tumor tissues

were spotted on an activated H-plate (EMD Chemicals, #5721-7). The TLC solvent was

made fresh by combining 30ml chloroform, 9ml methanol, 25ml 2-propanol, 6ml of

0.25M KCl and 18ml trimethylamine. The TLC plate was lightly sprayed with 0.1% (w/v)

ANSA (Fluka, #GA12046) and visualized under UV light. Phosphatifylcholine (PC) and

phosphatidylethanolamine (PE) were collected for analysis. A C17:0 FFA standard was

included using a Hamilton syringe.

Methylation. Methylation for serum, MGs and tumor samples was the same.

Hexane and 14% BF3-MeOH (Sigma, N1252) were added to each sample and incubated

for 90 min at 100˚C. Following methylation, 2ml of double distilled H2O was added to

stop methylation. Samples were centrifuged (1460rpm, 10 min), then the hexane layer

was collected and dried down under nitrogen before reconstitution in 2ml of hexane.

Fatty acid methyl esters were analyzed using a gas chromatography system (Agilent

Technologies, 7890B). Fatty acid composition was expressed as a percentage of total

fatty acids.

Sample size from each diet group was as follows: for both 6-and 20-week mice,

serum sample size (n) was equal to 5 per diet; for 6-week mice, 5th

MG tissue was taken

from mice in 10% safflower oil (n=5), 3% flaxseed oil (n=6), 10% flaxseed oil (n=6), and

3% menhaden oil (n=5); for 20-week mice, 4th MG tissue was taken from mice fed with

10% safflower oil (n=5), 3% flaxseed oil (n=4), 10% flaxseed oil (n=6), and 3%

menhaden oil (n=5); tumor tissues were taken from mice consumed 10% safflower oil

(n=6), 3% flaxseed oil (n=5), 10% flaxseed oil (n=5), and 3% menhaden oil (n=5).

Page 92: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

80

4.3.4.4 Western blot analysis

Western blotting was performed on the mouse mammary tumor tissues to analyze

the protein expression of HER2 and phospho-HER2. For each MMTV mice (n=6 per diet

group), a portion (about 20mg) of tumor tissues was homogenized in 1.5ml RIPA buffer

(cell signaling technology, #9806S) containing 15ul protease inhibitors (cell signaling

technology, #5817S). The homogenate was mixed gently on a rotator at 4˚C for 2 hours.

Next, the lysates were clarified by centrifugation at 10000xg at 4˚C for 10 min and

quantified for total protein concentration by using Coomassie Plus Protein Assay kit

(Thermo Sicentific, #1856210). Equal amount of tumor protein extract (20µg) from each

mouse were made up to 25µl with PBS and 5µl of 5×SDS PAGE loading dye. Then the

mixed tumor protein samples were separated by electrophoresis on 7.5% polyacrylamide

gels under denaturing conditions, and transferred to polyvinylidene difluoride membranes.

Membranes were incubated overnight at 4˚C in blocking buffer (5% skim milk in Tris-

buffered saline/0.1% Tween-20, TBST). After blocking, membranes were incubated for 2

hours under room temperature with primary rabbit anti-human antibodies, including

HER2/ErbB2 (1:2000, cell signaling technology, #4290) and Phospho-HER2

(Tyr1221/1222) (1:1000, cell signaling technology, #2243). The primary antibodies were

diluted in TBST containing 3% (v/w) bovine serum albumin (Sigma-Aldrich Canada Ltd).

The membranes were also probed under the same conditions with primary goat anti-

human antibody for β-actin (1:500, Santa Cruz Biotechnology, sc1616) to confirm even

protein loading. After extensive washing in TBST, membranes were probed with

secondary anti-rabbit IgG HRP-linked antibody (1:5000, cell signaling technology #7074)

for 1 hour. For β-actin, secondary donkey anti-goat IgG-HRP (1:2000, Santa Cruz

Biotechnology, sc2020) was used. The bound antibodies were visualized using an

enhanced chemiluminescence kit (PerkinElmer, Inc) and detected in FluorChem Imaging

System (Alpha Innotech). The intensity of bands was quantified using Alpha View SA

(Alpha Innotech), and the relative intensity unit of each tumor protein bands was

normalized with the average intensity of β-actin bands.

Page 93: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

81

4.3.4.5 ELISA

Mouse mammary tumor Akt and phosphor-Akt protein expression was quantified

by an ELISA kit (ebioscience). Tumor proteins were extracted as described previously

and equal amounts of tumor protein extract (40 µg) from each MMTV mouse (n=6 per

diet group) was used for the ELISA measurement. The relative abundance of total Akt

(ebioscience, #85-86047-11) and phospho-Akt 1/2/3 (Ser473) (ebioscience, #85-86042-

11) were measured in all the tumor protein samples according to the manufacturer’s

instructions.

4.3.4.6 Immunohistochemistry analysis (IHC)

To evaluate the dietary effect of n-3 PUFA on tumor cell proliferation and

apoptosis, Ki-67 and cleaved-caspases-3 protein expression levels in mouse mammary

tumor tissues were assessed by IHC. Briefly, paraffin tumor sections (n=6 per diet group)

were deparaffinized in xylene, rehydrated through graded ethanol solutions to distilled

water, and washed in phosphate-buffered saline (PBS). Endogenous peroxidase activity

was quenched by incubating in a dual endogenous enzyme-blocking reagent (Dako,

#S2003) for 5 min. Next, tumor sections were subject to heat-induced antigen retrieval in

a decloaking chamberTM

N×Gen (Biocare Medical) containing 0.01 M citrate buffer

(pH=6.0) According to the tested protein targets, the tumor sections were treated

differentially on boiling temperature and time during the antigen retrieval (i.e. Ki-67,

90°C for 25 min; cleaved-caspase 3, 95°C for 20 min). Nonspecific binding sites were

blocked using 3% (w/v) BSA for 1 hour. Tumor sections were then incubated overnight

at 4°C with 1:200 dilutions of rabbit monoclonal antibody against Ki67 (Abcam,

#ab16667) or 1:1000 dilutions of rabbit polyclonal antibody against cleaved-caspase-3

(Asp 175) (Cell Signaling, #9661). Instead of primary antibody, the tumor section

incubated with PBS was used as a negative control. In the following day, after several

washes with PBS, the tumor sections were incubated with secondary anti-rabbit

EnVision+ System-HRP-labelled polymer (Dako, #K400211-2) for 1 hour at room

temperature. To show antigens, tumor sections were treated with 3, 3’-diaminobenzidine

Page 94: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

82

chromogen solution and its specific substrate buffer (Dako, #K4010) according to the

manufacturer’s instructions for 10 min. The tumor sections were then counterstained with

hematoxylin, dehydrated and mounted. The tumor slides (n=6 per diet group) were read

blindly with a light microscopy (Olympus Bx46, ZK62809) at 400× magnification to

identity the samples. For each slide, 5-10 fields were randomly chosen to photograph by

Q-capture pro-7 system; the number of total cells and of the positive cells of Ki-67 and

cleaved-caspase-3 per field were scored blindly using image J software. Approximately

over 1000 cells were counted per field. Ki-67 and cleaved-caspase-3 labeling index was

expressed as a percentage of total tumor cells, and calculated as follows: (number of

positive cells / total cells counted) ×100.

4.3.5 Statistical analysis

The predetermined upper limit of probability for statistical significance was p ≤

0.05 for all analyses using SASv9.1. A one-way analysis of variance (ANOVA) was

conducted to determine differences in daily food intake, puberty onset, number and

density of TEB structures, tumor latency, final overall tumor weight, fatty acid

composition and protein expression between diet groups, and followed, if justified by

statistical significance, by Tukey’s Studentized Range test. When the data was not

normally distributed, variables were log or square root transformed to adjust for

nonnormality. Log-Rank test was used to analyze the difference in proportion of mice

free of tumors in each diet group throughout the 20-week study period. The Kruskal-

Wallis procedure was performed followed, if justified by statistical significance, by

Wilcoxon two-sample rank sums. A repeated-measures test was conducted for total tumor

volume and tumor multiplicity over the 20-week time course to determine differences

between diet groups over time, as well as for mouse body weight between diet groups

(116). Values are reported as mean SD.

For the purpose of this study, the following definitions for tumor parameters were

used. Tumor latency values were determined as time when the first tumor was palpated.

Tumor multiplicity values were determined by the total number of tumor palpated in each

Page 95: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

83

mouse. Total tumor volume values were calculated as the sum of all tumor volumes for

each mouse. Final tumor volume and multiplicity values were taken when mice reached

20 weeks of age, except the ones required enthanization prior to 20 weeks of age. Due to

ethical reasons, seven of 48 tumor-carrying mice were terminated before the 20 week

time point (4 mice from 10% safflower oil diet and 3 mice from 3% flaxseed oil diet

group were terminated between 18-19 weeks of age).

4.4 Results

4.4.1 Food intake, body weight gain and puberty onset

There were no significant differences between diet groups in daily food intake

(data not shown), body weight change from 3 to 20 weeks of age (Figure 4.1) or tissue

weights measured at euthanization (data not shown). The average daily intake for mouse

is ~2g per day. Timing of vaginal opening is a marker of puberty onset. The onset of

puberty was significantly (p<0.05) delayed in mice fed with n-3 PUFA diets compared to

mice that consumed an n-6 PUFA diet (10% safflower oil, 25.9 ± 0.8 days). A significant

delay (p<0.05) was also observed in mice fed 10% flaxseed oil (29.2 ± 1.6 days)

compared to mice fed 3% flaxseed oil (27.9 ± 1.4 days), 3% menhaden oil group were at

intermediate level (28.2 ± 1.6 days) (Figure 4.2).

Page 96: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

84

Figure 4.1 Mice body weight changes throughout 3 to 20 weeks of age. No significant

difference existed between diet groups determined by repeated measures analysis.

10

15

20

25

30

35

3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Aver

age

bod

y w

eigh

t (g

)

Time (Week)

10% safflower 3% flaxseed 10% flaxseed 3% menhaden

Page 97: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

85

Figure 4.2 Average pubertal onset as measured by vaginal opening in mice fed 10%

safflower oil (n-6 PUFA diet, n=27), 3% flaxseed oil (n-3 PUFA, n= 24), 10% flaxseed

oil (n-3 PUFA, n=24) or 3% menhaden oil (n-3 PUFA, n=26). A one-way ANOVA was

performed; different letters denote significant difference (p<0.05) between diet groups.

4.4.2 TEB structure in 6-week-old mice

At 6 weeks of age, significant differences in the number of TEB structures were

observed in the MG of mice between diet groups. See Figure 4.3 for representative

images of MG taken at 6 weeks of age from each diet group. When compared with mice

consuming 10% safflower oil (an n-6 PUFA diet), lifelong exposure to 10% flaxseed and

3% menhaden oil significantly (p<0.05) reduced the number and density of TEBs;

however, the mice exposed to 3% flaxseed oil did not have any significant effect on

numbers of TEB structures (Figure 4.4). There were no significant differences between

diet groups in ductal tree area and length of ductal infiltration (data not shown).

20

24

28

32

10% safflower (n-6)

3% flaxseed (n-3)

10% flaxseed (n-3)

3% menhaden (n-3)

Days

Post

nata

l

c

b

a

ab

Page 98: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

86

Figure 4.3 Representative stereoscopic wholemount images (20×) of the left fourth MG

of MMTV mice at 6 weeks of age fed a) 10% safflower oil, b) 3% flaxseed oil, c) 10%

flaxseed oil, and d) 3% menhaden oil. Arrows (→) show representative terminal end buds

(TEB) enumerated. Lymph node (LN).

a. 10% safflower

oil

b. 3% flaxseed oil

d. 3% menhaden oil c. 10% flaxseed oil

LN LN

LN LN

Page 99: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

87

Figure 4.4 Effect of lifelong exposure to plant- versus marine-derived n-3 PUFA on the

average total number and density of terminal end buds (TEB) in the MG of mice at 6

weeks of age (n=12-15 mice per diet group). The density of TEBs was calculated using

the following equation: (number of TEBs) / (ductal tree area). Bars not sharing a letter

are significantly different (p < 0.05) based on a one-way ANOVA test.

0

5

10

15

20

25

10% safflower (n-6)

3% flaxseed (n-3)

10% flaxseed (n-3)

3% menhaden (n=3)

Aver

age

nu

mb

er o

f T

EB

s

a

a

bb

(n-3)(n-3)

Page 100: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

88

65

75

85

95

105

115

125

10% safflower (n-6)

3% flaxseed (n-3)

10% flaxseed (n-3)

3% menhaden (n-3)

Days

Post

nata

l

a a

b

b

4.4.3 Tumor latency and tumor free status

Time to palpation of first tumor was tracked in the mice from four different diet

groups. Average tumor latency was delayed (p<0.05) in mice fed 10% flaxseed (105.3 ±

8.2 days) or 3% menhaden oil (103.9 ± 10.8 days) relative to 10% safflower oil (86.7 ±

7.7 days). However, there was no significant difference between mice fed 3% flaxseed

(93.9 ± 7.6 days) and 10% safflower oil diets (Figure 4.5). Similarly, tumor free status

followed the same trend (Figure 4.6). At the T50 threshold, the median age when

mammary tumor was induced in 50% of MMTV mice, n-3 PUFA diets delayed tumor

latency compared to mice fed 10% safflower oil. However, only T50 for mice fed with

either 10% flaxseed (107 days) or 3% menhaden oil (102 days) was significantly (p<0.05)

higher than mice fed 3% flaxseed and 10% safflower oil diets (93 and 86 days,

respectively).

Figure 4.5 Effect of lifelong exposure to plant- versus marine-derived n-3 PUFA on the

average tumor latency in mice (n=12 mice per diet group). Different letters denote

significant (p < 0.05) differences between means.

Page 101: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

89

Figure 4.6 The proportion of mice tumor free throughout the duration of the study fed 10% safflower (n-6 PUFA), 3% flaxseed (n-3

PUFA), 10% flaxseed (n-3 PUFA) or 3%menhaden oil (n-3 PUFA) (n=12 mice per diet group). Significant differences between diet

groups were analyzed by Log-Rank test (p < 0.05).

0

10

20

30

40

50

60

70

80

90

100

110

1 74 79 84 89 94 99 104 109 114 119 124 129 134 139

Pro

po

rtio

n o

f m

ice

tum

or

free

Age of Tumor Onset (day)

10% safflower (n-6)

3% flaxseed (n-3)

3% menhaden (n-3)

10% flaxseed (n-3)

TD 50

b

b

a

a

Page 102: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

90

Figure 4.7 Tumor volume. Average total tumor volume of mice within each diet group was tracked over 20 weeks (n=12 mice per diet

group). Length and width of palpated tumors were recorded, and tumor volume was calculated using the following equation: [length ×

(width) 2

]/2. Measurements were taken three times per week. Different letters denote significant differences (p<0.05) between diet

groups determined by repeated measures analysis.

0

500

1000

1500

2000

2500

3000

3500

4000

4500

5000

10 11 12 13 14 15 16 17 18 19 20

Av

era

ge

tota

l tu

mo

r v

olu

me (

mm

3)

per

mo

use

Time (week)

10% safflower (n-6)a

3% menhaden (n-3)c

10% flaxseed (n-3)c

3% flaxseed (n-3)b

Page 103: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

91

Figure 4.8 Tumor multiplicity. Average total numbers of palpated tumors per mouse within each diet group were tracked over 20

weeks (n=12 mice per diet group). Mice were palpated for tumors three times per week. Different letters denote significant

differences (p<0.05) between diet groups determined by repeated measures analysis.

0

1

2

3

4

5

6

7

10 11 12 13 14 15 16 17 18 19 20

Av

era

ge

tota

l n

um

ber

of

palp

tate

d t

um

ors

per

mou

se

Time (Week)

10% safflower (n-6)

3% Flaxseed (n-3)

3% Menhaden (n-3)

10% Flaxseed (n-3)

a

b

c

d

Page 104: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

92

4.4.4 Tumor volume

Total tumor volume accumulated over time was compared between diet groups by

repeated measures. Total tumor volume in mice fed any of the n-3 PUFA diets was

significantly (p<0.05) less than that carried by mice fed 10% safflower oil over the 20-

week time course (Figure 4.7). In comparison to the 10% safflower oil group, the total

tumor volume began to diverge significantly (p<0.05) at 15 weeks of age for mice fed

either 10% flaxseed or 3% menhaden oil, and at 17 weeks of age for mice fed 3%

flaxseed oil. There was a significant (p<0.05) dose-dependent reduction of total tumor

volume in mice fed with 3 and 10% flaxseed oil. At 20 weeks of age, the final tumor

volumes carried by mice were 24% and 78% lowered in 3% and 10% flaxseed oil groups

relative to 10% safflower oil, respectively.

When compared with 3% menhaden oil, total tumor volume significantly different

starting at week 16 relative to mice fed 3% flaxseed oil; while total tumor volume was

not different between 3% menhaden and 10% flaxseed oil diets. Moreover, 10% flaxseed

oil resulted in a similar reduction in final tumor volume as 3% menhaden oil (78% and

72%, respectively), but 3% flaxseed oil had a much weaker inhibitory effect (p<0.05) on

final tumor volume compare to 3% menhaden oil (24% vs.72%).

4.4.5 Tumor multiplicity

Total tumor multiplicity over time was compared between diet groups by repeated

measures. Feeding any of the n-3 PUFA diets (p<0.05) reduced the number of tumors

palpated in mice compare to 10% safflower oil. This reduction started at week 13 and

maintained until the end of the study at 20 weeks (Figure 4.8). A significant (p<0.05)

dose-dependent reduction was also observed in total tumor multiplicity in mice fed 3%

and 10% flaxseed oil. This was accompanied by a dose-dependent reduction (15% and

42%, respectively) in the final tumor multiplicity of 20-week-old mice on 3% and 10%

flaxseed oil diets relative to 10% safflower oil.

Page 105: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

93

When compared with 3% menhaden oil diet, feeding 10% flaxseed oil was more

potent in reducing total tumor multiplicity (p<0.05), which began to diverge significantly

at week 18 and resulted in a greater reduction in final tumor multiplicity (27% vs. 42%).

However, 3% flaxseed oil was significantly (p<0.05) weaker in reducing total tumor

multiplicity relative to 3% menhaden oil. This significant diverged starting at week 14

and led to a significant differences in final tumor multiplicity (15% vs. 27%) at the end of

20-week time course.

