You are on page 1of 76

Anti-Carcinogenic Effects of Morinda citrifolia (Noni): Identifying Signal Transduction

Pathways using ER Positive and ER Negative Human Breast Cancer Cell Lines

A thesis submitted to the Graduate Division of the University of Hawai’i at Mānoa in

partial fulfillment of the requirements for the degree of

Master of Science

in

Molecular Biosciences and Bioengineering

August 2012

By

Phoebe Hwang

Thesis Committee:

Pratibha V. Nerurkar – Chairperson

Dulal Borthakur

Scot Nelson

i
Student: Phoebe W.N. Hwang
Student ID#: 6096491
Degree: MS
Field: Molecular Biosciences Bioengineering
Graduation date: May 2012

Title: Anti-carcinogenic effects of Morindacitrifolia(noni): Identifying signal


transduction pathways using ER positive and ER negative human breast cancer cell
lines

We certify that we have read this Thesis and that, in our opinion, it is satisfactory in
scope and quality as a Thesis for the degree of Master of Science in Molecular
Bioscience Bioengineering.

Thesis Committee:

Names Signatures

PratibhaNerurkar, Chairperson ____________________________

DulalBorthakur ____________________________

Scot Nelson ____________________________

ii
Dedication

To my brother, Andy Hwang.

iii
Acknowledgments

Thanks to Dr. Pratibha Nerurkar for all her support and guidance. I would never be able

to complete my project if she had not accepted me into her lab and taught me everything.

I would like to express my gratitude to Dr. Dulal Borthakur and Dr. Scot Nelson for their

counsel and guidance.

I would also like to thank our lab members for all their moral support. Especially my

colleagues who trained me.

Last but not least, thanks to my friends and family for all the emotional support and

understanding.

iv
Abstract

It is predicted that one in every eight women in the United States will be diagnosed with

breast cancer in her lifetime, making breast cancer the second most prevalent cancer in

the nation. Native Hawaiians have the highest incidence as well as mortality rates

compared to other ethnic groups in Hawaii. An increase in the use of alternative

medicine, specifically by breast cancer patients prompted us to study the anti-cancer

effects of Hawaiian traditional medicine, Morinda citritfolia (noni). Estrogen receptor

positive (ER +ve) cells, MCF-7 and estrogen receptor negative (ER –ve) cells, MDAMB-

231 were initially treated with fNJ at varying concentrations for up to 96h. 10% fNJ

demonstrated cell death in 30 – 40% of the cells and 15% fNJ demonstrated cell death in

50 – 60% of the cells. In addition, when non-carcinoma breast cells, MCF-10A

underwent the same treatment, 15% fNJ only demonstrated 10 – 15% cell death in MCF-

10A. Cell proliferation assays suggest that fNJ is toxic to breast cancer cell lines, MCF-7

and MDAMB-231 but not normal breast cell line, MCF-10A. Apoptosis array data have

shown a significant decrease of a class of proteins called inhibitors of apoptosis proteins

(IAPs) such as survivin, Bcl-2, cIAP-1, and cIAP-2 in fNJ treated MCF-7 and MDAMB-

231 cells. There was also a significant decrease in proteins that assist with cell

replication such as claspin and phospho-Rad17. This demonstrates that fNJ induces cell

selective apoptosis and inhibits cell proliferation in breast cancer cell lines, MCF-7 and

MDAMB-231. [Grants: NCCAM (R21AT003719), USDA-CREES (2004-34135-

15182)]

v
Table of contents

Dedication .......................................................................................................................... iii

Acknowledgments.............................................................................................................. iv

Abstract ............................................................................................................................... v

Table of contents ................................................................................................................ vi

List of figures ................................................................................................................... viii

List of Tables ...................................................................................................................... x

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

Chapter 1: Introduction ....................................................................................................... 1

1.1 Breast Cancer ........................................................................................................... 1


1.1.1 Impact ............................................................................................................. 1
1.1.2 Factors and Causations ................................................................................... 1
1.1.3 Estrogen Receptor Negative vs. Estrogen Receptor Positive Breast Cancer
Cells ......................................................................................................................... 5

1.2 Complementary and Alternative Medicine .............................................................. 7


1.2.1 Usage in the United States ............................................................................. 7
1.2.2 Complementary and Alternative Medicine as Cancer Treatment ................... 8

1.3 Morinda citrofolia (Noni) ........................................................................................ 9


1.3.1 History............................................................................................................. 9
1.3.2 Traditional and Modern Uses........................................................................ 10
1.3.3 Fermented Noni Fruit Juice .......................................................................... 13

1.4 Noni and Cancer .................................................................................................. 14