4.4.6 Final total tumor weight

At euthanization, the final total tumor weight carried by mice fed either 10%

flaxseed or 3% menhaden oil was significantly (p<0.05) less than that carried by 10%

safflower oil treated mice (Figure 4.9). However, feeding 3% flaxseed oil did not result in

a reduction in the final total tumor weight compared to 10% safflower oil.

Figure 4.9 Effect of lifelong exposure to plant- versus marine-derived n-3 PUFA on the

final total tumor weight carried by mice at 20 weeks of age (n=12 mice per diet group).

Different letters denote significant (p < 0.05) differences between means.

Page 106: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

94

4.4.7 Fatty acid composition

4.4.7.1 Fatty acid composition in serum

Fatty acid analysis of serum from mice at 6 or 20 weeks of age was conducted to

verify the experimental diet intake (Table 4.3). Serum from 10% safflower oil fed mice

exhibited fatty acid profiles high in LA, AA and n-6 DPA (Docosapentaenoic acid, 22:5

n-6). In comparison, feeding n-3 PUFA diets significantly (p<0.05) increased the serum

level of these fatty acids in both 6-and 20-week-old mice. As expected, there was a

significant decrease in n-6/n-3 PUFA ratio (p<0.05) in the serum of mice exposed to n-3

PUFA diets relative to 10% safflower oil. In comparison to 3% menhaden oil, diets

containing flaxseed oil resulted in a significant (p<0.05) accumulation of ALA into serum,

and further 10% flaxseed oil significantly (p<0.05) increased serum level of EPA.

However, mice fed 3% menhaden oil had the greatest DHA in serum than any other

dietary treatments (p<0.05). Overall, there was no significant difference in serum fatty

acid composition in 6- and 20-week-old mice.

4.4.7.2 Fatty acid composition in mammary glands

Phospholipid analysis of MG from 6- or 20-week mice were assessed to

determine if the observed effects on tumor outcomes were related to the incorporation of

n-3 PUFA into the target tissue. Selected n-6 and n-3 PUFA in phosphatidylethanolamine

(PE) fraction shown in Figure 4.10, and complete details of phosphatidylcholine (PC) and

PE are shown in Table 4.4. Feeding 10% safflower oil increased n-6 PUFA incorporation

into MGs of mice. For both 6- and 20-week mice, there was a significant (p<0.05) dose-

dependent increase in ALA, EPA and n-3 DPA (22:5 n-3) in MGs of mice fed increasing

levels of flaxseed oil in comparison to mice fed 10% safflower oil. Correspondingly, a

significant (p<0.05) dose-dependent decrease in AA was observed. Moreover, exposure

to 3% menhaden oil markedly (p<0.05) increased EPA, DPA (n-3) and DHA content of

MG, with a corresponding decrease in AA and n-6 DPA, when compared with safflower

oil fed mice.

Page 107: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

95

Table 4.3 Serum fatty acid composition of mice at 6 or 20 weeks of age.

-Major n-6 and n-3 PUFA were displayed in the table. Other measured fatty acids are including 12:0, 14:0, 15:0, 16:0, 16:1n-7, 18:0,

18:1n-7, 18:1n-9, 18:3n-6, 20:0, 20:1n-9, 20:2n-6, 20:3n-6, 20:3n-3, 22:0, 22:1n-9, 22:2n-6, 22:4n-6. 22:3n-3, 24:0 and 24:1.

-One-way ANOVA was performed on serum fatty acids of 6- and 20-week-mice independently; different letters denote significant

difference (p < 0.05) between dietary treatments in each age group.

Serum

Fatty acid

6-week-old mice 20-week-old mice

10%

safflower

(n-6)

3% flaxseed

(n-3)

10%

flaxseed

(n-3)

3%

menhaden

(n-3)

10%

safflower

(n-6)

3% flaxseed

(n-3)

10%

flaxseed

(n-3)

3%

menhaden

(n-3)

LA (18:2n-6) 30.8 1.2b 33.6 1.9

a 20.4 2.0

c 33.2 1.0

a 34.6 0.8

a 33.1 0.8

a 21.6 1.1

c 29.6 2.3

b

AA (20:4n-6) 18.9 1.2a 9.1 0.8

b 2.3 0.3

d 6.8 0.5

c 18.4 4.3

a 10.8 1.3

b 3.0 0.4

c 10.6 1.8

b

DPA (22:5n-6) 1.8 0.1a 0.1 0.1

b 0.0 0.0

bc 0.1 0.1

b 1.6 0.2

a 0.0 0.0

b 0.0 0.0

b 0.1 0.0

b

ALA (18:3n-3) 0.1 0.0c 4.1 0.8

b 18.2 1.1

a 0.2 0.1

c 0.1 0.0

c 3.8 0.6

b 16.6 3.6

a 0.1 0.0

c

EPA (20:5n-3) 0.0 0.0d 1.0 0.2

c 6.7 0.6

a 4.2 0.7

b 0.0 0.0

d 0.7 0.1

c 6.2 1.4

a 2.5 0.9

b

DPA (22:5n-3) 0.1 0.0c 0.5 0.1

b 0.8 0.1

a 0.9 0.0

a 0.1 0.1

d 0.4 0.0

c 0.9 0.1

a 0.6 0.1

b

DHA (22:6n-3) 1.0 0.1c 4.1 0.7

b 3.8 0.4

b 6.7 0.3

a 1.1 0.1

c 4.4 0.3

b 4.2 0.7

b 6.8 0.6

a

Total n-6 53.6 1.5a 44.7 1.7

b 23.6 2.4

d 41.8 1.5

c 56.9 3.5

a 45.8 1.4

b 25.6 1.4

c 43.3 2.6

b

Total n-3 1.2 0.2c 10.0 0.8

d 30.4 0.8

a 12.3 0.7

b 1.3 0.2

c 9.7 0.7

b 28.7 2.2

a 10.3 1.0

b

n-6/n-3 Ratio 46.3 7.3a 4.5 0.3

b 0.8 0.1

d 3.4 0.3

c 45.4 5.9

a 4.7 0.4

b 0.9 0.1

c 4.3 0.7

b

Page 108: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

96

Figure 4.10 MG fatty acid composition. Upper panel: PE fraction in 6-week-old mice;

lower panel: PE fraction in 20-week-old mice. The fatty acid composition of MG

phospholipid differs markedly between dietary treatments (n=4-6 per diet). Bars not

sharing a letter are significantly different (p<0.05) based on a one-way ANOVA test. Full

fatty acid profile is displayed in Table 4.4.

Page 109: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

97

When compared with 3% menhaden oil, feeding low (3%) and high (10%)

flaxseed oil diets significantly (p<0.05) increased ALA incorporation into MG tissue.

Notably, mice fed 10% flaxseed oil had significantly (p<0.05) higher level of EPA than

mice fed 3% menhaden oil at 20 weeks, and displayed a significant (p<0.05) reduction of

AA at both age groups. Interestingly, DHA accumulation was significantly (p<0.05)

higher in the mice fed 3% menhaden oil than any flaxseed oil diets. Similar effects were

observed in the PC fraction (Table 4.4, Appendix).

4.4.7.3 Fatty acid composition in mammary tumors

There were significant differences in the fatty acid composition of phospholipid

from mammary tumors in the four different diet groups (n=5-6 per diet), which varied

between PC and PE phospholipid fractions. The PUFA composition in PC and PE are

shown in Figure 4.11 and complete details of all fatty acids are reported in Table 4.5.

Compared to mice fed 10% safflower oil, a significant (PC and PE, P<0.05) dose-

dependent decrease in AA and a significant dose-dependent increase in ALA, EPA and n-

3 DPA were observed in the tumors of mice fed increasing levels of flaxseed oil. In

contrast, a dose-dependent reduction of LA was only observed in tumor PE (p<0.05).

Likewise, tumors from mice receiving a 3% menhaden oil diet had significantly lower

levels of AA and n-6 DPA (PC and PE, p<0.05), higher levels of EPA, n-3 DPA and

DHA (PC and PE, p<0.05), and a significant reduction of LA in tumor PE (p<0.05),

when compared with mice on 10% safflower oil.

Page 110: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

98

Table 4.4 Fatty acid composition of MG phospholipids.

Fatty Acids

Mammary Gland of 20-week-old Mice

PC PE

10%

ssafflower

3%

flaxseed

10%

flaxseed

3%

menhaden

10%

safflower

3%

flaxseed

10%

flaxseed

3%

menhaden

12:0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0

14:0 0.3±0.4 0.2±0.0 0.4±03 0.5±0.2 0.8±0.6 0.8±0.2 1.0±0.6 0.6±0.7

15:0 0.1±0.1b 0.1±0.1

ab 0.1±0.1

a 0.2±0.0

ab 0.5±0.3 0.1±0.1 0.1±0.1 0.1±0.1

16:0 24.5±3.3 21.7±2.1 23.3±2.0 23.8±1.3 10.4±2.2 10.2±1.3 8.1±2.0 11.1±4.6

18:0 16.7±4.2 19.3±1.9 16.5±3.2 19.2±1.6 18.6±1.9 17.5±1.6 15.0±2.5 16.6±4.8

20:0 0.1±0.1 0.2±0.0 0.2±0.0 0.1±0.1 0.2±0.1 0.2±0.0 0.1±0.1 0.1±0.1

22:0 0.2±0.2 0.2±0.0 0.2±0.0 0.2±0.1 0.3±0.1 0.3±0.1 0.1±0.1 0.3±0.2

24:0 0.1±0.2 0.1±0.1 0.0±0.0 0.0±0.0 0.1±0.1 0.0±0.0 0.0±0.0 0.0±0.0

Total SFA 42.0±3.3 41.7±0.3 40.7±2.1 44.0±1.8 30.8±3.4 29.2±2.2 27.0±3.0 29.4±3.9

16:1n-7 1.5±0.6b 1.4±0.2

b 3.4±1.3

a 2.2±0.4

ab 1.6±0.3

b 2.1±0.5

ab 3.0±0.6

a 2.5±0.4

a

18:1n-7 10.4±1.6b 11.1±1.0

b 15.5±1.9

a 9.8±1.2

b 14.3±1.3

bc 15.5±2.0

b 20.1±1.9

a 11.1±2.8

c

18:1n-9 3.0±0.9 2.0±1.2 3.9±1.4 2.7±1.5 1.9±0.5b 2.5±0.4

ab 3.5±1.5

a 2.6±0.4

a

20:1n-9 0.1±0.2 0.2±0.1 0.1±0.2 0.1±0.1 0.4±0.3 0.3±0.3 0.5±0.3 0.4±0.3

22:1n-9 0.3±0.2 0.3±0.1 0.3±0.2 0.2±0.2 0.8±0.7 1.1±0.7 0.5±0.3 1.4±1.0

24:1 0.0±0.0 0.1±0.1 0.1±0.1 0.1±0.1 0.1±0.1ab

0.2±0.2ab

0.1±0.1b 0.4±0.2

a

Total

MUFA 15.3±2.9

b 15.1±1.5

b 23.4±4.4

a 15.1±2.2

b 19.2±1.3

b 21.7±2.2

ab 27.2±3.2

a 18.9±3.9

b

18:2n-6 26.9±5.3ab

30.7±1.3a 22.1±4.4

b 28.2±1.7

ab 18.9±1.3

a 19.8±1.9

a 12.3±3.3

b 13.3±3.3

b

18:3n-6 0.1±0.1 0.1±0.1 0.0±0.0 0.1±0.1 0.0±0.0 0.0±0.0 0.0±0.0 0.1±0.1

18:3n-3 0.0±0.0c 0.8±0.0

b 4.7±1.9

a 0.1±0.0

c 0.1±0.1

c 1.4±0.6

b 5.1±2.6

a 0.0±0.0

c

20:2n-6 1.0±0.2a 0.7±0.1

b 0.3±0.0

d 0.5±0.0

c 0.5±0.0

a 0.4±0.1

a 0.1±0.1

b 0.2±0.2

b

20:3n-6 1.1±0.2 1.0±0.1 09±0.4 1.1±0.2 0.7±0.3 0.9±0.2 1.2±0.8 0.8±0.3

20:4n-6 11.1 2.6a 6.1 0.6

b 2.4 1.0

c 4.9 0.7

b 19.7±1.9

a 14.6±2.0

b 8.0±1.8

c 12.6±4.3

bc

20:3n-3 0.0±0.0c 0.1±0.1

b 0.4±0.1

a 0.2±0.1

b 0.0±0.0

b 0.0±0.0

b 0.3±0.1

a 0.0±0.0

b

20:5n-3 0.0±0.0c 0.2±0.0

c 2.0±0.4

a 1.2±0.5

b 0.1±0.1

d 0.7±0.1

c 6.0±0.9

a 2.1±0.5

b

22:2n-6 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0

22:4n-6 1.0±0.4a 0.4±0.0

b 0.0±0.0

c 0.2±0.1

bc 3.9±0.5

a 1.5±0.4

b 0.3±0.1

c 0.7±0.3

c

22:3n-3 0.0±0.0c 0.1±0.1

b 0.4±0.1

a 0.2±0.1

b 0.0±0.0 0.0±0.0 0.0±0.0 0.1±0.1

22:5n-6 0.7±0.4a 0.1±0.1

b 0.0±0.0

b 0.0±0.0

b 3.0±0.8

a 0.0±0.0

c 0.1±0.1

bc 0.2±0.2

b

22:5n-3 0.0±0.0c 0.6±0.2

b 1.0±0.1

a 0.9±0.1

a 0.4±0.2

d 2.0±0.2

c 4.3±0.7

a 2.7±0.4

b

22:6n-3 0.2±0.2c 1.9±0.5

b 1.4±0.2

b 2.9±0.7

a 2.6±1.0

c 7.8±1.0

b 6.7±0.3

b 11.1±1.5

a

Total n-6 41.9±3.2a 39.0±1.2

ab 25.7±3.3

c 35.0±2.0

b 46.6±4.8

a 37.3±0.9

b 23.2±1.7

c 34.3±1.2

b

Total n-3 0.2±0.2c 3.6±0.8

b 9.8±1.8

a 5.3±1.2

b 3.3±1.5

d 11.8±0.8

c 22.7±2.2

a 17.5±4.0

b

n-6/n-3

ratio 209 16.1

a 11.1 2.0

b 2.7 0.8

d 6.8 1.5

c 17.1 7.7

a 3.2 0.3

b 1.0 0.1

d 2.0 0.3

c

Total PUFA 42.1±3.0a 42.6±1.5

a 35.5±3.2

b 40.3±2.4

a 49.9±3.2

ab 49.1±1.1

ab 46.0±2.6

a 51.8±2.3

b

Page 111: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

99

In comparison to 3% menhaden oil, there was greater incorporation of ALA in

tumors of mice fed increasing levels of flaxseed oil (p<0.05). However, tumors from

mice on low flaxseed oil (3%) diet had a significantly (p<0.05) lower EPA and DHA than

tumors from mice on 3% menhaden oil. Additionally, a significant increase in AA and

decrease in n-3 DPA were observed in the tumor PC of mice fed 3% flaxseed oil

compared with 3% menhaden oil (p<0.05). In contrast, feeding high dose of flaxseed oil

(10%) significantly reduced LA and AA and increased EPA and n-3 DPA incorporation

into tumors compared to mice fed 3% menhaden oil. Notably, DHA accumulation was

the highest (p<0.05) in tumors of mice exposed to 3% menhaden oil. Furthermore, mice

on n-3 PUFA diets also displayed a significant reduction of n-6/n-3 PUFA ratio in both

tumor and adjacent MG tissue compared to n-6 PUFA fed mice (Table 4.4 and 4.5,

p<0.05). In comparison to tumor PC, there was a larger reduction in n-6/n-3 PUFA ratio

in tumor PE, which declined from 16:1 in 10% safflower oil to 3:1 and 2:1 in 3%

flaxseed and 3% menhaden oil, respectively, as well as finally to 1:1 in mice exposed to

10% flaxseed oil (Table 4.5). In addition, the majority of n-3 PUFA in the form of EPA,

n-3 DPA and DHA were accumulated to a greater extent in PE relative to PC.

Page 112: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

100

0

5

10

15

20

25

30

35

18:2n-6 20:4n-6 22:5n-6 18:3n-3 20:5n-3 22:5n-3 22:6n-3

% C

om

po

siti

on

Tumor-PE

10% safflower 3% flaxseed

10% flaxseed 3% menhaden

ab

c b

a

ab

c

b

a

bb bc

b ac d bc

a

c ba

b

a

bb

c

0

5

10

15

20

25

18:2n-6 20:4n-6 22:5n-6 18:3n-3 20:5n-3 22:5n-3 22:6n-3

% C

om

posi

tion

Tumor-PC

10% safflower 3% flaxseed10% flaxseed 3% menhaden

bb

b

a

a

d

c

a cc ab bd

ac bd

ac

b

b b b c

ab b

Figure 4.11 Mammary tumor fatty acid composition. Percentage composition of major n-6 and n-3 PUFA in the representative PC

and PE phospholipid fraction of mouse mammary tumor tissue are shown in the bar graph. Upper panel: PC fraction; lower panel: PE

fraction. The fatty acid composition of tumor phospholipid classes differs markedly between dietary treatments (n=5-6 per diet). Bars

not sharing a letter are significantly different (p<0.05) based on a one-way ANOVA test. Full fatty acid profile is displayed in Table

4.5.

Page 113: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

101

Table 4.5 Fatty acid composition of mammary tumor phospholipids.