1.4.1 In vitro studies.............................................................................................. 14
1.4.2 In vivo studies ............................................................................................... 15
1.4.3 Molecular Mechanisms ................................................................................. 19

Chapter 2: Hypothesis and Aims ...................................................................................... 21


2.1 Hypothesis............................................................................................................ 21
2.2 Specific Aims ....................................................................................................... 22

vi
Chapter 3: Materials and Methods .................................................................................... 24
3.1 Preparation of fNJ with endotoxins removed ........................................................ 24
3.2 Cell Culture ............................................................................................................ 24
3.3 MTT assay ............................................................................................................. 25
3.4 Human apoptosis antibody array ........................................................................... 25

Chapter 4: Results ............................................................................................................. 28


4.1 fNJ decreases cell viability in breast carcinoma cells ............................................ 28
4.2 Apoptotic proteins identified in fNJ treated breast carcinoma cells ...................... 29
4.3 fNJ inhibits survivin in MCF-7 (ER+ve) cells ....................................................... 29
4.4 fNJ up-regulates cytosolic levels of cytochrome c in MCF-7 (ER+ve) cells ........ 30
4.5 fNJ does not affect cytosolic and nuclear levels of nFκB in MCF-7 (ER+ve) cells30
4.6 fNJ inhibits survivin in MDAMB-231 (ER-ve) cells ............................................ 30
4.7 fNJ does not affect cytosolic levels of cytochrome c in MDAMB-231 (ER-ve)
cells ............................................................................................................................... 31
4.8 fNJ does not affect cytosolic and nuclear levels of nFκB in MDAM-231 (ER-ve)
cells ............................................................................................................................... 31
4.9 Figures of results .................................................................................................... 32

Chapter 5: Discussion ....................................................................................................... 46

Chapter 6: Conclusion....................................................................................................... 53
6.1 Significance............................................................................................................ 53
6.2 Future studies ......................................................................................................... 54

Reference: ......................................................................................................................... 55

vii
List of figures

Figure Page

Figure 1. Ductal and Lobular Carcinoma In Situ ................................................................ 2

Figure 2. Modifiable risk factors that affect cancer ............................................................ 4

Figure 3. General transcription mechanisms of estrogen receptor positive and estrogen


receptor negative breast cancer cells........................................................................... 6

Figure 4. Total out-of-pocket complementary and alternative medicine costs among


American adults in 2007 ............................................................................................. 8

Figure 5. Noni fruit ............................................................................................................. 9

Figure 6. Encapsulated freeze-dried noni fruit ................................................................. 13

Figure 7. fNJ induces apoptosis through hypothesized pathways .................................... 21

Figure 8. Phase contrast photomicrographs of MCF-7 cells treated with fNJ .................. 32

Figure 9. Phase contrast photomicrographs of MDAMB-231 cells treated with fNJ) ..... 32

Figure 10. Effects of fNJ on MCF7 cell viability ............................................................. 33

Figure 11. Effects of fNJ on MDAMB-231 cell viability................................................. 34

Figure 12. Effects of fNJ on MCF-10A cell viability ....................................................... 35

Figure 13. Human apoptosis array overlay ....................................................................... 36

Figure 14. Effects of 15% fNJ (v/v) on apoptotic proteins in MCF-7 cells...................... 37

Figure 15. Effects of 15% fNJ (v/v) on apoptotic proteins in MDAMB-231 cells. ......... 37

Figure 16. fNJ reduced survivin levels in MCF-7 cells .................................................... 38

Figure 17. fNJ increases cytosolic cytochrome c in MCF-7 cells .................................... 39

Figure 18. fNJ did not affect changes in cytosolic nFkB levels in MCF-7 cells .............. 40

Figure 19. fNJ did not affect changes in nuclear nFkB in MCF-7 cells ........................... 41

viii
Figure 20. fNJ reduced survivin levels MDAMB231 ....................................................... 42

Figure 21. fNJ did not affect cytosolic levels of cytochrome c in MDAMB-231 cells .... 43

Figure 22. fNJ does not affect cytosolic nFkB in MDAMB-231 cells ............................. 44

Figure 23. fNJ did not affect nuclear nFkB levels in MDAMB-231 cells ........................ 45

Figure 24. Extrinsic and intrinsic pathways of apoptosis ................................................. 47

Figure 25. Roles of Bcl-2 .................................................................................................. 50

Figure 26.The role of NF-kB in apoptosis ........................................................................ 52

ix
List of Tables

Table 1. Traditional uses of the noni ................................................................................ 10

Table 2. Modern uses of noni .......................................................................................... 12

Table 3.In vitro studies on the effects of noni and cancer................................................ 17

Table 4. Summary of molecular mechanisms involved in the effects of noni against


cancer ........................................................................................................................ 20

x
List of Abbreviations

AP1 Activator protein 1

ATM Ataxia telangiecstasia mutated

Bcl-2 B-cell lymphoma 2

BMI Body mass index

BRCA1 Breast cancer type 1

CAM Complementary and alternative medicine

CDH1 Cadherin 1

CHEK2 Dysfunctional check point homolog 2

cIAP-1 Cellular inhibitor of apoptosis protein 1

DCIS Ductal carcinoma in situ

EGF Epidermal growth factor

eIF-2α Eukaryotic translation initiation factor-2α

ER Estrogen receptors

ER-ve Estrogen receptor negative

ER+ve Estrogen receptor positive

Fas/TNFRSF6 Fibroblast associated tumor necrosis factor receptor


super family member 6

fNJ Ferment noni fruit juice

HepG2 Human laryngeal carcinoma

HSP-60 Heat shock protein 60

IAP Inhibitors of apoptosis

IFN-γ Interferon-γ

xi
IL-1β Interleukin-1β

JNK-1 c-Jun N-terminal kinase-1

LAN5 Human neuroblastoma

LCIS Lobular carcinoma in situ

LLC Lewis lung carcinoma

MCF-7 Human breast ER+ve carcinoma

NAG-1 Nonsteroidal anti-inflammatory activated gene-1

NfκB Necrosis factor-kappa beta

NJ Noni fruit juice

NK Natural killer

NO Nitric oxide

Noni-ppt Noni fruit precipitate

phospho-Rad17 Phosphorylated Rad 17

PK-B Protein kinase B

TNF Tumor necrosis factor

TP53 Tumor protein 53

TNFR Tumor necrosis factor receptor

TPA 12-O-tedtradecanoylphorbol-13-acetate

TRAIL TNF related apoptosis-inducing ligand

xii
Chapter 1: Introduction

1.1 Breast Cancer

1.1.1 Impact
Breast cancer is the third highest cause of cancer-related mortality in Hawaii [1].

Coming in behind prostate cancer, it is the second most prevalent cancer in the United

States [2]. An estimated number of 226,870 women in the United States will be

diagnosed with breast cancer by the end of 2012. Of these women, 1,120 will be from

Hawaii [1]. Although breast cancer mortality seems to remain relatively constant for the

past 30 years, the incidence rates have been slowly increasing from 1975 – 2008 [3].

1.1.2 Factors and Causations


Cancer is characterized as uncontrolled growth of damaged or abnormal cells in the

body. Unlike normal cells that proliferate and die and under apoptosis, these malignant

(cancerous) cells do not undergo cell cycle arrest and will continue to rigorously grow in

its abnormal state (www.cancer.org). Sometimes the cancer metastasizes and travels to

distant organs. The cancer is named after the body part where the tumor originates [3].

Thus, breast cancer originates from the breast tissue. Some breast cancers are formed

within the ducts or lobules of breasts and are therefore referred to as ductal carcinoma in

situ (DCIS) or lobular carcinoma in situ (LCIS), respectively (figure 1). Most breast

cancers are invasive and travel from ducts and lobules to nearby breast tissues [3]. Many

1
factors including age, sex, family history, physical fitness, and diet influence the type of

breast.

Figure 1. Ductal and Lobular Carcinoma In Situ [source: www.virtualmedicinecentre.com]

Aside from being female, age is the biggest risk factor for breast cancer. Invasive

breast cancer is found in 1 out of 8 women under the age of 45, whereas it is found in 2

out of 3 women over the age of 55 [4]. A woman at the age of 30 has a 0.43% risk of

developing breast cancer, compared to a woman at the age of60 who has a 3.45% risk of

developing breast cancer [3]. This evidence provided by the American Cancer Society

suggests that the probability of developing breast cancer increases with age. It is

believed that the increased likelihood of developing breast cancer at an older age may be

due to a longer life expectancy inmodern times. Other factors such as menopausal

hormone use, changes in reproductive patterns, rising obesity rates, and improved

detection and screening methods, also contribute to this increase.

Family history and genetics influence a woman’s likelihood of developing breast

cancer. Studies have shown that 5 – 10% of diagnosed breast cancer is thought to be

2
hereditary [4, 5]. The more closely related a woman is to a family member who has

breast cancer, the greater her chance of developing breast cancer [6]. Studies have shown

that a number of individuals with genes such as breast cancer type 1 (BRCA1) and

BRCA2 [7]and dysfunctional check point homolog 2 (CHEK2)[8], tumor protein 53

(TP53)[9], ataxia telangiecstasia mutated (ATM)[10], and cadherin 1 (CDH1)[11] genes

have a higher risk of developing breast cancer.