Fatty Acids

Mammary Tumors

PC PE

10%

ssafflower

3%

flaxseed

10%

flaxseed

3%

menhaden

10%

safflower

3%

flaxseed

10%

flaxseed

3%

menhaden

12:0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0

14:0 1.3±0.3 1.3±0.2 1.3±0.2 1.0±0.5 0.3±0.1 0.2±0.1 0.2±0.1 0.2±0.1

15:0 0.2±0.0 0.2±0.0 0.2±0.0 0.2±0.0 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0

16:0 31.7±1.2 30.9±0.9 29.6±0.9 27.9±4.6 6.8±1.1 6.7±0.8 6.5±0.5 6.9±0.8

18:0 8.6±1.3 8.6±0.9 7.7±1.4 10.6±3.8 14.1±1.7 13.7±1.9 13.1±2.3 14.8±1.1

20:0 0.1±0.0 0.1±0.0 0.1±0.0 0.1±0.0 0.1±0.0 0.1±0.0 0.1±0.0 0.1±0.0

22:0 0.0±0.0b 0.1±0.0

ab 0.1±0.0

ab 0.1±0.1

a 0.1±0.1 0.1±0.0 0.1±0.0 0.1±0.0

24:0 0.0±0.0 0.1±0.0 0.0±0.0 0.1±0.1 0.1±0.1 0.0±0.0 0.1±0.0 0.0±0.0

Total SFA 42.0±1.3 41.3±1.2 38.9±1.7 40.0±3.8 21.5±2.7 20.8±2.6 19.9±2.0 22.2±1.1

16:1n-7 3.0±0.4b 3.3±0.7

b 5.2±1.0

a 3.3±0.9

b 1.6±0.3

b 1.7±0.3

b 2.7±0.7

a 1.9±0.4

b

18:1n-7 12.7±0.7b 14.1±1.9

b 18.7±1.5

a 12.7±2.7

b 17.8±2.8

b 20.5±3.0

ab 23.9±1.3

a 18.2±1.8

b

18:1n-9 5.0±0.6b 5.4±0.6

ab 7.0±1.2

a 5.1±1.4

b 4.0±1.3

b 4.4±1.2

ab 6.9±2.2

a 4.8±1.1

ab

20:1n-9 0.3±0.1b 0.5±0.2

ab 0.7±0.3

a 0.4±0.2

b 0.5±0.4 0.8±0.2 0.6±0.2 0.5±0.2

22:1n-9 0.1±0.0 0.2±0.1 0.2±0.1 0.1±0.1 0.3±0.3 0.3±0.2 0.2±0.0 0.3±0.1

24:1 0.1±0.0 0.2±0.1 0.3±0.1 0.2±0.2 0.2±0.0 0.2±0.1 0.2±0.1 0.1±0.0

Total MUFA 21.4±1.5b 23.7±1.3

b 32.1±2.0

a 21.9±4.9

b 24.4±4.2

b 27.8±3.9

ab 34.6±3.5

a 25.8±2.8

b

18:2n-6 11.6±1.3b 14.0±0.8

b 11.7±0.2

b 19.3±1.4

a 12.1±4.1

a 10.3±2.1

b 8.0±0.8

c 9.7±0.4

b

18:3n-6 0.2±0.1a 0.1±0.1

ab 0.1±0.0

b 0.1±0.0

b 0.1±0.0 0.1±0.0 0.1±0.0 0.1±0.0

18:3n-3 0.0±0.0c 0.3±0.0

b 1.6±0.4

a 0.1±0.0

c 0.1±0.1

c 0.4±0.2

b 1.6±0.2

a 0.1±0.0

c

20:2n-6 1.5±0.5a 1.4±0.4

a 0.7±0.2

b 1.1±0.4

ab 1.4±0.3

a 1.0±0.2

ab 0.5±0.1

c 0.9±0.3

bc

20:3n-6 2.9±0.6 3.0±1.2 2.5±0.5 2.8±1.1 3.5±1.7 4.0±0.7 3.0±0.6 3.5±0.5

20:4n-6 17.1±1.0a 12.1±0.9

b 4.1±1.1

d 8.4±2.1

c 25.3±2.6

a 22.2±1.6

ab 10.0±1.5

c 18.1±3.8

b

20:3n-3 0.0±0.0c 0.1±0.0

b 0.8±0.3

a 0.1±0.0

c 0.0±0.0

b 0.1±0.0

b 0.4±0.2

a 0.1±0.0

b

20:5n-3 0.0±0.0d 0.3±0.1

c 2.8±0.4

a 1.2±0.6

b 0.1±0.1

d 0.8±0.1

c 6.4±0.9

a 1.4±0.2

b

22:2n-6 0.1±0.1a 0.1±0.0

a 0.0±0.0

b 0.1±0.1

ab 0.1±0.0

a 0.1±0.0

a 0.0±0.0

b 0.1±0.0

a

22:4n-6 0.9±0.1a 0.4±0.1

b 0.1±0.0

d 0.2±0.0

c 3.9±1.1

a 1.3±0.4b 0.2±0.1

c 0.8±0.2

b

22:3n-3 0.0±0.0b 0.1±0.1

b 0.8±0.3

a 0.1±0.0

b 0.0±0.0

b 0.0±0.0

b 0.1±0.0

a 0.0±0.0

b

22:5n-6 0.7±0.3a 0.1±0.0

b 0.1±0.1

b 0.1±0.0

b 4.2±1.7

a 0.2±0.0

b 0.1±0.0

b 0.3±0.0

b

22:5n-3 0.1±0.0d 0.5±0.1

c 1.4±0.2

a 0.9±0.2

b 0.5±0.3

c 2.7±0.6

b 6.2±0.6

a 3.3±0.4

b

22:6n-3 0.8±0.1c 1.7±0.3

b 1.8±0.2

b 3.0±0.6

a 2.9±1.5

c 8.4±1.0

b 9.0±0.7

b 13.7±1.8

a

Total n-6 35.0±1.7a 31.2±1.7

b 19.3±1.0

c 32.1±2.8

ab 50.5±2.2

a 39.1±1.8

b 21.8±2.1

d 33.4±3.1

c

Total n-3 1.0±0.1d 3.0±0.4

c 9.1±1.1

a 5.3±1.2

b 3.6±1.3

d 12.3±0.8

c 23.7±1.8

a 18.6±1.9

b

n-6/n-3

ratio 36.1 5.3

a 10.5 1.7

b 2.14 0.3

d 6.4 1.8

c 15.8 6.5

a 3.2 0.3

b 0.9 0.14

d 1.8 0.3

c

Total PUFA 36.0±1.6a 34.2±1.6

a 28.5±0.7

b 37.4±3.3

a 54.1±3.0

a 51.4±1.6

a 45.5±1.7

b 52.0±2.5

a

Page 114: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

102

0

5

10

15

20

25

10% safflower

(n-6)

3% flaxseed

(n-3)

10% flaxseed

(n-3)

3% menhaden

(n-3)

Rela

tive in

ten

sity

un

it

HER2

185kDa

β-actin

10%saf 3%flax 10%flax 3%men

a ab

bb

0

5

10

15

20

25

10% safflower

(n-6)

3% flaxseed

(n-3)

10% flaxseed

(n-3)

3% menhaden

(n-3)

Rela

tive in

ten

sity

un

it

a

pHER 2

185kDa

β-actin

10%saf 3%flax 10%flax 3%men

bb

a

a. b.

4.4.8 Protein analysis

4.4.8.1 HER-2 and pHER-2

HER-2 (human epidermal growth factor receptor 2) is a 185-kD transmembrane

receptor tyrosine kinase that is involved in human mammary oncogenesis. The term neu

is homologous to HER-2 and used when referring to the murine gene or cDNA (73, 214).

Protein expression of HER-2 and its activated form phosphorylated HER-2 (pHER-2) in

tumors were analyzed by Western Blotting as shown in Figure 4.12. Compared to 10%

safflower oil, exposure to 10% flaxseed or 3% menhaden oil significantly lowered the

protein expression of HER-2 in mouse mammary tumors, and simultaneously down-

regulated the tumor protein expression of pHER-2 (p<0.05). However, HER-2 and

pHER-2 expression was not different between 3% flaxseed oil when compared to mice

fed 10% safflower oil.

Figure 4.12 Effect of lifelong exposure to plant- versus marine-derived n-3 PUFA on

protein expression of (a) HER-2 and (b) pHER-2 in mouse mammary tumors (n=6 per

diet). Different letters denote significant difference (p<0.05) between diet groups was

based on a one-way ANOVA test.

Page 115: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

103

0.00

0.20

0.40

0.60

0.80

1.00

1.20

10% safflower

(n-6)

3% flaxseed

(n-3)

10% flaxseed

(n-3)

3% menhaden

(n-3)

Rela

tive a

bso

rb

an

ce o

f to

tal A

kt

a. Total Akt

0

0.2

0.4

0.6

0.8

1

1.2

1.4

10% safflower

(n-6)

3% flaxseed

(n-3)

10% flaxseed

(n-3)

3% menhaden

(n-3)

Rela

tive a

bso

rb

an

ce o

f p

Ak

t(S

er 4

73)

b. Phospho-Akt (Ser 473)

a

b

bb

4.4.8.2 Akt and pAkt

Akt is a downstream serine/threonine kinase involved in HER-2 signaling

pathways and functions as an anti-apoptotic signaling molecule (94). Protein expression

of total Akt and its activated form pAkt (phosphorylation at Ser473 site) were assessed by

ELISA as shown in Figure 4.13. Although total Akt was not significantly different

between the four dietary treatments, a significant effect was observed in pAktSer 473

. All n-

3 PUFA diets significantly reduced the pAktSer 473

compared to 10% safflower oil fed

mice.

Figure 4.13 Effect of lifelong exposure to plant- versus marine-derived n-3 PUFA on

protein expression of (a) total Akt and (b) pAkt

Ser 473in mouse mammary tumors (n=6 per

diet). Different letters denote significant difference (p<0.05) between diet groups was

based on a one-way ANOVA test.

4.4.8.3 Ki67 and Cleaved-caspase-3

Ki67 protein is expressed by proliferating cells and serves as a biomarker of

tumor cell replication and progression (103). While caspase-3 is a cytoplasmic protein

Page 116: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

104

0

5

10

15

20

25

30

35

10% safflower

(n-6)

3% flaxseed

(n-3)

10% flaxseed

(n-3)

3% menhaden

(n-3)

KI-

67 l

ab

elli

ng i

nd

ex (

%)

a. Ki67

0

2

4

6

8

10

12

10% safflower

(n-6)

3% flaxseed

(n-3)

10% flaxseed

(n-3)

3% menhaden

(n-3)

Cle

aved

-casp

ase

-3 lab

ellin

g I

nd

ex (

%)

b. Cleaved-caspase-3

a

b

c c cc

a

b

that involved in the activation cascade of caspases responsible for cell apoptosis (227,

228). The tumor protein expression of Ki67 and cleaved caspase-3 (activated caspase-3

by a cleavage adjacent to Asp175) were assessed by immunohistochemistry. The results

are presented as labeling index, which is the percentage of labeled cells relative to total

cells (Figure 4.14 a, b). The photomicrographs of representative immunohistochemical

stained mammary tumor section of MMTV mice are shown in Figures 4.15 and 4.16.

When compared to 10% safflower oil, fed mice any of the n-3 PUFA diets can

significantly reduce the expression of Ki67. Moreover, mice exposed to10% flaxseed and

3% menhaden oil had significant lower levels of Ki67 relative to 3% flaxseed oil.

Exposure to 10% flaxseed and 3% menhaden oil resulted in a significant (p<0.05)

increase in cleaved-caspase-3 compared to 10% safflower oil. Mice fed 3% menhaden oil

diet had significantly (p<0.05) higher cleaved-caspase-3 than mice fed 10% flaxseed oil.

However, no significant difference on cleaved-caspase-3 was observed between mice on

3% flaxseed and 10% safflower oil diets.

Figure 4.14 Effect of lifelong exposure to plant- versus marine-derived n-3 PUFA on

protein expression of (a) Ki-67 and (b) cleaved-caspase-3 in mouse mammary tumors

(n=6 per diet). Different letters denote significant difference (p<0.05) between diet

groups was based on a one-way ANOVA test.

Page 117: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

105

Figure 4.15 Immuno-localization of Ki67 in the mammary tumor section of MMTV-neu

(ndl)-YD5 mice fed four different dietary treatments a) 10% safflower oil, b) 3% flaxseed

oil, c) 3% menhaden oil and d) 10% flaxseed oil. Immunohistochemical staining of

paraffinembedded mouse mammary tumor sections stained with Ki-67 antibody (1:200).

Ki67 (dark brown cells) displays a nuclear localization pattern which correlates with it

function in cell cycle progression (magnification: 400×, counterstained with hematoxilin).

Arrows (→) indicate Ki67 immunopositive cells in mouse mammary tumors.

a. 10% safflower

oil

b. 3% flaxseed oil

d. 3% menhaden oil c. 10% flaxseed oil

Page 118: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

106

Figure 4.16 Immuno-localization of cleaved-caspase-3 in the mammary tumor section of

MMTV-neu (ndl)-YD5 mice fed four different dietary treatments a) 10% safflower oil, b)

3% flaxseed oil, c) 3% menhaden oil and d) 10% flaxseed oil. Immunohistochemical

staining of paraffinembedded mouse mammary tumor sections stained with cleaved-case-

ase-3 antibody (1:1000). Cleaved-caspase-3 (cells with dark brown outer layers) displays

a cytoplasmic localization pattern which correlates with it function in cell cycle

progression (magnification: 400×, counterstained with hematoxylin). Arrows (→)

indicate cleaved-caspase-3 immunopositive cells in mouse mammary tumors.

a. 10% safflower oil b. 3% flaxseed oil

d. 3% menhaden oil c. 10% flaxseed oil

Page 119: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

107

4.5 Discussion

The present study is the first to compare the relative inhibitory potency of plant-

versus marine-derived n-3 PUFA on mammary tumor development. Epidemiological and

experimental studies to date provide evidence in support of a beneficial effect of marine-

based n-3 PUFA in reducing the risk of developing BC. However, a major gap remaining

is substantiating the anti-cancer effect of plant-derived ALA, the principle source of n-3

PUFA consumed in North American. Using the MMTV-neu (ndl)YD5 transgenic mouse

model, the present study demonstrated that lifelong exposure to n-3 PUFA, whether from

plant or marine sources, can prevent breast carcinogenesis and mitigate tumor outcomes

through modulating MG morphology, and signaling protein molecules involved in cell

proliferation and apoptosis. Conservatively, marine-based n-3 PUFA (EPA and DHA)

were 8 times more potent than plant-derived ALA in inhibiting mammary tumorigenesis.

4.5.1 Role of plant- and marine-derived n-3 PUFA in MG development and BC risk

First, the present study provides evidence that the onset of puberty was

significantly (p<0.05) delayed in mice fed either flaxseed or menhaden oil diet as

compared to mice fed 10% safflower oil. Pubertal onset has been linked to BC as earlier

menarches place females at a greater risk of BC (229-231). Therefore, lifelong exposure

to both plant- and marine-derived n-3 PUFA may reduce the risk of BC. Moreover,

estrogen is known as a key regulator of puberty, given that elevated estrogen is typically

associated with accelerated pubertal onset (168, 219, 230). This suggests that n-3 PUFA

may influence circulating estrogen levels. Hilakivi-Clarke et al. has shown that maternal

intake of n-3 PUFA diets significantly increased pregnancy 17β-estradiol levels, but

decreased mammary tumor incidence among the female rat offspring (232).

Unfortunately, serum estrodiol level was not assessed in the present study; future

investigation is needed to verify the role of n-3 PUFA in regulating hormonal effects on

puberty and risk of BC.

Page 120: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

108

Second, feeding either 10% flaxseed oil or 3% menhaden oil significantly (p<0.05)

decreased the number and density of TEBs in the MG of mice at 6 weeks of age when

compared with 10% safflower oil. TEBs are directly associated with MG development

and BC risk since they are tumor initiation sites that give rise to mammary tumors upon

exposure to a chemical carcinogen (169). It has been proposed that more TEBs at the

time the gland is exposed to a carcinogen, the higher the risk of BC (168). These findings

demonstrate that lifelong exposure to both plant- and marine-derived n-3 PUFA can

influence MG development at an early life stage, and may lower future BC risk. However,

at the same dosage, 3% menhaden oil was more effective in reducing TEBs relative to 3%

flaxseed oil, given that analysis of TEB structures showed no significant difference

between mice fed 3% flaxseed and 10% safflower oil. As a result, marine-based n-3

PUFA was more potent than plant-derived ALA in altering MG structures. These

observations were attributed to the type and level of PUFA present in MGs of mice at 6

weeks of age. An enrichment of n-3 PUFA and a corresponding decrease in AA in mice

fed flaxseed and menhaden oil diets contributed to reduced TEB structures at 6 weeks

(Figure 4.10). More specifically, a significant (p<0.05) increase of ALA and EPA in mice

fed 10% flaxseed oil, as well as higher levels of EPA and DHA in mice fed 3% menhaden

oil was negatively associated with the number of TEBs in MG. Therefore, changes in

fatty acid composition of MG during early periods of rapid growth and development can

influence the long term health of mammary tissue and modify the risk of mammary

tumorigenesis later in life.

The findings from the present study are in agreement with previous rodent studies.

Exposure of female rats to ~3.5% (w/w) menhaden oil or 4% (w/w) flaxseed oil at

discrete developmental stages such as in utero, during suckling to puberty, or throughout

lifetime, significantly (p<0.05) lowered the density of TEBs and subsequently decreased

mammary tumor incidence (110, 173, 174, 176, 220, 233). In contrast, exposure after

weaning had no beneficial effects on TEB structures (173), which suggests timing of

dietary exposure is an important factor to consider in MG development. Moreover, a

recent study showed that female rat offspring exposed to marine n-3 PUFA enriched fish

oil in utero via maternal intake had a greater effect on preventing mammary tumors than

supplementation during puberty or adulthood (29). Therefore, early life stages such as in

Page 121: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

109

utero and during suckling are critical periods for inducing structural changes in MG that

reduce BC risk later in life.

4.5.2 Role of plant- and marine-derived n-3 PUFA in mammary tumor development

Multiple parameters in this present study suggest a preventive effect of plant- and

marine-derived n-3 PUFA on BC development. Compared to 10% safflower oil, intake of

10% flaxseed and 3% menhaden oil delayed tumor latency; whereas no significant effect

on tumor latency was found when mice were fed a low (3%) flaxseed oil diet. Likewise,

mice exposed to either 10% flaxseed or 3% menhaden oil diets were tumor free longer

over the whole 20-week-period relative to mice fed 3% flaxseed oil. This demonstrated

that marine-based n-3 PUFA was more effective than plant-derived ALA in delaying

mammary tumor onset.

Mammary tumor development was assessed by measuring tumor volume and

multiplicity which was significant (p<0.05) lower in mice fed either flaxseed or

menhaden oil diet relative to mice fed 10% safflower oil. These results demonstrate that

lifelong exposure to both plant- and marine-derived n-3 PUFA can attenuate aspects of

mammary tumorigenesis in mice expressing MMTV-neu (ndl)YD5, a relevant model of

HER-2 positive BC. In addition, the reduced tumor multiplicity also demonstrated that

both plant- and marine-derived n-3 PUFA can inhibit subsequent tumor initiation.