In light of current studies on lifestyle choices and cancer, obesity, physical fitness[12,

13], and nutrition have been implicated as risk factors (figure 2)[14, 15]. Garnet

Anderson’s study showed a 70% increase risk factor of breast cancer in obese

premenopausal women compared to normal weight premenopausal women [16]. This

suggests that women with higher than normal body mass index (BMI) have an increased

risk of developing breast cancer. Besides having a high BMI, women with higher than

normal central adiposity (measured by waist circumference) also have a higher risk of

developing breast cancer [17]. Obese individuals tend to have higher concentrations of

pro-inflammatory markers circulating in the blood causing the person to be in a chronic

inflammatory state [18-20]. An activated pro-inflammatory marker, necrosis factor-

kappa beta (NF-κB), has been observed in mammary adipose tissues obtained from high

fat diet fed mice [21] and obese women with breast cancer [22]. These findings show

that breast tissues with high adipocyte density have elevated levels of NF-κB.

Furthermore, the correlation between adipocyte size, BMI, and inflamed mammary

glands supports the idea that obesity is associated with an increased risk of breast cancer

on a cellular level. In 2010, 35.8% of the women in the United States were considered

obese (www.cdc.org), indicating that 40.6 million American women have a high risk of

3
developing breast cancer.

Figure 2.Modifiable risk factors that affect cancer [source: Food, nutrition, physical activity and the
prevention of cancer: a global perspective. World Cancer Research Fund/American Institute for Cancer
Research, 2007: p. 30-46]

4
Whether or not an individual’s diet affects their risk of developing breast cancer is

still a controversial topic. A number of studies carried out to discover the correlation

between foods and the onset of breast cancer have yielded conflicting results[3, 15].

Foods such as bitter melon [23], curcumin[24], resveratrol [25], and other foods high in

antioxidants and fiber [26, 27] have been shown to decrease the onset of breast cancer.

Whereas high temperature cooked meats [28], red meats[29], and high fat foods [21]

have been shown to increase the risks. More research is needed to accurately determine

the effects of dietary consumption on breast cancer. Other factors such as periods and

amount of food consumption, family history, food-food and food-drug interactions might

also play an important role in the development of breast cancer and need to be further

analyzed.

1.1.3 Estrogen Receptor Negative vs. Estrogen Receptor Positive Breast Cancer Cells
Estrogen is a steroid hormone that is commonly found in women to regulate

pregnancy, bone density, menstrual cycle, liver function, breast and uterine cancer.

Estrogen receptors (ER) are molecules that bind to estrogen hormones and initiate a

number of signaling pathways [30]. The presence of intracellular ER indicates the cell’s

receptiveness to estrogen hormones (figure 3) [30, 31]. Consequently, elevated levels of

circulating estrogen increases the risk of developing breast cancer for individuals with

estrogen receptor positive (ER+ve) breast cells[32-34].

5
Figure 3. General transcription mechanisms of estrogen receptor positive and estrogen receptor negative
breast cancer cells [source: Food, nutrition, physical activity and the prevention of cancer: a global
perspective. World Cancer Research Fund/American Institute for Cancer Research, 2007: p. 30-46

Due to the lack of estrogen receptors, estrogen receptor negative (ER-ve) breast cells

are unresponsive to selective hormonal therapy compared to ER+ve breast cells [35].

Consequently, studies have shown that since the amount of ER present on each cell

varies, not all ER+ve breast cells respond well to hormonal treatment. As the amount of

ER decreases, so does the efficacy of the treatment [19, 36]. This raises a problem

because 15% of the breast cancer patients in the United States are ER-, leaving

approximately 450,000 women incurable [37]. Unfortunately, only 30% of breast cancer

patients have overexpressing ER that is highly susceptible to selective treatment [35],

leaving the rest of the women to rely on other non-selective cancer treatments such as

chemical and radiation therapy. Side effects from these non-selective cancer treatments

such as hair loss, lowered immune system, fatigue, nausea and abnormal digestive

processes (www.cancercare.org) are incentives to further investigate novel selective

breast cancer treatments.

6
1.2 Complementary and Alternative Medicine

1.2.1 Usage in the United States


Complementary and alternative medicine (CAM) covers a spectrum of medical

and health care practices that range from traditional to modern methods to prevent or cure

certain diseases and ailments [38, 39]. Complementary medicines are used alongside

with other conventional medicines. Whereas, alternative medicines are used to substitute

conventional medicines [39]. There are two types of CAM therapies: practitioner

therapies and self-care therapies [40]. Commonly used practitioner-type CAM therapies

include deep breathing, chiropractic treatments, massage therapy, acupuncture, and

hypnotism therapy. Commonly used self-care therapies include natural product

consumption, diet based therapy, meditation, and yoga [41, 42].