Interestingly, a significant dose-dependent reduction in both tumor volume and

multiplicity was found in mice fed increasing levels of flaxseed oil. This provides strong

evidence that mammary tumor development was dose-dependently inhibited by plant-

derived ALA in female MMTV-neu (ndl)YD5 mice. Although 3% menhaden oil was

more potent than 3% flaxseed oil in decreasing tumor volume and multiplicity,

supplementation of a high (10%) flaxseed oil diet resulted in a similar reduction of tumor

volume and a greater reduction of tumor multiplicity. Furthermore, overall final tumor

weight was consistent with the observed effects on tumor growth. Exposure to 10%

flaxseed and 3% menhaden oil produced a similar reduction in final tumor weight, which

was significantly (p<0.05) lower than the final tumor weight observed in mice fed 3%

Page 122: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

110

flaxseed and 10% safflower oil. Since 3% menhaden oil diet contains 3.9% EPA and 3.4%

DHA, and 10% flaxseed oil diet contains 57.6% ALA, it is concluded that plant-derived

ALA is ~1/8 as potent as marine-based n-3 PUFA (EPA and DHA) in inhibiting

mammary tumorigenesis in MMTV-neu (ndl)YD5 mouse model.

Observations from the present study reinforce our previous findings which have

also shown beneficial outcomes of marine-derived n-3 PUFA in mitigating mammary

tumor development. In one study, the anti-tumorigenic effect of marine-based n-3 PUFA

was examined via both complementary genetic and conventional dietary approaches

(116). Mice carrying the fat-1 transgene, which are capable of endogenously producing n-

3 PUFA from dietary n-6 PUFA, were crossed with MMTV-neu (ndl)YD5 mouse model

to create a novel double-hybrid progeny (116, 234). It was shown that both in the genetic

arm as well as, and more importantly, in the dietary arm of the study, the volume and

number of tumors was significantly decreased in mice fed marine-based n-3 PUFA.

Particularly, final tumor volume and multiplicity carried by MMTV mice that receiving a

3% (w/w) menhaden oil diet were 64% and 40% reduced in comparison to MMTV mice

fed 10% (w/w) safflower oil, respectively (116). Consistent with previous finding, the

same menhaden oil diet was used in the present study and showed a similar reduction

(72%) in final tumor volume and a slightly smaller reduction (27%) in final tumor

multiplicity, when compared with 10% safflower oil. Furthermore, in a recent study, we

examined the dose-response relationship between dietary marine-based n-3 PUFA and

mammary tumor development (21). A significant dose-dependent reduction was observed

in tumor multiplicity in MMTV-neu (ndl)YD5 mice fed increasing levels of menhaden oil

(21). In support of these findings, Yee et al. (2005) examined mammary tumor

development in MMTV-neu mice fed menhaden oil from 7-8 weeks of age onwards and

showed a significant reduction in tumor incidence (30%) and final tumor multiplicity

(55%) by the end of 15 month (115). The MMTV-neu mouse model utilized in this study

is much less aggressive than MMTV-neu (ndl)YD5, and thus requiring a period to induce

mammary tumors (117). More importantly, our study used less amount of menhaden oil

than this study, but the inhibitory effect on tumor multiplicity in our study was greater.

This suggests a potential benefit of lifelong exposure to n-3 PUFA in our study may add

protection against mammary tumor development.

Page 123: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

111

A few studies have also investigated the protective effect of plant-derived n-3

PUFA on MMTV-c-neu mouse model. One study looked at the effect of increasing levels

of flaxseed oil (0.05, 0.1 and 0.2 ml) on mammary tumor development, and demonstrated

that daily exposure to 0.2 ml flaxseed oil (equivalent to 186 mg/day) from 4 weeks of age

onwards delayed tumor incident and reduced overall tumor weight (208). This effective

dose (186 mg/day) of flaxseed oil was comparable to the amount provided by a 10%

(w/w) flaxseed oil diet in the present study (average mouse daily intake is 200 mg/day).

However, lifelong exposure to 10% (w/w) flaxseed oil had more diverse anti-tumorigenic

effects, such as reducing tumor volume and multiplicity. This further emphasizes the

importance of investigating the effect of lifelong exposure in the current study. Moreover,

the anti-tumorigenic effect of a low dose of flaxseed oil was also observed. It has been

shown that mice fed a 0.5% (w/w) flaxseed diet, which contain ~0.2 (w/w) flaxseed oil,

significantly reduced the tumor incidence and the numbers of large tumors (> 6mm

diameter) (190). In contrast, 3% (w/w) flaxseed oil in the present study had no significant

effect on tumor latency. The reason for this discrepancy may be due to the lignan

component in the flaxseed diet. Flaxseed is the richest source of mammalian lignan

precursors, namely secoisolariciresinol diglycoside (SDG), which is known as a type of

phytoestrogen that have estrogenic or anti-estrogenic properties (157, 162, 163). Thus,

the anticancer effect of the 0.5% (w/w) flaxseed diet may be partially attributed to SDG.

Altogether, findings from the current study and several rodent studies employing MMTV-

neu related models provide concordant evidence for an anti-cancer effect of both plant-

and marine-derived n-3 PUFA on modulating neu-related mammary tumor development.

This is critical given that nearly one third of invasive human BC cases are HER-2/neu

related (215, 216).

4.5.3 Potential mechanisms of anti-tumorigenic actions

4.5.3.1 Change membrane fatty acid composition

Total lipid analysis of serum from 20-week mice reflected the type and level of

PUFA from the diets. The anti-tumorigenic action of n-3 PUFA may be attributed to

Page 124: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

112

changes in membrane fatty acid composition of both tumor tissue and the adjacent MG

tissue. Analysis of tumor and adjacent MG phospholipid fatty acid composition revealed

significant increases in n-3 PUFA, particularly ALA, EPA and DHA, as well as

significant decreases in AA and overall n-6/n-3 PUFA ratio in mice fed flaxseed or

menhaden oil diet compared to 10% safflower oil (p<0.05). A significant (p<0.05)

increase was also found in n-3 DPA content, demonstrating that EPA is incorporated and

further elongated. These changes in membrane fatty acid composition suggest a potential

inhibitory effect of n-3 PUFA on the synthesis of eicosanoid derived from AA.

Overlapping metabolic pathways of n-6 and n-3 PUFA are likely to be one of the most

salient mechanisms by which n-3 PUFA inhibited tumor development. Both AA and EPA

are the substrate for eicosanoid synthesis; while eicosanoids derived from AA are pro-

inflammatory and cancer promoting, in contrast to EPA and DHA, which are anti-

inflammatory and cancer inhibitory (31, 58, 61). Thus, intake of plant- and marine-

derived n-3 PUFA reduced AA incorporation and thus, less AA was available to produce

pro-inflammatory eicosanoids, and hence prevent BC development. In addition, these

changes may also affect plasma membrane properties. N-3 PUFA can significantly affect

membrane fluidity, permeability, and resident protein activity (52, 235).

Analysis of tumor and adjacent MG phospholipids fatty acid composition also

suggests a differential role of individual n-3 PUFA on cancer prevention. Specifically, the

observed dose-dependent reductions on tumor outcomes with increased flaxseed oil

intake were mainly associated with a dose-dependent increase in ALA and EPA. An

earlier study reported in vivo and in vitro evidence that ALA was the likely bioactive

anticancer agent (78). Unfortunately, in the present study we cannot determine whether

the anti-tumorigenic effect was directly attribute to ALA itself, or the downstream

metabolic products, such as EPA and DHA. Although the conversion of ALA to EPA and

DHA is limited, supplementation of the flaxseed oil diets also resulted in a higher

incorporation of EPA and DHA relative to ALA into tumor membranes. This apparent

mismatch between dietary intakes and levels of incorporation may indicate a negative

selection for ALA into tumor cell membrane phospholipids, or preferred in conversion to

EPA and DHA. Thus, it will be important to determine the independent effect of ALA in

the future studies.

Page 125: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

113

Moreover, the observed tumor inhibitory effect of 3% menhaden oil was primarily

related to significant increases in EPA and DHA (p<0.05), and more importantly, it is

noted that DHA is considerably more concentrated than EPA in tumor and adjacent MG

tissue. This observation may indicate a preference by tumor cells for DHA over EPA.

Interestingly, evidence has shown that DHA was more predominantly localized to the sn-

2 position on the glycerol backbone of glycerophospholipids compare to EPA (236, 237);

the position was usually linked to an n-6 PUFA, such as AA (52). With the increased

intake of marine-derived n-3 PUFA, more DHA may replace AA at the sn-2 position of

glycerophospholipid relative to EPA. Therefore, we proposed that DHA may be the more

bioactive component of menhaden oil that exerts these anti-tumorigenic effects. This was

in accordance with a recent analysis that demonstrated DHA was more effective than EPA

in suppressing mammary carcinogenesis (120). Furthermore, it is interesting to note there

is a higher level of marine-based n-3 PUFA incorporated in tumor PE relative to PC.

Previous studies also noted a similar observation in brain tissue, which demonstrated the

proportion of PUFA in PE, particularly DHA, was much greater than in PC (238-240). In

addition, a significant (p<0.05) higher incorporation of LA in tumor PE was observed in

mice fed 3% menhaden oil compared to 10% safflower oil. This may indicate a feedback

inhibition of delta-6 desaturase, which is the rate-limiting enzyme responsible for both

the synthesis of AA from LA and the conversion of EPA and DHA from ALA(241). The

presences of DHA or EPA act as a feedback inhibitor of delta-6 desaturase that may

reduce the flow of LA to AA, causing the accumulation of LA in tumor tissue.

Nevertheless, the incorporation of these n-3 PUFA into both the MGs and tumor tissue

demonstrates their direct involvement in modulating mammary tumor development,

which warrants further investigation into mechanisms of action.

4.5.3.2 Modulate cellular signaling molecules

Recent evidence suggest that membrane fatty acid compositional changes may

perturb lipid rafts, which are enriched with many regulatory proteins and growth factor

receptors (52, 235). Lipid rafts are important heterogeneous membrane microdomains

Page 126: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

114

that are rich in cholesterol and saturated phospholipids that, when clustered together,

function as operating platforms for signaling molecules (52, 235) . The highly

unsaturated nature of n-3 PUFA make them sterically incompatible with cholesterol,

which disrupt the formation of lipid rafts and thus, suppress raft-mediated cell signaling

transductions (52, 235). The MMTV-neu (ndl)YD5 model used in the current study

represents HER-2 positive BC, therefore, the signaling pathways related to HER-2 over-

expression were considered. HER-2 falls under the family of epidermal growth receptors,

which are involved in BC progression when over-expressed or mutated (214). Once

HER-2 protein is activated, the phsophotyrosine residues serve as docking sites for the

adaptor proteins, such as Shc and Grb2, resulting in the PI3K/Akt and mitogen-activated

protein kinase pathway activation, which promote cell survival and proliferation (242). In

the present study, 3% menhaden and 10% flaxseed oil reduced HER-2 and pHER-2

protein expression in mouse mammary tumors, which then led to down-regulation of

pAkt protein expression downstream. Although the low (3%) flaxseed oil diet did not

affect tumor protein expression of HER-2 or pHER-2, a significant (p<0.05) reduction

was observed in pAkt. These results matched with the observed effects on tumor

outcomes in MMTV-neu (ndl)YD5 mice, which suggest the anti-tumorigenic actions of

plant- and marine-derived n-3 PUFA may in part be due to reduced growth factor

signaling.

Several independent studies have provided consistent data showing that

exogenous supplementation with plant- or marine-derived n-3 PUFA, especially ALA and

DHA, can decrease the expression of HER-2 oncoprotein in BT-474 and SkBr-3 human

BC cells that naturally amplify the HER-2 oncogene (5, 138). Similarly, in athymic mice

implanted with HER-2-overexpressing BT-474 cells, feeding 8% (w/w) flaxseed oil was

found to reduce tumor cell proliferation, and this was likely due to the lowered protein

levels of pHER-2 and pAkt (207, 243). Akt was also shown to be susceptible to EPA,

since the treatment of EPA can decrease protein expression of both total and pAkt in

transfected MCF-7 cells (97). More importantly, treatment of EPA and DHA

independently was shown to disrupt the formation of lipid rafts in BC cell lines and led to

growth arrest (6, 22, 56). These observations were accompanied by the inhibition of

signaling initiated by growth factor receptors, such as HER2 and epidermal growth factor

Page 127: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

115

receptor (EFGR) (6, 22, 56). These supportive data may help to explain the differential

effect of plant- and marine-based n-3 PUFA in the current study. Compared to ALA, the

higher degree of unsaturation of EPA and DHA may lead to a greater disruption of lipid

raft, and consequently affect HER-2 mediated signaling pathways. Although little is

known about lipid rafts, evidence from previous studies and the present study support the

idea that n-3 PUFA may be a useful dietary treatment for controlling HER-2-mediated

oncogenesis. Future work will need to examine the effects of plant-derived ALA on lipid

raft disruption.

To further determine the potential mechanism of these anti-tumorigenic actions,

expression of proteins involved in cell proliferation and apoptosis were examined.

Exposure to either flaxseed or menhaden oil diets was shown to reduce tumor protein

expression of Ki67, a nuclear protein widely used as a prognostic or predictive marker of

BC (103). However, at the same dosage, 3% menhaden oil was more potent in reducing

the expression of Ki67 compared to 3% flaxseed oil. These results suggest that marine-

based n-3 PUFA is more effective than plant-derived ALA in inhibiting cell proliferation.

It is also consistent with previous in vitro studies demonstrating that a higher dosage of

ALA (72μM) is required to achieve similar inhibitory effect as EPA (50μM) and DHA

(30μM) at lower levels (86, 124, 146). However, it is not appropriate to compare the

effective dosage of individual n-3 PUFA from different BC cell lines.

In addition to cell proliferation, exposure to flaxseed or menhaden oil diet also

affected tumor cell apoptosis. It has been well-established that apoptosis is regulated and

executed by the activated cysteine-aspartic proteases, called caspases, which comprise

two distinct classes, the initiators and the effectors (228, 244). Caspase-3 is a key

executioner of apoptosis, which is cleaved by initiator caspases at Asp28 or Asp175 to

generate the active large (p17) and small (p12) subunits, forming an active heterotetramer

(228). In the present study, intake of 3% menhaden and 10% flaxseed oil significantly

(p<0.05) increased tumor protein expression of cleaved-caspase-3 compare to 10%

safflower oil, and this elevation was greatest in mice fed a 3% menhaden oil diet than any

other flaxseed oil diets. This demonstrated that dietary exposure to n-3 PUFA can

promote cell apoptosis, but marine-based n-3 PUFA was more potent than plant-derived

Page 128: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

116

ALA. Furthermore, it is noted that the actions of n-3 PUFA on modulating protein

expression involved in cell proliferation and apoptosis are consistent with their inhibitory

effect on tumor outcomes. This suggests that plant- and marine-derived n-3 PUFA

manifest their anti-tumorigenic properties by inhibiting cell growth and promoting tumor

cell death.

4.5.4 Physiological relevance to human intake

In regards to physiological relevance, all the n-3 PUFA diets in the present study

provided mice with 22% of total calories from fat. Based on the average mouse daily

intake (2 g/day), the 3% and 10% flaxseed oil diets provided 35 mg/day and 115 mg/day

of ALA, respectively. These amounts corresponding to 3.8% and 12.5% of total calories

per day, respectively. The doses of ALA in the 3% and 10% flaxseed oil diets calculated

on the basis of total caloric intake are equivalent to human doses of 8.4 g and 27.8 g of

ALA per day, respectively, assuming 2000 kcal of caloric intake per person. These

amounts are much higher compared to the typical North American diet that provides

approximately 1.4 g of ALA per day (15). While these dosages of ALA seem unrealistic,

an earlier clinical study showed the effectiveness of daily intake of 25 g flaxseed (5.7

g/day of ALA) in reducing tumor cell proliferation and HER-2 protein expression, and

increasing apoptosis in tumors of postmenopausal BC patients (50). However, it cannot

rule out the beneficial effect of the SDG component in this flaxseed diet.

When considering the marine-based n-3 PUFA diet in the present study,

supplementation of 3% menhaden oil provided mice with 7.8 mg/day of EPA and 6.8

mg/day of DHA. In total, EPA and DHA accounted for 1.6% of the total daily calories

consumed by the mice. In terms of human intake, it is similar to a traditional Japanese

diet which contains 1-2% of daily energy as EPA and DHA, and corresponds to 3.5 g of

EPA and DHA per day (245). Given that 100 g salmon provided approximately 2.2 g of

EPA and DHA, the equivalent quantity of EPA and DHA from the 3% menhaden oil diet

is achievable in humans through the consumption of ~2 servings of 75g salmon steak a

day. A previous cohort study demonstrated that daily doses up to 7.56 g EPA+DHA, for 6

Page 129: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

117

month, were well tolerated with excellent compliance in participants at high risk of BC

(118). Further, they found that daily intake of 1.7 g of EPA+DHA either from canned

salmon+albacore or capsule supplements increased EPA and DHA content of breast

adipose tissue over 3 months without significant differences between treatment arms

(246). These clinical data are supportive, and provide evidence that the equivalent dosage

of EPA and DHA from the 3% menhaden oil diet is tolerated by humans, and may

effective in increasing EPA and DHA incorporation into breast adipose tissue of women

to decrease BC risk.

4.6 Conclusions

In conclusion, the present study has shown that lifelong exposure to both plant-

and marine-derived n-3 PUFA can influence MG development and prevent mammary

tumoirgenesis in an aggressive HER-2 positive BC model. These anti-tumorigenic effects

of n-3 PUFA are mediated by altering fatty acid composition in the target tissue and

modulating the expression of tumor proteins involved in cell proliferation and apoptosis.

These findings support the potential importance of plant- and marine derived n-3 PUFA

in maternal and childhood diets throughout the lifecycle as a simple and effective means

of improving overall MG health and mitigating lifelong BC risk. Also, this study has, for

the first time, demonstrated that plant-derived ALA is only 1/8 as potent as marine-based

n-3 PUFA, especially EPA and DHA. This knowledge is very important for the

development of dietary strategies to prevent BC in Western countries.