The use of CAM in the United States has been increasing over the past decade. In

2002, 36.0% of American adults reported that they used CAM. In 2007, this number

jumped to 38.3% [38, 42]. With over a third of the American adults and 11.8% of the

American children using CAM, the total out-of-pocket expenses spent on CAM healing

philosophies, therapies and products was $33.9 billion [40]. 44% of the out-of-pocket

costs were spent on non-vitamin, non-mineral products indicating the popularity of

natural products (figure 4) [40]. Unfortunately by definition, CAM is not considered a

conventional medicine due to the lack of evidence to prove how safe and effective they

are [43]. Due to the increasing popularity of natural products in the United States,

additional research is needed to inform the public of their effectiveness and side effects,

if any.

7
Figure 4.Total out-of-pocket complementary and alternative medicine costs among American adults in
2007 [source: Nahin, Richard, Barnes, Patricia, Stussman, Barbara J., and Bloom, Barbara, Costs of
Complementary and Alternative Medicine (CAM) and Frequency of Visits to CAM Practitioners: United
States, 2007.National Health Statistics Report, 2009. 18.]

1.2.2 Complementary and Alternative Medicine as Cancer Treatment


CAM treatments have been used for certain diseases and conditions such as head

or chest cold, anxiety, digestive illnesses, high cholesterol, and cancer [41, 42]. Many

cancer patients find additional ways to improve their diseased state or to prevent side

effects caused by conventional medicine [44]. An international survey showed that

35.9% of cancer patients used CAM [45]. In a 2005 study conducted by Molassiotiset al,

44.7% of the breast cancer patients studied (n=956) used CAM to either treat the disease

or increase physical and emotional well-being [46]. For a multitude of reasons, some

cancer patients turn to natural CAM products for treatment. 40.1% of the cancer patients

that utilize CAM rely on natural products [45]. Therefore, a solid investigation into the

effectiveness of CAM against cancer will be of great benefit to patients who utilize it.

8
1.3 Morindacitrofolia (Noni)

1.3.1 History
Morindacitrifolia(noni), also known as Indian Mulberry, nono, or cheese fruit, is

a small shrub that is native to South Asia and grows prominently in the tropics [47].

Noni shrub grows about two to six meters tall, with either rounded, elliptic or long leaves

and globular fruits that range from three to 20 cm long [48, 49]. Noni has been

traditionally used in Hawaii, Polynesia and Southeast Asia as either a dye, famine food,

[50] and is considered the most important medicinal plant based on reports of usage

across Polynesian cultures [48, 51, 52].

Figure 5.Noni fruit [source: www.resorthealth.com]

9
1.3.2 Traditional and Modern Uses
Traditional healers from Hawaii, Samoa, Rotuma, and Fiji have been known to

utilize different parts of the plant to treat wounds, infections, menstrual cramps and

bowel irregularities[48, 53]. Table 1 summarizes the traditional uses of noni prior to the

1800s. Contrary to popular belief, traditional healers do not commonly utilize the ripe

fruits. The most commonly used part of the noni plant was the leaf. Traditional healers

would heat noni leaves over a small fire then use it as a poultice by wrapping it around

the wounded or infected area [54]. Yet the use of noni fruits is popular as a modern

dietary supplement [48].

Noni plant part Traditional treatment for

Leaves Topical burns, headaches, fevers, neonatal inability to


breathe, bone fractures, menstrual cramps, back pain*,
insect infestations, boils, rheumatic pain, ulcers, gout,
internal bleeding, ringworm[48, 50, 55-57]

Fruit Sores in mouth, peeling or cracking of toes, boils, pimples,


blood impurities*, kava intoxication, insect infestations,
blotchy skin*, heart trouble , stomach pain*, menstrual
cramps*, heartburns , sore throat [48, 55, 56, 58]

Bark Stonefish poisoning, topical infections , asthma*, [55, 57,


58]

Root Topical infections, stomach pain* [55]

Stem Hernia [49]

Seeds

Flower Sore eyes, topical burns [57]

Table 1.Traditional uses of the noni

10
In the 1980s, commercial noni fruit juice (NJ) gained popularity fueled by Ralph

Heinicke’s claim that it contains Proxeronine, a precursor for Xeronine. Heinicke

suggested that Xeronine is an enzyme that can modify dysfunctional proteins to its

appropriate conformation[48, 53, 59]. In a study conducted in 1999, noni ranked the

second most commonly used complementary medicine[57]. The Nutrition Business

Journal reported that noni juice is the number one sales of single herbs in American in

2005 (www.nutritionbusiness.com). Noni was commonly used topically to treat

ailments up until the 1900s, when people began consuming noni products for its

medicinal properties. Fermented noni juice is popularly known as the “traditional”

method of preparing noni. It is believed that the Chinese, which composed a significant

proportion of the population in Hawaii, influenced fermenting the fruit. Reports have

shown that in the 1930s fermented noni juice was commonly prepared and consumed by

households in Hawaii. Furthermore, studies show an increase of demand for noni fruit

from early to late 1990’s[57]. Since then, multimillion-dollar companies have found

ways to package and sell this plant.

Due to the rising popularity of noni, many studies have been conducted to test its

efficacy as a medicine and have shown its anti-cancerous[60-62], anti-diabetic[63], anti-

tuberculosis[64], anti-viral[65], and anti-bacterial[66-68] effects. These studies were

already reviewed by Pawlus, Wang, Blanco, and Brown[49, 53, 69, 70]. The up to date

modern uses of noni are summarized in Table 2.

11
Noni plant part Treatment for
Leaves Tuberculosis ,helmintic infections, oxidative stress ,
open wounds , hyperlipidemia [64, 71-73]

Fruit Bacterial infections , cancer , pain , memory


impairment, reduced cerebral blood flow , diabetes ,
nausea , gastric ulcers , liver disease , low humoral
immunity , neuronal damage , stress-induced cognitive
impairment , helminthic infections , lung cancer,
oxidative stress, inflammation, post surgery nausea,
hyperglycemic, liver diseases [62-64, 66-68, 72, 74-88]

Bark
Root Viruses , hypotension , colorectal cancer , NF-kB
associated inflammation , hyperlipidemia , diabetes,
pain [62, 65, 71, 73, 89, 90]

Stem
Seeds Hyperlipidemia , oxidative stress , melanoma cancer
[91-93]
Flower
Table 2.Modern uses of noni

12
From skincare products to supplement capsules and tea, noni has been packaged

in multiple ways with claims to a better and healthier lifestyle. In 1992, a Maui massage

therapist encapsulated the dry powder form of noni fruit and sold it as supplement[57].

In 1996, John Wadsworth and Stephen Story introduced

Tahitian Noni Juice as a natural healthy supplement to the

United States (http://www.tahitian-juice.com). Other noni

juice companies such as, Virgin Noni Juice, Healing Noni,

and Noni Maui, sprouted since then with their own line of

noni juices. These bottles juices are sold either non-

fermented or fermented.

Figure 6.Encapsulated freeze-dried noni fruit

[source: www.estatenoni.com]

1.3.3 Fermented Noni Fruit Juice


To present date, there are two popular methods to prepare noni fruit juice:

fermented and non-fermented. In the fermented method, ripe yellow fruits are picked,

washed and left in glass air sealed containers for about two weeks to two months. The

juice is extracted and the pulp is removed. Some fermented juices are pasteurized.

Pasteurized noni has a distinctively different taste as well as different pH. To prepare

non-fermented noni juice, fresh ripe fruits are pressed through a juicer. The juice can be

consumed pure, or diluted with water or other fruit juices. In some juices, the starchy

13
pulp of the noni is mixed with either apple or grape fruit juice to increase palatability

[94].

Most noni fruits are consumed as a naturally fermented juice [95]. It is suggested

that in Hawaii, this type of noni preparation might have originated from the prominent

Chinese ethnic influence in the mid-1800s [57]. Based on local records, Hawaii residents

commonly picked, fermented, and consumed noni fruits before meals to treat diabetes,

heart trouble and blood pressure [96, 97]. Recent literature suggests that the probiotic

properties of most fermented foods are beneficial to your health [98, 99]. The beneficial

effects include improved lactose digestion, controlled gastrointestinal infection, reduced

cholesterol levels and stimulated immune system [100]. Besides the difference in biotic

cultures found in fermented foods compared to non-fermented foods, the chemical

constituents may vary as well. In 2007, Shu-Chuan Yang et al demonstrated that the

chemical composition of fermented noni juice (fNJ) differed from that of non-fermented

NJ. The fNJ yielded higher phenolic compounds, condensed tannins, flavonoids and

scopoletin than non-fermented NJ. Additional results from the study showed that fNJ has

higher anti-oxidative activity as compared to non-fermented NJ [101].