Page 130: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

118

Chapter Five

General Discussion, Limitations, Future Directions and

Concluding Remarks

Page 131: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

119

5.1 General Discussion

Studies have shown that fish consumption or intake of marine-derived long-chain

n-3 PUFA, such as EPA and DHA, may prevent mammary carcinogenesis. However, the

anticancer effect of ALA, a plant-based and major n-3 PUFA in the Western diet, remains

equivocal. The present study has for the first time compared the relative potency between

plant- and marine-derived n-3 PUFA on mammary tumor development in MMTV-neu-

(ndl)YD5 mice, an aggressive transgenic mouse model of HER-2 positive BC. First, this

study has clearly shown that lifelong exposure to plant and marine-derived n-3 PUFA can

delay puberty onset, reduce the number of TEB and changed the fatty acid profile in the

MG of mice during the early life. These early changes in MG had a lasting impact on the

long term health of mammary tissue, influencing MG development and subsequently

lowering the risk of mammary tumorigenesis later in life. Second, this study provided

convincing evidence that lifelong exposure to plant and marine-derived n-3 PUFA can

attenuate mammary tumor development by reducing tumor volume and multiplicity, as

well as overall final tumor weight. As phospholipid membranes are the sites for

communication between cells and their environment, it is likely that n-3 PUFA exert their

anti-tumorigenic effect through the alteration in the membrane fatty acid composition in

tumor and MG tissue, and consequently HER-2-mediated signaling and tumor protein

expression related to cell proliferation and apoptosis. Most importantly, this study

demonstrates plant-derived ALA is only 1/8 as potent as marine-based n-3 PUFA in

inhibiting mammary tumorigenesis. This is useful for providing recommendations for

improving the dietary strategies and reducing BC risk in Western countries, where the

intake of EPA/DHA is considerably low (15). The fact that a food nutrient, like n-3 PUFA,

can have a significant effect on mammary tumor development is remarkable and as such

has considerable implications in the field of BC prevention. This study also emphasized

the potential importance of lifetime exposure, from conception onwards, throughout all

critical periods of development. Thus, it is important for pregnant mothers and women

planning to conceive, as well as young girls, to increase their n-3 PUFA intake in order to

mitigate lifelong BC risk.

Page 132: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

120

5.2 Limitations

Despite careful consideration of variables and confounding factors, a few

limitations are identified in this study. First, the MMTV-neu-(ndl)YD5 mouse model is

specific to tumors over-expressing HER-2, and though this type of mammary

tumroigenesis occurs in about 25-30% of invasive human BC cases, the nature of the

disease is very complex (215, 216). Indeed, BC can be divided into several molecular

subtypes based on the expression of cell estrogen receptor, progesterone receptor and

HER2 (209). In addition, BC cannot be assumed to have only one pathway of action

initiating and progressing tumor growth. Other mechanisms of mammary tumor

development are likely involved. This study can speculate that lifelong exposure to plant-

and marine-derived n-3 PUFA would likely attenuate the tumor growth of other subtypes

of BC.

The objective of this study was to compare the relative potency between marine-

based n-3 PUFA, mainly EPA and DHA, and plant-derived ALA on mammary tumor

development. Due to the nature of the MMTV-neu (ndl)YD5 mouse model, the

endogenous production of EPA and DHA from ALA obtained from the flaxseed oil diets

still remains. As a result, we cannot rule out the protective effect of EPA and DHA on

tumor development of mice that consumed flaxseed oil diets. Although the dose-

dependent effect of flaxseed oil demonstrates a role for ALA in cancer prevention, it is

not possible to determine whether this effect is directly due to ALA itself or in part from

the conversion of ALA to EPA and DHA.

The present study emphasized the importance of n-3 PUFA exposure throughout

life, however, it was not designed to investigate the protective role of n-3 PUFA exposure

at various stages of life cycle in MMTV-neu (ndl)YD5 mice. Thus, it is difficult to

predict the effect of plant and marine-derived n-3 PUFA exposure during important

developmental stages, such as in utero, during suckling, post-weaning, and in adulthood.

Also, we cannot compare the anti-tumorigenic effect of early life exposure to n-3 PUFA

versus n-3 PUFA exposure during adulthood.

Finally, the positive control diet utilized in the present study contained 10% (w/w)

Page 133: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

121

safflower oil which is abundant in LA (~72% LA in total fatty acids) and scarce in other

n-6 PUFA. The n-6 and n-3 PUFA families refer to many structurally similar molecules

which may have different biological effects. Therefore, in this study, we can only

conclude that in comparison to an LA rich n-6 PUFA diet, plant- and marine- derived n-3

PUFA are protective against mammary tumor development. Furthermore, the

interpretation may differ if saturated or monounsaturated fatty acids are used.

5.2 Future Directions

The results from this study demonstrate that dietary exposure to plant- and

marine-derived n-3 PUFA affected tumor protein expression of HER-2, pHER-2, pAkt

and cleaved-casepase-3, and thus it will be of interest to determine the direct effects of n-

3 PUFA on these protein targets at a transcriptional level. Moreover, additional analysis

of expression of genes involved in eicosanoid biosynthesis, inflammatory process, cell

proliferation and apoptosis will also be of interest, to provide a comprehensive

understanding of how plant- and marine-derived n-3 PUFA modulate molecular

mechanisms underlying BC development and progression.

To date, studies examined the protective roles of n-3 PUFA on lipid raft-mediated

signaling pathways remain inconclusive. Future analysis will be of interest to investigate

the effect of plant- and marine-derived n-3 PUFA on the formation of lipid raft in

mammary tumor tissue. More specifically, it will be important to know whether the

anticancer effect of n-3 PUFA was due to decrease the protein level of HER-2 or

inhibition of HER-2 phosphorylation in lipid rafts.

Estrogen is known as an important factor involved in the development and

progression of BC. Future analysis of serum estradiol and other hormones (progesterone,

etc.) will be of interested to assess whether dietary exposure to plant- and marine-derived

n-3 PUFA could modulate the metabolism of these hormones and further modify the risk

of BC.

Trastuzumab (TRAS, Herceptin™) is the most commonly prescribed drugs for

Page 134: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

122

HER-2 positive BC (140). It will be of interest to investigate the interactions between

plant- or marine-derived n-3 PUFA and TRAS in future studies. This will help to

determine whether dietary of plant- or marine-derived n-3 PUFA can interfere, enhance,

or have no effect on the effectiveness of TRAS in mitigating mammary tumor

development in MMTV-neu (ndl)-YD5 mice.

The next logical step to take from here would be to develop a novel double-hybrid

mouse model by crossing of a MMTV-neu-(ndl)-YD5 mouse with a delta-6 desaturase

knockout model, which blocking the conversion of ALA to EPA and DHA. This double-

hybrid mouse model will allow us to compare the independent effect of ALA versus that

of EPA and DHA on mammary tumorigenesis. From that, more definitive

recommendations can be made for using plant- or marine-derived n-3 PUFA as nutritional

interventions in the prevention and treatment of BC.

5.3 Concluding Remarks

This study has shown that lifelong exposure to plant- and marine-derived n-3

PUFA can influence MG development and attenuate mammary tumorigenesis in a

relevant model of HER-2 positive BC. However, plant-derived ALA was only 1/8 as

potent compared to marine-based EPA and DHA. The current study provides evidence for

potential dietary modulation of specific BC subtypes and warrants further investigation in

prospective human trials of BC prevention. It also supports the therapeutic potential of

ALA, EPA and DHA supplementation in the treatment of HER-2-positive BC. Nutritional

intervention provides a promising approach to enhancing conventional therapeutics

without negatively affecting a patient's quality of life. In light of these findings,

Westerners that typically consume low levels of n-3 PUFA should be encouraged to

increase their dietary intake of n-3 PUFA enriched food products, including fish or

flaxseed, especially for pregnant mothers and young females for reducing lifelong BC

risk.

Page 135: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

123

Chapter Six

References and Appendix

Page 136: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

124

References

1. DeSantis, C., Ma, J., Bryan, L. & Jemal, A. (2014) Breast cancer statistics, 2013. CA

Cancer J. Clin. 64: 52-62.

2. Canadian Cancer Statistics 2013. Canadian Cancer Society's Streering Committee on

Cancer Statistics. Canadian Cancer Society. 2013.

3. Gerber, M. (2012) Omega-3 fatty acids and cancers: a systematic update review of

epidemiological studies. Br. J. Nutr. 107 Suppl 2: S228-S239.

4. Iwasaki, M. & Tsugane, S. (2011) Risk factors for breast cancer: epidemiological

evidence from Japanese studies. Cancer Sci. 102: 1607-1614.

5. Menendez, J. A., Lupu, R. & Colomer, R. (2005) Exogenous supplementation with

omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA; 22:6n-3) synergistically

enhances taxane cytotoxicity and downregulates Her-2/neu (c-erbB-2) oncogene

expression in human breast cancer cells. Eur. J. Cancer Prev. 14: 263-270.

6. Ravacci, G. R., Brentani, M. M., Tortelli, T., Jr., Torrinhas, R. S., Saldanha, T., Torres,

E. A. & Waitzberg, D. L. (2013) Lipid raft disruption by docosahexaenoic acid induces

apoptosis in transformed human mammary luminal epithelial cells harboring HER-2

overexpression. J. Nutr. Biochem. 24: 505-515.

7. Zheng, J. S., Hu, X. J., Zhao, Y. M., Yang, J. & Li, D. (2013) Intake of fish and marine

n-3 polyunsaturated fatty acids and risk of breast cancer: meta-analysis of data from 21

independent prospective cohort studies. BMJ 346: f3706.

8. Simopoulos, A. P. (2002) The importance of the ratio of omega-6/omega-3 essential

fatty acids. Biomed. Pharmacother. 56: 365-379.

9. Saadatian-Elahi, M., Norat, T., Goudable, J. & Riboli, E. (2004) Biomarkers of dietary

fatty acid intake and the risk of breast cancer: a meta-analysis. Int. J. Cancer 111: 584-

591.

10. Boyd, N. F., Stone, J., Vogt, K. N., Connelly, B. S., Martin, L. J. & Minkin, S. (2003)

Dietary fat and breast cancer risk revisited: a meta-analysis of the published literature. Br.

J. Cancer 89: 1672-1685.

11. Anderson, B. M. & Ma, D. W. (2009) Are all n-3 polyunsaturated fatty acids created

equal? Lipids Health Dis. 8: 33.

12. Zou, Z., Bidu, C., Bellenger, S., Narce, M. & Bellenger, J. (2014) n-3 polyunsaturated

fatty acids and HER2-positive breast cancer: interest of the fat-1 transgenic mouse model

over conventional dietary supplementation. Biochimie 96: 22-27.

13. Michael Murray (13 A.D.) omega-3 polyunsaturated fatty acids and their metabolites.

Phytochem rev 13: 139-156.

Page 137: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

125

14. Akhtar, S., Ismail, T. & Riaz, M. (2013) Flaxseed - a miraculous defense against

some critical maladies. Pak. J. Pharm. Sci. 26: 199-208.

15. Kris-Etherton, P. M., Harris, W. S. & Appel, L. J. (2002) Fish consumption, fish oil,

omega-3 fatty acids, and cardiovascular disease. Circulation 106: 2747-2757.

16. Institute of Medicine. Dietary Reference Intakes for Energy, Carbohydrate, Fiber, Fat,

Fatty Acids, Cholesterol, Protein, and Amino Acids (Macronutrients). 2002.

Washington DC, National Academy Press.

17. Vannice, G. & Rasmussen, H. (2014) Position of the academy of nutrition and

dietetics: dietary fatty acids for healthy adults. J. Acad. Nutr. Diet. 114: 136-153.

18. Bagga, D., Anders, K. H., Wang, H. J. & Glaspy, J. A. (2002) Long-chain n-3-to-n-6

polyunsaturated fatty acid ratios in breast adipose tissue from women with and without

breast cancer. Nutr. Cancer 42: 180-185.

19. Chajes, V., Torres-Mejia, G., Biessy, C., Ortega-Olvera, C., ngeles-Llerenas, A.,

Ferrari, P., Lazcano-Ponce, E. & Romieu, I. (2012) omega-3 and omega-6

Polyunsaturated fatty acid intakes and the risk of breast cancer in Mexican women:

impact of obesity status. Cancer Epidemiol. Biomarkers Prev. 21: 319-326.

20. Rose, D. P., Connolly, J. M., Rayburn, J. & Coleman, M. (1995) Influence of diets

containing eicosapentaenoic or docosahexaenoic acid on growth and metastasis of breast

cancer cells in nude mice. J. Natl. Cancer Inst. 87: 587-592.

21. Leslie, M. A., Abdelmagid, S. A., Perez, K., Muller, W. J. & Ma, D. W. (2014)

Mammary tumour development is dose-dependently inhibited by n-3 polyunsaturated

fatty acids in the MMTV-neu(ndl)-YD5 transgenic mouse model. Lipids Health Dis. 13:

96.

22. Schley, P. D., Brindley, D. N. & Field, C. J. (2007) (n-3) PUFA alter raft lipid

composition and decrease epidermal growth factor receptor levels in lipid rafts of human

breast cancer cells. J. Nutr. 137: 548-553.

23. Silva, V., Barazzoni, R. & Singer, P. (2014) Biomarkers of fish oil omega-3

polyunsaturated fatty acids intake in humans. Nutr. Clin. Pract. 29: 63-72.

24. Ziegler, R. G., Hoover, R. N., Pike, M. C., Hildesheim, A., Nomura, A. M., West, D.

W., Wu-Williams, A. H., Kolonel, L. N., Horn-Ross, P. L. et al. (1993) Migration

patterns and breast cancer risk in Asian-American women. J. Natl. Cancer Inst. 85: 1819-

1827.

25. Singh, G. K. & Hiatt, R. A. (2006) Trends and disparities in socioeconomic and

behavioural characteristics, life expectancy, and cause-specific mortality of native-born

and foreign-born populations in the United States, 1979-2003. Int. J. Epidemiol. 35: 903-

919.

Page 138: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

126

26. Manna, S., Janarthan, M., Ghosh, B., Rana, B., Rana, A. & Chatterjee, M. (2010) Fish

oil regulates cell proliferation, protect DNA damages and decrease HER-2/neu and c-

Myc protein expression in rat mammary carcinogenesis. Clin. Nutr. 29: 531-537.

27. Manna, S., Chakraborty, T., Ghosh, B., Chatterjee, M., Panda, A., Srivastava, S.,

Rana, A. & Chatterjee, M. (2008) Dietary fish oil associated with increased apoptosis and

modulated expression of Bax and Bcl-2 during 7,12-dimethylbenz(alpha)anthracene-

induced mammary carcinogenesis in rats. Prostaglandins Leukot. Essent. Fatty Acids 79:

5-14.

28. Manna, S., Chakraborty, T., Damodaran, S., Samanta, K., Rana, B. & Chatterjee, M.

(2007) Protective role of fish oil (Maxepa) on early events of rat mammary

carcinogenesis by modulation of DNA-protein crosslinks, cell proliferation and p53

expression. Cancer Cell Int. 7: 6.

29. Su, H. M., Hsieh, P. H. & Chen, H. F. (2010) A maternal high n-6 fat diet with fish

oil supplementation during pregnancy and lactation in rats decreases breast cancer risk in

the female offspring. J. Nutr. Biochem. 21: 1033-1037.

30. Hardman, W. E., Avula, C. P., Fernandes, G. & Cameron, I. L. (2001) Three percent

dietary fish oil concentrate increased efficacy of doxorubicin against MDA-MB 231

breast cancer xenografts. Clin. Cancer Res. 7: 2041-2049.

31. Larsson, S. C., Kumlin, M., Ingelman-Sundberg, M. & Wolk, A. (2004) Dietary long-

chain n-3 fatty acids for the prevention of cancer: a review of potential mechanisms. Am.

J. Clin. Nutr. 79: 935-945.

32. Terry, P. D., Rohan, T. E. & Wolk, A. (2003) Intakes of fish and marine fatty acids

and the risks of cancers of the breast and prostate and of other hormone-related cancers: a

review of the epidemiologic evidence. Am. J. Clin. Nutr. 77: 532-543.

33. Stoll, B. A. (2002) N-3 fatty acids and lipid peroxidation in breast cancer inhibition.

Br. J. Nutr. 87: 193-198.

34. MacLennan, M. & Ma, D. W. (2010) Role of dietary fatty acids in mammary gland

development and breast cancer. Breast Cancer Res. 12: 211.

35. Wakai, K., Tamakoshi, K., Date, C., Fukui, M., Suzuki, S., Lin, Y., Niwa, Y., Nishio,

K., Yatsuya, H. et al. (2005) Dietary intakes of fat and fatty acids and risk of breast

cancer: a prospective study in Japan. Cancer Sci. 96: 590-599.

36. Gago-Dominguez, M., Yuan, J. M., Sun, C. L., Lee, H. P. & Yu, M. C. (2003)

Opposing effects of dietary n-3 and n-6 fatty acids on mammary carcinogenesis: The

Singapore Chinese Health Study. Br. J. Cancer 89: 1686-1692.

37. Murff, H. J., Shu, X. O., Li, H., Yang, G., Wu, X., Cai, H., Wen, W., Gao, Y. T. &

Zheng, W. (2011) Dietary polyunsaturated fatty acids and breast cancer risk in Chinese

women: a prospective cohort study. Int. J. Cancer 128: 1434-1441.

Page 139: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

127

38. Shannon, J., King, I. B., Lampe, J. W., Gao, D. L., Ray, R. M., Lin, M. G., Stalsberg,

H. & Thomas, D. B. (2009) Erythrocyte fatty acids and risk of proliferative and

nonproliferative fibrocystic disease in women in Shanghai, China. Am. J. Clin. Nutr. 89:

265-276.

39. Kim, J., Lim, S. Y., Shin, A., Sung, M. K., Ro, J., Kang, H. S., Lee, K. S., Kim, S. W.

& Lee, E. S. (2009) Fatty fish and fish omega-3 fatty acid intakes decrease the breast

cancer risk: a case-control study. BMC. Cancer 9: 216.

40. Brasky, T. M., Lampe, J. W., Potter, J. D., Patterson, R. E. & White, E. (2010)

Specialty supplements and breast cancer risk in the VITamins And Lifestyle (VITAL)

Cohort. Cancer Epidemiol. Biomarkers Prev. 19: 1696-1708.

41. Goodstine, S. L., Zheng, T., Holford, T. R., Ward, B. A., Carter, D., Owens, P. H. &

Mayne, S. T. (2003) Dietary (n-3)/(n-6) fatty acid ratio: possible relationship to

premenopausal but not postmenopausal breast cancer risk in U.S. women. J. Nutr. 133:

1409-1414.