1.4 Noni and Cancer

1.4.1 In vitro studies


In 1999, Hiramizu and Furusawa tested the antitumor properties of a

polysaccharide-rich ethanol extract of noni fruit precipitate (noni-ppt). They found that

noni-pptsuppressed the growth of Lewis lung (LLC) peritoneal carcinoma cells by up

14
regulating several mediator and murine effector cells[61]. In 2006, Arpornsuwan and

Punjanon also conducted a study investigating the effects of noni fruit extract against

cancer cells. They observed cytotoxic effects against human laryngeal carcinoma

(HepG2), human breast ER+vecarcinoma (MCF7), and human neuroblastoma (LAN5)

cell lines[102]. An interesting study conducted in 2003 by Hornicket al. looked at the

anti-angiogenic effects of noni juice in human breast tumor explants by using a three-

dimensional fibrin clot matrix model. The commercial noni juice used demonstrated

anti-angiogenic and degenerative effects against the tumor explants[103].

Aside frominvestigating various noni fruit extracts, several studies have shown

the anti-cancerous effects of chemical extractions from other noni plant parts. In 1993,

Hiramatsuet al. demonstrated that the damnacathal and anthraquinones extracted from

noni root inhibited ras functions in rat kidney cells [104]. Later in 2011, Nualsanitet al.

also utilized damnacanthal and anthraquinone extracts from the root and found that the

extracts exhibited pro-apoptotic effects by inducing caspase activity in human colorectal

cancer cells [105]. New chemical compounds such as 6-O-(beta-D-glucopyranosyl)-1-O-

octanoyl-beta-D-glucopyranose and asperulosidic acid extracted from noni fruit juice also

displayed anti-cancerous properties against mouse epidermal cells[106].

1.4.2 In vivo studies


In vivo studies play a crucial role in determining the efficacy of the plant on

humans. Table 3is a table compiled by Brown that summarizes the in vivo studies

conducted since 1994 to determine the effects of noni fruit juice against cancer. For all

the studies mentioned in Table 3, subjects that received noni juice displayed a significant

15
amount of anti-cancerous effects. Taskinet al.’s Ehrlich ascites tumor injected Balb-c

mice showed a reduction in tumor size in mice that have received oral dosages of noni

juice as compared to contraol. Consequently, the same results can be seen in Stoner et

al.’s study done on rats. A reduction in tumor size has been observed in rats that received

noni juice as a part of their diet, compared to control.

16
Table 3. In vitro studies on the effects of noni and cancer

17
Table 3. (continued)

18
1.4.3 Molecular Mechanisms
Although there have been many studies conducted on the efficacy of noni against

cancer, many of them do not explain the molecular mechanisms behind the anti-

cancerous effects. Table 4 summarizes studies done thus far that demonstrate signal

transduction pathways caused by noni.In 2007, Takashima et al. studied the apoptotic

effects of noni leaf extract via tumor necrosis factor (TNF) and TNF related apoptosis-

inducing ligand (TRAIL)[107]. Other studies done by Hirazumi and Furusawa also

examined the apoptotic effects of noni through TNF. In addition, they also demonstrated

that increased levels of interleukin-1β (IL-1β), IL-10, IL-12, interferon-γ (IFN-γ), and

nitric oxide (NO) may have contributed to the anti-cancerous effects [61]. Other studies

demonstrate how noni induces apoptosis by affecting the immune system. In 2008, Li et

al. showed that fermented noni juice increased levels of granulocytes and natural killer

(NK) cells in the peripheral blood,peritoneum, and spleen therefore contributing to the

decrease in tumor size of mice[108]. Soon after, Stoner et al. demonstrated that noni fruit

powder inhibited tumorigenesis by reducing the levels of cytokines in rats [109].

Although many studies have demonstrated anti-cancerous effects of noni, most

fail to identify the molecular mechanisms involved. Thus far, only eight mechanisms

have been identified and additional studies are warranted. Identifying the proteins and

signal transduction pathways involved in noni’s anti-cancerous properties may aide in

determining the benefits of noni against cancer.

19
Type of Noni Extract Model used Molecules observed and measurable outcomes
Damnacathal and anthraquinone Ras transformed normal rat kidney Inhibited ras function [104]
extracted from noni roots using cells
chloroform

Noni-ppt Lewis lung carcinoma cells TNF-α, IL-1β, IL-10, IL-12, IFN-γ, and NO levels
subcutaneously injected in C57BL/6 increased [61]
mice
6-O-(beta-D-glucopyranosyl)-1-O- Mouse epidermal JB6 cells Suppressed 12-O-tedtradecanoylphorbol-13-acetate
octanoyl-beta-D-glucopyranose and (TPA) and epidermal growth factor (EGF) induced
asperulosidic acid extracted from activator protein 1 (AP1) transactivation[106]
noni fruit juice