42. Thiebaut, A. C., Chajes, V., Gerber, M., Boutron-Ruault, M. C., Joulin, V., Lenoir, G.,

Berrino, F., Riboli, E., Benichou, J. & Clavel-Chapelon, F. (2009) Dietary intakes of

omega-6 and omega-3 polyunsaturated fatty acids and the risk of breast cancer. Int. J.

Cancer 124: 924-931.

43. Stripp, C., Overvad, K., Christensen, J., Thomsen, B. L., Olsen, A., Moller, S. &

Tjonneland, A. (2003) Fish intake is positively associated with breast cancer incidence

rate. J. Nutr. 133: 3664-3669.

44. Witt, P. M., Christensen, J. H., Schmidt, E. B., Dethlefsen, C., Tjonneland, A.,

Overvad, K. & Ewertz, M. (2009) Marine n-3 polyunsaturated fatty acids in adipose

tissue and breast cancer risk: a case-cohort study from Denmark. Cancer Causes Control

20: 1715-1721.

45. Maillard, V., Bougnoux, P., Ferrari, P., Jourdan, M. L., Pinault, M., Lavillonniere, F.,

Body, G., Le, F. O. & Chajes, V. (2002) N-3 and N-6 fatty acids in breast adipose tissue

and relative risk of breast cancer in a case-control study in Tours, France. Int. J. Cancer

98: 78-83.

46. Patterson, R. E., Flatt, S. W., Newman, V. A., Natarajan, L., Rock, C. L., Thomson, C.

A., Caan, B. J., Parker, B. A. & Pierce, J. P. (2011) Marine fatty acid intake is associated

with breast cancer prognosis. J. Nutr. 141: 201-206.

47. Kuriki, K., Hirose, K., Wakai, K., Matsuo, K., Ito, H., Suzuki, T., Hiraki, A., Saito, T.,

Iwata, H. et al. (2007) Breast cancer risk and erythrocyte compositions of n-3 highly

unsaturated fatty acids in Japanese. Int. J. Cancer 121: 377-385.

48. Shannon, J., King, I. B., Moshofsky, R., Lampe, J. W., Gao, D. L., Ray, R. M. &

Thomas, D. B. (2007) Erythrocyte fatty acids and breast cancer risk: a case-control study

in Shanghai, China. Am. J. Clin. Nutr. 85: 1090-1097.

Page 140: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

128

49. Signori, C., DuBrock, C., Richie, J. P., Prokopczyk, B., Demers, L. M., Hamilton, C.,

Hartman, T. J., Liao, J., El-Bayoumy, K. & Manni, A. (2012) Administration of omega-3

fatty acids and Raloxifene to women at high risk of breast cancer: interim feasibility and

biomarkers analysis from a clinical trial. Eur. J. Clin. Nutr. 66: 878-884.

50. Thompson, L. U., Chen, J. M., Li, T., Strasser-Weippl, K. & Goss, P. E. (2005)

Dietary flaxseed alters tumor biological markers in postmenopausal breast cancer. Clin.

Cancer Res. 11: 3828-3835.

51. Menendez, J. A. & Lupu, R. (2007) Fatty acid synthase and the lipogenic phenotype

in cancer pathogenesis. Nat. Rev. Cancer 7: 763-777.

52. Gu, Z., Suburu, J., Chen, H. & Chen, Y. Q. (2013) Mechanisms of omega-3

polyunsaturated fatty acids in prostate cancer prevention. Biomed. Res. Int. 2013: 824563.

53. Wassall, S. R. & Stillwell, W. (2008) Docosahexaenoic acid domains: the ultimate

non-raft membrane domain. Chem. Phys. Lipids 153: 57-63.

54. Chenais, B. & Blanckaert, V. (2012) The janus face of lipids in human breast cancer:

how polyunsaturated Fatty acids affect tumor cell hallmarks. Int. J. Breast Cancer 2012:

712536.

55. Murai, T. (2012) The role of lipid rafts in cancer cell adhesion and migration. Int. J.

Cell Biol. 2012: 763283.

56. Rogers, K. R., Kikawa, K. D., Mouradian, M., Hernandez, K., McKinnon, K. M.,

Ahwah, S. M. & Pardini, R. S. (2010) Docosahexaenoic acid alters epidermal growth

factor receptor-related signaling by disrupting its lipid raft association. Carcinogenesis 31:

1523-1530.

57. Ma, D. W., Seo, J., Davidson, L. A., Callaway, E. S., Fan, Y. Y., Lupton, J. R. &

Chapkin, R. S. (2004) n-3 PUFA alter caveolae lipid composition and resident protein

localization in mouse colon. FASEB J. 18: 1040-1042.

58. Burdge, G. C. & Calder, P. C. (2005) Conversion of alpha-linolenic acid to longer-

chain polyunsaturated fatty acids in human adults. Reprod. Nutr. Dev. 45: 581-597.

59. Young, L. R., Kurzer, M. S., Thomas, W., Redmon, J. B. & Raatz, S. K. (2011)

Effect of dietary fat and omega-3 fatty acids on urinary eicosanoids and sex hormone

concentrations in postmenopausal women: a randomized controlled feeding trial. Nutr.

Cancer 63: 930-939.

60. Healy, D. A., Wallace, F. A., Miles, E. A., Calder, P. C. & Newsholm, P. (2000)

Effect of low-to-moderate amounts of dietary fish oil on neutrophil lipid composition and

function. Lipids 35: 763-768.

Page 141: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

129

61. Benoit, V., Relic, B., Leval, X., X, Chariot, A., Merville, M. P. & Bours, V. (2004)

Regulation of HER-2 oncogene expression by cyclooxygenase-2 and prostaglandin E2.

Oncogene 23: 1631-1635.

62. Richards, J. A. & Brueggemeier, R. W. (2003) Prostaglandin E2 regulates aromatase

activity and expression in human adipose stromal cells via two distinct receptor subtypes.

J. Clin. Endocrinol. Metab 88: 2810-2816.

63. Kelley, D. S., Taylor, P. C., Nelson, G. J., Schmidt, P. C., Ferretti, A., Erickson, K. L.,

Yu, R., Chandra, R. K. & Mackey, B. E. (1999) Docosahexaenoic acid ingestion inhibits

natural killer cell activity and production of inflammatory mediators in young healthy

men. Lipids 34: 317-324.

64. Wei, N., Wang, B., Zhang, Q. Y., Mi, M. T., Zhu, J. D., Yu, X. P., Yuan, J. L., Chen,

K., Wang, J. & Chang, H. (2008) Effects of different dietary fatty acids on the fatty acid

compositions and the expression of lipid metabolic-related genes in mammary tumor

tissues of rats. Nutr. Cancer 60: 810-825.

65. Serhan, C. N. (2005) Novel eicosanoid and docosanoid mediators: resolvins,

docosatrienes, and neuroprotectins. Curr. Opin. Clin. Nutr. Metab Care 8: 115-121.

66. Serhan, C. N., Arita, M., Hong, S. & Gotlinger, K. (2004) Resolvins, docosatrienes,

and neuroprotectins, novel omega-3-derived mediators, and their endogenous aspirin-

triggered epimers. Lipids 39: 1125-1132.

67. Weylandt, K. H., Kang, J. X., Wiedenmann, B. & Baumgart, D. C. (2007) Lipoxins

and resolvins in inflammatory bowel disease. Inflamm. Bowel. Dis. 13: 797-799.

68. Kang, J. X. & Weylandt, K. H. (2008) Modulation of inflammatory cytokines by

omega-3 fatty acids. Subcell. Biochem. 49: 133-143.

69. Calder, P. C. (2006) n-3 polyunsaturated fatty acids, inflammation, and inflammatory

diseases. Am. J. Clin. Nutr. 83: 1505S-1519S.

70. Zandi, R., Larsen, A. B., Andersen, P., Stockhausen, M. T. & Poulsen, H. S. (2007)

Mechanisms for oncogenic activation of the epidermal growth factor receptor. Cell Signal.

19: 2013-2023.

71. Bhargava, R., Gerald, W. L., Li, A. R., Pan, Q., Lal, P., Ladanyi, M. & Chen, B.

(2005) EGFR gene amplification in breast cancer: correlation with epidermal growth

factor receptor mRNA and protein expression and HER-2 status and absence of EGFR-

activating mutations. Mod. Pathol. 18: 1027-1033.

72. Corsetto, P. A., Montorfano, G., Zava, S., Jovenitti, I. E., Cremona, A., Berra, B. &

Rizzo, A. M. (2011) Effects of n-3 PUFAs on breast cancer cells through their

incorporation in plasma membrane. Lipids Health Dis. 10: 73.

Page 142: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

130

73. Hynes, N. E. & Stern, D. F. (1994) The biology of erbB-2/neu/HER-2 and its role in

cancer. Biochim. Biophys. Acta 1198: 165-184.

74. Tovey, S. M., Brown, S., Doughty, J. C., Mallon, E. A., Cooke, T. G. & Edwards, J.

(2009) Poor survival outcomes in HER2-positive breast cancer patients with low-grade,

node-negative tumours. Br. J. Cancer 100: 680-683.

75. Moasser, M. M. (2007) The oncogene HER2: its signaling and transforming functions

and its role in human cancer pathogenesis. Oncogene 26: 6469-6487.

76. Bollig-Fischer, A., Dziubinski, M., Boyer, A., Haddad, R., Giroux, C. N. & Ethier, S.

P. (2010) HER-2 signaling, acquisition of growth factor independence, and regulation of

biological networks associated with cell transformation. Cancer Res. 70: 7862-7873.

77. Yee, L. D., Agarwal, D., Rosol, T. J., Lehman, A., Tian, M., Hatton, J., Heestand, J.,

Belury, M. A. & Clinton, S. K. (2013) The inhibition of early stages of HER-2/neu-

mediated mammary carcinogenesis by dietary n-3 PUFAs. Mol. Nutr. Food Res. 57: 320-

327.

78. Truan, J. S., Chen, J. M. & Thompson, L. U. (2010) Flaxseed oil reduces the growth

of human breast tumors (MCF-7) at high levels of circulating estrogen. Mol. Nutr. Food

Res. 54: 1414-1421.

79. Desvergne, B. & Wahli, W. (1999) Peroxisome proliferator-activated receptors:

nuclear control of metabolism. Endocr. Rev. 20: 649-688.

80. Georgiadi, A. & Kersten, S. (2012) Mechanisms of gene regulation by fatty acids.

Adv. Nutr. 3: 127-134.

81. Gurnell, M., Savage, D. B., Chatterjee, V. K. & O'Rahilly, S. (2003) The metabolic

syndrome: peroxisome proliferator-activated receptor gamma and its therapeutic

modulation. J. Clin. Endocrinol. Metab 88: 2412-2421.

82. Flachs, P., Rossmeisl, M., Bryhn, M. & Kopecky, J. (2009) Cellular and molecular

effects of n-3 polyunsaturated fatty acids on adipose tissue biology and metabolism. Clin.

Sci. (Lond) 116: 1-16.

83. Kliewer, S. A., Sundseth, S. S., Jones, S. A., Brown, P. J., Wisely, G. B., Koble, C. S.,

Devchand, P., Wahli, W., Willson, T. M. et al. (1997) Fatty acids and eicosanoids

regulate gene expression through direct interactions with peroxisome proliferator-

activated receptors alpha and gamma. Proc. Natl. Acad. Sci. U. S. A 94: 4318-4323.

84. Clay, C. E., Namen, A. M., Atsumi, G., Willingham, M. C., High, K. P., Kute, T. E.,

Trimboli, A. J., Fonteh, A. N., Dawson, P. A. & Chilton, F. H. (1999) Influence of J

series prostaglandins on apoptosis and tumorigenesis of breast cancer cells.

Carcinogenesis 20: 1905-1911.

Page 143: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

131

85. Mueller, E., Sarraf, P., Tontonoz, P., Evans, R. M., Martin, K. J., Zhang, M., Fletcher,

C., Singer, S. & Spiegelman, B. M. (1998) Terminal differentiation of human breast

cancer through PPAR gamma. Mol. Cell 1: 465-470.

86. Barascu, A., Besson, P., Le, F. O., Bougnoux, P. & Jourdan, M. L. (2006) CDK1-

cyclin B1 mediates the inhibition of proliferation induced by omega-3 fatty acids in

MDA-MB-231 breast cancer cells. Int. J. Biochem. Cell Biol. 38: 196-208.

87. Jiang, W., Zhu, Z., McGinley, J. N., El, B. K., Manni, A. & Thompson, H. J. (2012)

Identification of a molecular signature underlying inhibition of mammary carcinoma

growth by dietary N-3 fatty acids. Cancer Res. 72: 3795-3806.

88. Elmore, S. (2007) Apoptosis: a review of programmed cell death. Toxicol. Pathol. 35:

495-516.

89. Jacobson, M. D., Burne, J. F., King, M. P., Miyashita, T., Reed, J. C. & Raff, M. C.

(1993) Bcl-2 blocks apoptosis in cells lacking mitochondrial DNA. Nature 361: 365-369.

90. van Slooten, H. J., Clahsen, P. C., van Dierendonck, J. H., Duval, C., Pallud, C.,

Mandard, A. M., obelle-Deroide, A., van, d., V & van, d., V (1996) Expression of Bcl-2

in node-negative breast cancer is associated with various prognostic factors, but does not

predict response to one course of perioperative chemotherapy. Br. J. Cancer 74: 78-85.

91. Oltvai, Z. N., Milliman, C. L. & Korsmeyer, S. J. (1993) Bcl-2 heterodimerizes in

vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell 74:

609-619.

92. Raisova, M., Hossini, A. M., Eberle, J., Riebeling, C., Wieder, T., Sturm, I., Daniel, P.

T., Orfanos, C. E. & Geilen, C. C. (2001) The Bax/Bcl-2 ratio determines the

susceptibility of human melanoma cells to CD95/Fas-mediated apoptosis. J. Invest

Dermatol. 117: 333-340.

93. Ion, G., Akinsete, J. A. & Hardman, W. E. (2010) Maternal consumption of canola oil

suppressed mammary gland tumorigenesis in C3(1) TAg mice offspring. BMC. Cancer

10: 81.

94. Vivanco, I. & Sawyers, C. L. (2002) The phosphatidylinositol 3-Kinase AKT

pathway in human cancer. Nat. Rev Cancer 2: 489-501.

95. Bai, D., Ueno, L. & Vogt, P. K. (2009) Akt-mediated regulation of NFkappaB and the

essentialness of NFkappaB for the oncogenicity of PI3K and Akt. Int. J. Cancer 125:

2863-2870.

96. Schley, P. D., Jijon, H. B., Robinson, L. E. & Field, C. J. (2005) Mechanisms of

omega-3 fatty acid-induced growth inhibition in MDA-MB-231 human breast cancer

cells. Breast Cancer Res. Treat. 92: 187-195.

Page 144: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

132

97. deGraffenried, L. A., Friedrichs, W. E., Fulcher, L., Fernandes, G., Silva, J. M.,

Peralba, J. M. & Hidalgo, M. (2003) Eicosapentaenoic acid restores tamoxifen sensitivity

in breast cancer cells with high Akt activity. Ann. Oncol. 14: 1051-1056.

98. Nicholson, K. M. & Anderson, N. G. (2002) The protein kinase B/Akt signalling

pathway in human malignancy. Cell Signal. 14: 381-395.

99. Hussain, A. R., Ahmed, S. O., Ahmed, M., Khan, O. S., Al, A. S., Platanias, L. C.,

Al-Kuraya, K. S. & Uddin, S. (2012) Cross-talk between NFkB and the PI3-kinase/AKT

pathway can be targeted in primary effusion lymphoma (PEL) cell lines for efficient

apoptosis. PLoS. One. 7: e39945.

100. Dolcet, X., Llobet, D., Pallares, J. & Matias-Guiu, X. (2005) NF-kB in development

and progression of human cancer. Virchows Arch. 446: 475-482.

101. Wu, J. T. & Kral, J. G. (2005) The NF-kappaB/IkappaB signaling system: a

molecular target in breast cancer therapy. J. Surg. Res. 123: 158-169.

102. Mayo, M. W., Wang, C. Y., Cogswell, P. C., Rogers-Graham, K. S., Lowe, S. W.,

Der, C. J. & Baldwin, A. S., Jr. (1997) Requirement of NF-kappaB activation to suppress

p53-independent apoptosis induced by oncogenic Ras. Science 278: 1812-1815.

103. Yerushalmi, R., Woods, R., Ravdin, P. M., Hayes, M. M. & Gelmon, K. A. (2010)

Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol. 11: 174-183.

104. Scholzen, T. & Gerdes, J. (2000) The Ki-67 protein: from the known and the

unknown. J. Cell Physiol 182: 311-322.

105. Saggar, J. K., Chen, J., Corey, P. & Thompson, L. U. (2010) Dietary flaxseed lignan

or oil combined with tamoxifen treatment affects MCF-7 tumor growth through estrogen

receptor- and growth factor-signaling pathways. Mol. Nutr. Food Res. 54: 415-425.

106. Taftachi, R., Ayhan, A., Ekici, S., Ergen, A. & Ozen, H. (2005) Proliferating-cell

nuclear antigen (PCNA) as an independent prognostic marker in patients after

prostatectomy: a comparison of PCNA and Ki-67. BJU. Int. 95: 650-654.

107. Paunesku, T., Mittal, S., Protic, M., Oryhon, J., Korolev, S. V., Joachimiak, A. &

Woloschak, G. E. (2001) Proliferating cell nuclear antigen (PCNA): ringmaster of the

genome. Int. J. Radiat. Biol. 77: 1007-1021.

108. Malkas, L. H., Herbert, B. S., bdel-Aziz, W., Dobrolecki, L. E., Liu, Y., Agarwal, B.,

Hoelz, D., Badve, S., Schnaper, L. et al. (2006) A cancer-associated PCNA expressed in

breast cancer has implications as a potential biomarker. Proc. Natl. Acad. Sci. U. S. A

103: 19472-19477.

109. Strzalka, W. & Ziemienowicz, A. (2011) Proliferating cell nuclear antigen (PCNA):

a key factor in DNA replication and cell cycle regulation. Ann. Bot. 107: 1127-1140.

Page 145: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

133

110. Olivo, S. E. & Hilakivi-Clarke, L. (2005) Opposing effects of prepubertal low- and

high-fat n-3 polyunsaturated fatty acid diets on rat mammary tumorigenesis.

Carcinogenesis 26: 1563-1572.

111. Kim, I. S. & Baek, S. H. (2010) Mouse models for breast cancer metastasis.

Biochem. Biophys. Res. Commun. 394: 443-447.

112. Karmali, R. A., Marsh, J. & Fuchs, C. (1984) Effect of omega-3 fatty acids on

growth of a rat mammary tumor. J. Natl. Cancer Inst. 73: 457-461.

113. Gonzalez, M. J., Schemmel, R. A., Gray, J. I., Dugan, L., Jr., Sheffield, L. G. &

Welsch, C. W. (1991) Effect of dietary fat on growth of MCF-7 and MDA-MB231

human breast carcinomas in athymic nude mice: relationship between carcinoma growth

and lipid peroxidation product levels. Carcinogenesis 12: 1231-1235.

114. Hubbard, N. E., Lim, D. & Erickson, K. L. (1998) Alteration of murine mammary

tumorigenesis by dietary enrichment with n-3 fatty acids in fish oil. Cancer Lett. 124: 1-7.

115. Yee, L. D., Young, D. C., Rosol, T. J., Vanbuskirk, A. M. & Clinton, S. K. (2005)

Dietary (n-3) polyunsaturated fatty acids inhibit HER-2/neu-induced breast cancer in

mice independently of the PPARgamma ligand rosiglitazone. J. Nutr. 135: 983-988.

116. MacLennan, M. B., Clarke, S. E., Perez, K., Wood, G. A., Muller, W. J., Kang, J. X.

& Ma, D. W. (2013) Mammary tumor development is directly inhibited by lifelong n-3

polyunsaturated fatty acids. J. Nutr. Biochem. 24: 388-395.

117. Dankort, D., Maslikowski, B., Warner, N., Kanno, N., Kim, H., Wang, Z., Moran, M.

F., Oshima, R. G., Cardiff, R. D. & Muller, W. J. (2001) Grb2 and Shc adapter proteins

play distinct roles in Neu (ErbB-2)-induced mammary tumorigenesis: implications for

human breast cancer. Mol. Cell Biol. 21: 1540-1551.

118. Yee, L. D., Lester, J. L., Cole, R. M., Richardson, J. R., Hsu, J. C., Li, Y., Lehman,

A., Belury, M. A. & Clinton, S. K. (2010) Omega-3 fatty acid supplements in women at

high risk of breast cancer have dose-dependent effects on breast adipose tissue fatty acid

composition. Am. J. Clin. Nutr. 91: 1185-1194.

119. Luijten, M., Verhoef, A., Dormans, J. A., Beems, R. B., Cremers, H. W.,

Nagelkerke, N. J., Adlercreutz, H., Penalvo, J. L. & Piersma, A. H. (2007) Modulation of

mammary tumor development in Tg.NK (MMTV/c-neu) mice by dietary fatty acids and

life stage-specific exposure to phytoestrogens. Reprod. Toxicol. 23: 407-413.

120. Yuri, T., Danbara, N., Tsujita-Kyutoku, M., Fukunaga, K., Takada, H., Inoue, Y.,

Hada, T. & Tsubura, A. (2003) Dietary docosahexaenoic acid suppresses N-methyl-N-

nitrosourea-induced mammary carcinogenesis in rats more effectively than

eicosapentaenoic acid. Nutr. Cancer 45: 211-217.

Page 146: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

134

121. Rose, D. P. & Connolly, J. M. (1990) Effects of fatty acids and inhibitors of

eicosanoid synthesis on the growth of a human breast cancer cell line in culture. Cancer

Res. 50: 7139-7144.

122. Grammatikos, S. I., Subbaiah, P. V., Victor, T. A. & Miller, W. M. (1994) n-3 and

n-6 fatty acid processing and growth effects in neoplastic and non-cancerous human

mammary epithelial cell lines. Br. J. Cancer 70: 219-227.

123. Kachhap, S. K., Dange, P. P., Santani, R. H., Sawant, S. S. & Ghosh, S. N. (2001)

Effect of omega-3 fatty acid (docosahexanoic acid) on BRCA1 gene expression and

growth in MCF-7 cell line. Cancer Biother. Radiopharm. 16: 257-263.

124. Chajes, V., Sattler, W., Stranzl, A. & Kostner, G. M. (1995) Influence of n-3 fatty

acids on the growth of human breast cancer cells in vitro: relationship to peroxides and

vitamin-E. Breast Cancer Res. Treat. 34: 199-212.

125. Rose, D. P., Connolly, J. M. & Coleman, M. (1996) Effect of omega-3 fatty acids on

the progression of metastases after the surgical excision of human breast cancer cell solid

tumors growing in nude mice. Clin. Cancer Res. 2: 1751-1756.

126. Chamras, H., Ardashian, A., Heber, D. & Glaspy, J. A. (2002) Fatty acid modulation

of MCF-7 human breast cancer cell proliferation, apoptosis and differentiation. J. Nutr.

Biochem. 13: 711-716.

127. Arterburn, L. M., Hall, E. B. & Oken, H. (2006) Distribution, interconversion, and

dose response of n-3 fatty acids in humans. Am. J. Clin. Nutr. 83: 1467S-1476S.

128. Harper, C. R., Edwards, M. J., DeFilippis, A. P. & Jacobson, T. A. (2006) Flaxseed

oil increases the plasma concentrations of cardioprotective (n-3) fatty acids in humans. J.

Nutr. 136: 83-87.

129. Pawlosky, R. J., Hibbeln, J. R., Novotny, J. A. & Salem, N., Jr. (2001) Physiological

compartmental analysis of alpha-linolenic acid metabolism in adult humans. J. Lipid Res.

42: 1257-1265.

130. Brenna, J. T. (2002) Efficiency of conversion of alpha-linolenic acid to long chain n-

3 fatty acids in man. Curr. Opin. Clin. Nutr. Metab Care 5: 127-132.

131. Ezaki, O., Takahashi, M., Shigematsu, T., Shimamura, K., Kimura, J., Ezaki, H. &

Gotoh, T. (1999) Long-term effects of dietary alpha-linolenic acid from perilla oil on

serum fatty acids composition and on the risk factors of coronary heart disease in

Japanese elderly subjects. J. Nutr. Sci. Vitaminol. (Tokyo) 45: 759-772.

132. Ghafoorunissa, Vani, A., Laxmi, R. & Sesikeran, B. (2002) Effects of dietary alpha-

linolenic acid from blended oils on biochemical indices of coronary heart disease in

Indians. Lipids 37: 1077-1086.

Page 147: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

135

133. Klein, V., Chajes, V., Germain, E., Schulgen, G., Pinault, M., Malvy, D., Lefrancq,

T., Fignon, A., Le, F. O. et al. (2000) Low alpha-linolenic acid content of adipose breast

tissue is associated with an increased risk of breast cancer. Eur. J. Cancer 36: 335-340.

134. Bougnoux, P., Koscielny, S., Chajes, V., Descamps, P., Couet, C. & Calais, G.

(1994) alpha-Linolenic acid content of adipose breast tissue: a host determinant of the

risk of early metastasis in breast cancer. Br. J. Cancer 70: 330-334.

135. Fritsche, K. L. & Johnston, P. V. (1990) Effect of dietary alpha-linolenic acid on

growth, metastasis, fatty acid profile and prostaglandin production of two murine

mammary adenocarcinomas. J. Nutr. 120: 1601-1609.

136. Hardman, W. E. (2007) Dietary canola oil suppressed growth of implanted MDA-

MB 231 human breast tumors in nude mice. Nutr. Cancer 57: 177-183.

137. Mabasa, L., Cho, K., Walters, M. W., Bae, S. & Park, C. S. (2013) Maternal dietary

canola oil suppresses growth of mammary carcinogenesis in female rat offspring. Nutr.

Cancer 65: 695-701.

138. Menendez, J. A., Vazquez-Martin, A., Ropero, S., Colomer, R. & Lupu, R. (2006)

HER2 (erbB-2)-targeted effects of the omega-3 polyunsaturated fatty acid, alpha-

linolenic acid (ALA; 18:3n-3), in breast cancer cells: the "fat features" of the

"Mediterranean diet" as an "anti-HER2 cocktail". Clin. Transl. Oncol. 8: 812-820.

139. Menendez, J. A., Ropero, S., Lupu, R. & Colomer, R. (2004) Dietary fatty acids

regulate the activation status of Her-2/neu (c-erbB-2) oncogene in breast cancer cells.

Ann. Oncol. 15: 1719-1721.

140. Mason, J. K., Fu, M. H., Chen, J., Yu, Z. & Thompson, L. U. (2013) Dietary

flaxseed-trastuzumab interactive effects on the growth of HER2-overexpressing human

breast tumors (BT-474). Nutr. Cancer 65: 451-459.

141. Chiu, L. C. & Wan, J. M. (1999) Induction of apoptosis in HL-60 cells by

eicosapentaenoic acid (EPA) is associated with downregulation of bcl-2 expression.

Cancer Lett. 145: 17-27.

142. Senzaki, H., Iwamoto, S., Ogura, E., Kiyozuka, Y., Arita, S., Kurebayashi, J.,

Takada, H., Hioki, K. & Tsubura, A. (1998) Dietary effects of fatty acids on growth and

metastasis of KPL-1 human breast cancer cells in vivo and in vitro. Anticancer Res. 18:

1621-1627.

143. Sauer, L. A., Dauchy, R. T., Blask, D. E., Krause, J. A., Davidson, L. K. & Dauchy,

E. M. (2005) Eicosapentaenoic acid suppresses cell proliferation in MCF-7 human breast

cancer xenografts in nude rats via a pertussis toxin-sensitive signal transduction pathway.

J. Nutr. 135: 2124-2129.

144. Chambrier, C., Bastard, J. P., Rieusset, J., Chevillotte, E., Bonnefont-Rousselot, D.,

Therond, P., Hainque, B., Riou, J. P., Laville, M. & Vidal, H. (2002) Eicosapentaenoic

Page 148: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

136

acid induces mRNA expression of peroxisome proliferator-activated receptor gamma.

Obes. Res. 10: 518-525.

145. Bougnoux, P., Hajjaji, N., Ferrasson, M. N., Giraudeau, B., Couet, C. & Le, F. O.

(2009) Improving outcome of chemotherapy of metastatic breast cancer by

docosahexaenoic acid: a phase II trial. Br. J. Cancer 101: 1978-1985.

146. Kang, K. S., Wang, P., Yamabe, N., Fukui, M., Jay, T. & Zhu, B. T. (2010)

Docosahexaenoic acid induces apoptosis in MCF-7 cells in vitro and in vivo via reactive

oxygen species formation and caspase 8 activation. PLoS. One. 5: e10296.

147. Blanckaert, V., Ulmann, L., Mimouni, V., Antol, J., Brancquart, L. & Chenais, B.

(2010) Docosahexaenoic acid intake decreases proliferation, increases apoptosis and

decreases the invasive potential of the human breast carcinoma cell line MDA-MB-231.

Int. J. Oncol. 36: 737-742.

148. Chiu, L. C., Wong, E. Y. & Ooi, V. E. (2004) Docosahexaenoic acid from a cultured

microalga inhibits cell growth and induces apoptosis by upregulating Bax/Bcl-2 ratio in

human breast carcinoma MCF-7 cells. Ann. N. Y. Acad. Sci. 1030: 361-368.

149. Tsujita-Kyutoku, M., Yuri, T., Danbara, N., Senzaki, H., Kiyozuka, Y., Uehara, N.,

Takada, H., Hada, T., Miyazawa, T. et al. (2004) Conjugated docosahexaenoic acid

suppresses KPL-1 human breast cancer cell growth in vitro and in vivo: potential

mechanisms of action. Breast Cancer Res. 6: R291-R299.

150. Connolly, J. M., Gilhooly, E. M. & Rose, D. P. (1999) Effects of reduced dietary

linoleic acid intake, alone or combined with an algal source of docosahexaenoic acid, on

MDA-MB-231 breast cancer cell growth and apoptosis in nude mice. Nutr. Cancer 35:

44-49.

151. Jourdan, M. L., Maheo, K., Barascu, A., Goupille, C., De Latour, M. P., Bougnoux,

P. & Rio, P. G. (2007) Increased BRCA1 protein in mammary tumours of rats fed marine

omega-3 fatty acids. Oncol. Rep. 17: 713-719.

152. Bernard-Gallon, D. J., Vissac-Sabatier, C., ntoine-Vincent, D., Rio, P. G., Maurizis,

J. C., Fustier, P. & Bignon, Y. J. (2002) Differential effects of n-3 and n-6

polyunsaturated fatty acids on BRCA1 and BRCA2 gene expression in breast cell lines.

Br. J. Nutr. 87: 281-289.

153. Rahman, M. M., Veigas, J. M., Williams, P. J. & Fernandes, G. (2013) DHA is a

more potent inhibitor of breast cancer metastasis to bone and related osteolysis than EPA.

Breast Cancer Res. Treat. 141: 341-352.

154. Bougnoux, P., Chajes, V., Germain, E., Hubert, B., Lhuillery, C., Le, F. O., Body, G.

& Calais, G. (1999) Cytotoxic drug efficacy correlates with adipose tissue

docosahexaenoic acid level in locally advanced breast carcinoma. Lipids 34 Suppl: S109.

Page 149: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

137

155. Flower, G., Fritz, H., Balneaves, L. G., Verma, S., Skidmore, B., Fernandes, R.,

Kennedy, D., Cooley, K., Wong, R. et al. (2013) Flax and Breast Cancer: A Systematic

Review. Integr. Cancer Ther. 13: 181-192.

156. Singh, K. K., Mridula, D., Rehal, J. & Barnwal, P. (2011) Flaxseed: a potential

source of food, feed and fiber. Crit Rev Food Sci. Nutr. 51: 210-222.

157. Patterson, R. E. (2011) Flaxseed and breast cancer: what should we tell our patients?

J. Clin. Oncol. 29: 3723-3724.

158. Kaul, N., Kreml, R., Austria, J. A., Richard, M. N., Edel, A. L., Dibrov, E., Hirono,

S., Zettler, M. E. & Pierce, G. N. (2008) A comparison of fish oil, flaxseed oil and

hempseed oil supplementation on selected parameters of cardiovascular health in healthy

volunteers. J. Am. Coll. Nutr. 27: 51-58.

159. Mason, J. K. & Thompson, L. U. (2014) Flaxseed and its lignan and oil components:

can they play a role in reducing the risk of and improving the treatment of breast cancer?

Appl. Physiol Nutr. Metab 39: 663-678.

160. Ganry, O. (2002) Phytoestrogen and breast cancer prevention. Eur. J. Cancer Prev.

11: 519-522.

161. Thompson, L. U., Boucher, B. A., Liu, Z., Cotterchio, M. & Kreiger, N. (2006)

Phytoestrogen content of foods consumed in Canada, including isoflavones, lignans, and

coumestan. Nutr. Cancer 54: 184-201.

162. Prasad, K. (2000) Flaxseed: a source of hypocholesterolemic and antiatherogenic

agents. Drug News Perspect. 13: 99-104.

163. Prasad, K. (2001) Secoisolariciresinol diglucoside from flaxseed delays the

development of type 2 diabetes in Zucker rat. J. Lab Clin. Med. 138: 32-39.

164. Liu, J. & Ma, D. W. (2014) The role of n-3 polyunsaturated fatty acids in the

prevention and treatment of breast cancer. Nutrients. 6: 5184-5223.

165. Francois, C. A., Connor, S. L., Bolewicz, L. C. & Connor, W. E. (2003)

Supplementing lactating women with flaxseed oil does not increase docosahexaenoic acid

in their milk. Am. J. Clin. Nutr. 77: 226-233.

166. Boon, H. S., Olatunde, F. & Zick, S. M. (2007) Trends in complementary/alternative

medicine use by breast cancer survivors: comparing survey data from 1998 and 2005.

BMC. Womens Health 7: 4.

167. Boucher, B. A., Cotterchio, M., Curca, I. A., Kreiger, N., Harris, S. A., Kirsh, V. A.

& Goodwin, P. J. (2012) Intake of phytoestrogen foods and supplements among women

recently diagnosed with breast cancer in Ontario, Canada. Nutr. Cancer 64: 695-703.

Page 150: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

138

168. Hilakivi-Clarke, L., Cho, E., deAssis, S., Olivo, S., Ealley, E., Bouker, K. B., Welch,

J. N., Khan, G., Clarke, R. & Cabanes, A. (2001) Maternal and prepubertal diet,

mammary development and breast cancer risk. J. Nutr. 131: 154S-157S.

169. Hilakivi-Clarke, L. (2007) Nutritional modulation of terminal end buds: its

relevance to breast cancer prevention. Curr. Cancer Drug Targets. 7: 465-474.

170. Russo, I. H. & Russo, J. (1998) Role of hormones in mammary cancer initiation and

progression. J. Mammary. Gland. Biol. Neoplasia. 3: 49-61.

171. Russo, J. & Russo, I. H. (1978) DNA labeling index and structure of the rat

mammary gland as determinants of its susceptibility to carcinogenesis. J. Natl. Cancer

Inst. 61: 1451-1459.

172. Russo, J. & Russo, I. H. (2008) Breast development, hormones and cancer. Adv.

Exp. Med. Biol. 630: 52-56.

173. Tou, J. C. & Thompson, L. U. (1999) Exposure to flaxseed or its lignan component

during different developmental stages influences rat mammary gland structures.

Carcinogenesis 20: 1831-1835.

174. Ward, W. E., Jiang, F. O. & Thompson, L. U. (2000) Exposure to flaxseed or

purified lignan during lactation influences rat mammary gland structures. Nutr. Cancer 37:

187-192.

175. Tan, K. P., Chen, J., Ward, W. E. & Thompson, L. U. (2004) Mammary gland

morphogenesis is enhanced by exposure to flaxseed or its major lignan during suckling in

rats. Exp. Biol. Med. (Maywood. ) 229: 147-157.

176. Chen, J., Tan, K. P., Ward, W. E. & Thompson, L. U. (2003) Exposure to flaxseed

or its purified lignan during suckling inhibits chemically induced rat mammary

tumorigenesis. Exp. Biol. Med. (Maywood. ) 228: 951-958.

177. Khan, G., Penttinen, P., Cabanes, A., Foxworth, A., Chezek, A., Mastropole, K., Yu,

B., Smeds, A., Halttunen, T. et al. (2007) Maternal flaxseed diet during pregnancy or

lactation increases female rat offspring's susceptibility to carcinogen-induced mammary

tumorigenesis. Reprod. Toxicol. 23: 397-406.

178. Yu, B., Khan, G., Foxworth, A., Huang, K. & Hilakivi-Clarke, L. (2006) Maternal

dietary exposure to fiber during pregnancy and mammary tumorigenesis among rat

offspring. Int. J. Cancer 119: 2279-2286.

179. Callegaro, M. G., Milbradt, B. G., Alves, E., Diettrich, T., Kemerich, D. M., Hausen,

B. S., Duarte, F. A., Flores, E. M., Dressler, V. L. & Emanuelli, T. (2011) Effect of wheat

bran and flaxseed on cadmium effects and retention in rats. Hum. Exp. Toxicol. 30: 981-

991.

Page 151: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

139

180. Serraino, M. & Thompson, L. U. (1991) The effect of flaxseed supplementation on

early risk markers for mammary carcinogenesis. Cancer Lett. 60: 135-142.

181. Serraino, M. & Thompson, L. U. (1992) The effect of flaxseed supplementation on

the initiation and promotional stages of mammary tumorigenesis. Nutr. Cancer 17: 153-

159.

182. Thompson, L. U., Rickard, S. E., Orcheson, L. J. & Seidl, M. M. (1996) Flaxseed

and its lignan and oil components reduce mammary tumor growth at a late stage of

carcinogenesis. Carcinogenesis 17: 1373-1376.

183. Rickard, S. E., Yuan, Y. V., Chen, J. & Thompson, L. U. (1999) Dose effects of

flaxseed and its lignan on N-methyl-N-nitrosourea-induced mammary tumorigenesis in

rats. Nutr. Cancer 35: 50-57.

184. Chen, J., Stavro, P. M. & Thompson, L. U. (2002) Dietary flaxseed inhibits human

breast cancer growth and metastasis and downregulates expression of insulin-like growth

factor and epidermal growth factor receptor. Nutr. Cancer 43: 187-192.

185. Saarinen, N. M., Huovinen, R., Warri, A., Makela, S. I., Valentin-Blasini, L.,

Sjoholm, R., Ammala, J., Lehtila, R., Eckerman, C. et al. (2002) Enterolactone inhibits

the growth of 7,12-dimethylbenz(a)anthracene-induced mammary carcinomas in the rat.

Mol. Cancer Ther. 1: 869-876.

186. Power, K. A., Chen, J. M., Saarinen, N. M. & Thompson, L. U. (2008) Changes in

biomarkers of estrogen receptor and growth factor signaling pathways in MCF-7 tumors

after short- and long-term treatment with soy and flaxseed. J. Steroid Biochem. Mol. Biol.

112: 13-19.

187. Lee, S. H., Lee, J. K., Jin, S. M., Lee, K. C., Sohn, J. H., Chae, S. W. & Kim, D. H.

(2010) Expression of cell-cycle regulators (cyclin D1, cyclin E, p27kip1, p57kip2) in

papillary thyroid carcinoma. Otolaryngol. Head Neck Surg. 142: 332-337.

188. Chen, J., Power, K. A., Mann, J., Cheng, A. & Thompson, L. U. (2007) Flaxseed

alone or in combination with tamoxifen inhibits MCF-7 breast tumor growth in

ovariectomized athymic mice with high circulating levels of estrogen. Exp. Biol. Med.

(Maywood. ) 232: 1071-1080.

189. Chen, J., Power, K. A., Mann, J., Cheng, A. & Thompson, L. U. (2007) Dietary

flaxseed interaction with tamoxifen induced tumor regression in athymic mice with

MCF-7 xenografts by downregulating the expression of estrogen related gene products

and signal transduction pathways. Nutr. Cancer 58: 162-170.

190. Birkved, F. K., Mortensen, A., Penalvo, J. L., Lindecrona, R. H. & Sorensen, I. K.

(2011) Investigation into the cancer protective effect of flaxseed in Tg.NK (MMTV/c-neu)

mice, a murine mammary tumor model. Genes Nutr. 6: 403-411.

Page 152: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

140

191. Lee, J. & Cho, K. (2012) Flaxseed sprouts induce apoptosis and inhibit growth in

MCF-7 and MDA-MB-231 human breast cancer cells. In Vitro Cell Dev. Biol. Anim 48:

244-250.

192. Christofidou-Solomidou, M., Pietrofesa, R., Arguiri, E., McAlexander, M. A. &

Witwer, K. W. (2014) Dietary flaxseed modulates the miRNA profile in irradiated and

non-irradiated murine lungs: a novel mechanism of tissue radioprotection by flaxseed.

Cancer Biol. Ther. 15: 930-937.

193. Cotterchio, M., Boucher, B. A., Kreiger, N., Mills, C. A. & Thompson, L. U. (2008)

Dietary phytoestrogen intake--lignans and isoflavones--and breast cancer risk (Canada).

Cancer Causes Control 19: 259-272.

194. Lowcock, E. C., Cotterchio, M. & Boucher, B. A. (2013) Consumption of flaxseed,

a rich source of lignans, is associated with reduced breast cancer risk. Cancer Causes

Control 24: 813-816.

195. Buck, K., Zaineddin, A. K., Vrieling, A., Heinz, J., Linseisen, J., Flesch-Janys, D. &

Chang-Claude, J. (2011) Estimated enterolignans, lignan-rich foods, and fibre in relation

to survival after postmenopausal breast cancer. Br. J. Cancer 105: 1151-1157.

196. Hutchins, A. M., Martini, M. C., Olson, B. A., Thomas, W. & Slavin, J. L. (2001)

Flaxseed consumption influences endogenous hormone concentrations in postmenopausal

women. Nutr. Cancer 39: 58-65.

197. Haggans, C. J., Hutchins, A. M., Olson, B. A., Thomas, W., Martini, M. C. & Slavin,

J. L. (1999) Effect of flaxseed consumption on urinary estrogen metabolites in

postmenopausal women. Nutr. Cancer 33: 188-195.

198. Brooks, J. D., Ward, W. E., Lewis, J. E., Hilditch, J., Nickell, L., Wong, E. &

Thompson, L. U. (2004) Supplementation with flaxseed alters estrogen metabolism in

postmenopausal women to a greater extent than does supplementation with an equal

amount of soy. Am. J. Clin. Nutr. 79: 318-325.

199. Schneider, J., Kinne, D., Fracchia, A., Pierce, V., Anderson, K. E., Bradlow, H. L. &

Fishman, J. (1982) Abnormal oxidative metabolism of estradiol in women with breast

cancer. Proc. Natl. Acad. Sci. U. S. A 79: 3047-3051.

200. Luijten, M., Singh, A. V., Bastian, C. A., Westerman, A., Pisano, M. M., Pennings, J.

L., Verhoef, A., Green, M. L., Piersma, A. H. et al. (2013) Lasting effects on body weight

and mammary gland gene expression in female mice upon early life exposure to n-3 but

not n-6 high-fat diets. PLoS. One. 8: e55603.

201. Cameron, E. R. B. J. M. R. (1989) DIvergent effects of omega-6 and omega-3 fatty

acids on mammary tumor development in C3H/Heston mice treated with DMBA.

Nutrition Research 9: 383-393.

Page 153: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

141

202. Wang, L., Chen, J. & Thompson, L. U. (2005) The inhibitory effect of flaxseed on

the growth and metastasis of estrogen receptor negative human breast cancer xenograftsis

attributed to both its lignan and oil components. Int. J. Cancer 116: 793-798.

203. Chen, J., Wang, L. & Thompson, L. U. (2006) Flaxseed and its components reduce

metastasis after surgical excision of solid human breast tumor in nude mice. Cancer Lett.

234: 168-175.

204. Bergman, J. M., Thompson, L. U. & Dabrosin, C. (2007) Flaxseed and its lignans

inhibit estradiol-induced growth, angiogenesis, and secretion of vascular endothelial

growth factor in human breast cancer xenografts in vivo. Clin. Cancer Res. 13: 1061-

1067.

205. Chen, J., Saggar, J. K., Corey, P. & Thompson, L. U. (2011) Flaxseed cotyledon

fraction reduces tumour growth and sensitises tamoxifen treatment of human breast

cancer xenograft (MCF-7) in athymic mice. Br. J. Nutr. 105: 339-347.

206. Chen, J., Saggar, J. K., Ward, W. E. & Thompson, L. U. (2011) Effects of flaxseed

lignan and oil on bone health of breast-tumor-bearing mice treated with or without

tamoxifen. J. Toxicol. Environ. Health A 74: 757-768.

207. Mason, J. K., Chen, J. & Thompson, L. U. (2010) Flaxseed oil-trastuzumab

interaction in breast cancer. Food Chem. Toxicol. 48: 2223-2226.

208. Rao, G. N., Ney, E. & Herbert, R. A. (2000) Effect of melatonin and linolenic acid

on mammary cancer in transgenic mice with c-neu breast cancer oncogene. Breast Cancer

Res. Treat. 64: 287-296.

209. Wiggins, A. K., Mason, J. K. & Thompson, L. U. (2015) Growth and gene

expression differ over time in alpha-linolenic acid treated breast cancer cells. Exp. Cell

Res. 333: 147-154.

210. Chen, J., Saggar, J. K., Corey, P. & Thompson, L. U. (2009) Flaxseed and pure

secoisolariciresinol diglucoside, but not flaxseed hull, reduce human breast tumor growth

(MCF-7) in athymic mice. J. Nutr. 139: 2061-2066.

211. Canadian Cancer Statistics 2014. Canadian Cancer Society's Steering Committee on

Cancer Statistics. Canadian Cancer Society . 2015.

Ref Type: Electronic Citation

212. Taneja, P., Frazier, D. P., Kendig, R. D., Maglic, D., Sugiyama, T., Kai, F., Taneja,

N. K. & Inoue, K. (2009) MMTV mouse models and the diagnostic values of MMTV-

like sequences in human breast cancer. Expert. Rev Mol. Diagn. 9: 423-440.

213. Muller, W. J., Sinn, E., Pattengale, P. K., Wallace, R. & Leder, P. (1988) Single-step

induction of mammary adenocarcinoma in transgenic mice bearing the activated c-neu

oncogene. Cell 54: 105-115.

Page 154: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

142

214. Siegel, P. M., Ryan, E. D., Cardiff, R. D. & Muller, W. J. (1999) Elevated

expression of activated forms of Neu/ErbB-2 and ErbB-3 are involved in the induction of

mammary tumors in transgenic mice: implications for human breast cancer. EMBO J. 18:

2149-2164.

215. Kroese, M., Zimmern, R. L. & Pinder, S. E. (2007) HER2 status in breast cancer--an

example of pharmacogenetic testing. J. R. Soc. Med. 100: 326-329.

216. Slamon, D. J., Clark, G. M., Wong, S. G., Levin, W. J., Ullrich, A. & McGuire, W.

L. (1987) Human breast cancer: correlation of relapse and survival with amplification of

the HER-2/neu oncogene. Science 235: 177-182.

217. Bargmann, C. I., Hung, M. C. & Weinberg, R. A. (1986) The neu oncogene encodes

an epidermal growth factor receptor-related protein. Nature 319: 226-230.

218. Slamon, D. J. (1990) Studies of the HER-2/neu proto-oncogene in human breast

cancer. Cancer Invest 8: 253.

219. Hilakivi-Clarke, L., Clarke, R., Onojafe, I., Raygada, M., Cho, E. & Lippman, M.

(1997) A maternal diet high in n - 6 polyunsaturated fats alters mammary gland

development, puberty onset, and breast cancer risk among female rat offspring. Proc. Natl.

Acad. Sci. U. S. A 94: 9372-9377.

220. Anderson, B. M., MacLennan, M. B., Hillyer, L. M. & Ma, D. W. (2014) Lifelong

exposure to n-3 PUFA affects pubertal mammary gland development. Appl. Physiol Nutr.

Metab 39: 699-706.

221. Russo, J., Balogh, G. A., Heulings, R., Mailo, D. A., Moral, R., Russo, P. A., Sheriff,

F., Vanegas, J. & Russo, I. H. (2006) Molecular basis of pregnancy-induced breast cancer

protection. Eur. J. Cancer Prev. 15: 306-342.

222. Russo, J. & Russo, I. H. (1999) Cellular basis of breast cancer susceptibility. Oncol.

Res. 11: 169-178.

223. Russo, J., Gusterson, B. A., Rogers, A. E., Russo, I. H., Wellings, S. R. & van

Zwieten, M. J. (1990) Comparative study of human and rat mammary tumorigenesis. Lab

Invest 62: 244-278.

224. Cardiff, R. D. & Wellings, S. R. (1999) The comparative pathology of human and

mouse mammary glands. J. Mammary. Gland. Biol. Neoplasia. 4: 105-122.

225. Dankort, D., Jeyabalan, N., Jones, N., Dumont, D. J. & Muller, W. J. (2001)

Multiple ErbB-2/Neu Phosphorylation Sites Mediate Transformation through Distinct

Effector Proteins. J. Biol. Chem. 276: 38921-38928.

226. Folch, J., LEES, M. & SLOANE STANLEY, G. H. (1957) A simple method for the

isolation and purification of total lipides from animal tissues. J. Biol. Chem. 226: 497-

509.

Page 155: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

143

227. McIlwain, D. R., Berger, T. & Mak, T. W. (2015) Caspase functions in cell death

and disease. Cold Spring Harb. Perspect. Biol. 7.

228. Kamada, S., Kikkawa, U., Tsujimoto, Y. & Hunter, T. (2005) Nuclear translocation

of caspase-3 is dependent on its proteolytic activation and recognition of a substrate-like

protein(s). J. Biol. Chem. 280: 857-860.

229. Bodicoat, D. H., Schoemaker, M. J., Jones, M. E., McFadden, E., Griffin, J.,

Ashworth, A. & Swerdlow, A. J. (2014) Timing of pubertal stages and breast cancer risk:

the Breakthrough Generations Study. Breast Cancer Res. 16: R18.

230. Kelsey, J. L., Gammon, M. D. & John, E. M. (1993) Reproductive factors and breast

cancer. Epidemiol. Rev 15: 36-47.

231. Hsieh, C. C., Trichopoulos, D., Katsouyanni, K. & Yuasa, S. (1990) Age at

menarche, age at menopause, height and obesity as risk factors for breast cancer:

associations and interactions in an international case-control study. Int. J. Cancer 46: 796-

800.

232. Hilakivi-Clarke, L., Cho, E., Cabanes, A., deAssis, S., Olivo, S., Helferich, W.,

Lippman, M. E. & Clarke, R. (2002) Dietary modulation of pregnancy estrogen levels

and breast cancer risk among female rat offspring. Clin. Cancer Res. 8: 3601-3610.

233. MacLennan, M. B., Anderson, B. M. & Ma, D. W. (2011) Differential mammary

gland development in FVB and C57Bl/6 mice: implications for breast cancer research.

Nutrients. 3: 929-936.

234. Kang, J. X., Wang, J., Wu, L. & Kang, Z. B. (2004) Transgenic mice: fat-1 mice

convert n-6 to n-3 fatty acids. Nature 427: 504.

235. Turk, H. F. & Chapkin, R. S. (2013) Membrane lipid raft organization is uniquely

modified by n-3 polyunsaturated fatty acids. Prostaglandins Leukot. Essent. Fatty Acids

88: 43-47.

236. Ackman, R. G. (1992) The absorption of fish oils and concentrates. Lipids 27: 858-

862.

237. Sadou, H., Leger, C. L., Descomps, B., Barjon, J. N., Monnier, L. & Crastes de, P. A.

(1995) Differential incorporation of fish-oil eicosapentaenoate and docosahexaenoate into

lipids of lipoprotein fractions as related to their glyceryl esterification: a short-term

(postprandial) and long-term study in healthy humans. Am. J. Clin. Nutr. 62: 1193-1200.

238. Tam, O. & Innis, S. M. (2006) Dietary polyunsaturated fatty acids in gestation alter

fetal cortical phospholipids, fatty acids and phosphatidylserine synthesis. Dev. Neurosci.

28: 222-229.

239. Sastry, P. S. (1985) Lipids of nervous tissue: composition and metabolism. Prog.

Lipid Res. 24: 69-176.

Page 156: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

144

240. Tacconi, M. & Wurtman, R. J. (1985) Rat brain phosphatidyl-N,N-

dimethylethanolamine is rich in polyunsaturated fatty acids. J. Neurochem. 45: 805-809.

241. He, C., Qu, X., Wan, J., Rong, R., Huang, L., Cai, C., Zhou, K., Gu, Y., Qian, S. Y.

& Kang, J. X. (2012) Inhibiting delta-6 desaturase activity suppresses tumor growth in

mice. PLoS. One. 7: e47567.

242. Reese, D. M. & Slamon, D. J. (1997) HER-2/neu signal transduction in human

breast and ovarian cancer. Stem Cells 15: 1-8.

243. Mason, J. K., Fu, M. H., Chen, J., Yu, Z. & Thompson, L. U. (2013) Dietary

flaxseed-trastuzumab interactive effects on the growth of HER2-overexpressing human

breast tumors (BT-474). Nutr. Cancer 65: 451-459.

244. Riedl, S. J. & Shi, Y. (2004) Molecular mechanisms of caspase regulation during

apoptosis. Nat. Rev Mol. Cell Biol. 5: 897-907.

245. Kim, W., McMurray, D. N. & Chapkin, R. S. (2010) n-3 polyunsaturated fatty acids-

-physiological relevance of dose. Prostaglandins Leukot. Essent. Fatty Acids 82: 155-158.

246. Straka, S., Lester, J. L., Cole, R. M., Andridge, R. R., Puchala, S., Rose, A. M.,

Clinton, S. K., Belury, M. A. & Yee, L. D. (2015) Incorporation of eicosapentaenioic and

docosahexaenoic acids into breast adipose tissue of women at high risk of breast cancer:

A randomized clinical trial of dietary fish and n-3 fatty acid capsules. Mol. Nutr. Food

Res.

Page 157: Plant- and Marine-derived n-3 Polyunsaturated Fatty Acids ...

145

Appendix - MG fatty acid composition (PC fraction)

Upper panel: PC fraction in 6-week-old mice; lower panel: PC fraction in 20-week-old

mice. The fatty acid composition of MG phospholipid differs markedly between dietary

treatments (n=4-6 per diet). Bars not sharing a letter are significantly different (p<0.05)

based on a one-way ANOVA test. Full fatty acid profile is displayed in Table 4.4.