Anthraquinone compound extracted Immortalized human T-lymphocyte Increased TRAIL induced cytotoxic activity [107]
from noni leaves using methanol Jurkat cells
Fermented noni exudate C57BL/6J mice Increased levels of granulocytes and NK cells in the
peripheral blood,peritoneum, and spleen[108]
Noni fruit powder Rats Inhibited N-nitrosomethylbenzylamine
(NMBA) induced tumorigenesis in the rat
esophagus and reduced cytokine, IL-5 and GRO/KC
(rat homologue for human IL-8 serum levels) [109]
Damnacanthal extracted from noni Human colorectal carcinoma cells Induced nonsteroidal anti-inflammatory activated
roots gene-1 (NAG-1), caspase, and transcription factor
CCAAT/enhancer binding protein β (C/EBPβ) [105]

Noni fruit juice Human lung carcinoma A549 cells Induced phosphorylation of protein kinase B (PKB),
extracellular regulated protein kinase 1/2 (ERK-1/2),
c-Jun N-terminal kinase-1 (JNK-1), and eukaryotic
translation initiation factor-2α (eIF-2α)[110]
Table 4.Summary ofmolecular mechanisms involved in the effects of noni against cancer

20
Chapter 2: Hypothesis and Aims

2.1 Hypothesis

Based on our preliminary data and literature review, we hypothesize that fermented noni

fruit juice (fNJ) will prevent proliferation and induce programmed cell death (apoptosis)

in MCF-7, and ER –veMDAMB-231cancer cells by modulating cell cycle arrest proteins

as well as inhibiting cell death proteins called “inhibitors of apoptosis” (IAP).

Figure 7.fNJ induces apoptosis through hypothesized pathways

21
2.2 Specific Aims

2.2.1 Specific Aim 1

To investigate the effects of fNJinnormal human breast, MCF10A and cancerous human

breast cells, MCF-7 and MDAMB-231.

Objective 1: Identify dosage of fNJ that are non-toxic in normal cells, but will

induce cell death in MCF-7and MDAMB-231 cells.

Approach: Toxicity of varying doses of fNJ will be measured in cells after 24, 48,

72 and 96 h by using commercial 96 well non-radioactive cell proliferation

(MTT) assay kit.

Objective 2: Identify the apoptotic proteins present infNJtreated human breast

cancer cells that affects proliferation or causes cell death in MCF-7 and

MDAMB-231.

Approach:Apoptosis will be measured using an apoptosis array in cells treated

with two doses of fNJ that will achieve about 60-80% cell death.

2.2.2 Specific Aim 2

To identify the anti-proliferative and apoptotic mechanisms inducedbyfNJin MCF-7

and MDAMB-231 cells.

Objective 1: Identify fNJ-induced apoptotic proteins in MCF-7 and the apoptotic

signaling mechanisms.

22
Approach: Proteins will be identified from apoptosis array and confirmed by

western blotting.

Objective 2:Identify fNJ-induced apoptotic proteins in MDAMB-231 and the

apoptotic signaling mechanisms.

Approach 1: Proteins will be identified by apoptosis array will be confirmed by

western blotting.

Approach 2: Nuclear and cytosolic proteins of fNJ treated MCF-7 and MDAMB-

231 cells will be extracted and be confirmed by western blotting.

23
Chapter 3: Materials and Methods

3.1 Preparation of fNJ with endotoxins removed


Morindacitrofolia(noni) fruits were obtained from noni trees on the University of Hawaii

ManoaMagoon facility, Honolulu, Hawaii. The fruits were rinsed, dried, and left to

ferment in airtight jars placed in indirect sunlight for 14 days. The fermented juice was

extracted and centrifuged at 3000 rpm at room temperature for 15 minutes. The

supernatant was filtered with a 0.45μm filter then a 0.20μm filter. The pH of the

fermented noni juice (fNJ) was adjusted to pH 7 using 10mM NaOH .

Endotoxins were removed from the fNJ using Detoxi-Gel Endotoxin Removing Columns

(Pierce Biotechnology, Rockford, IL, U.S.A) as per manufacturer’s instructions. The fNJ

with removed endotoxins were then aliquoted into 1.5 mL tubes and store in -80°C, then

thawed once for cell treatment.

3.2 Cell Culture


Estrogen receptor positive (ER +ve) human breast cancer cells, MCF-7 were generously

donated by Dr. June Panee. MCF-7 cells were maintained in Dulbecco’s modified eagle

medium (DMEM) (ATCC, Manassas, VA, U.S.A.) with 10% heat inactivate fetal bovine

serum (FBS), 1% penicillin/streptomycin, and 10mg/ml insulin.

Estrogen receptor negative (ER -ve) MDA-MB-231 cells were generously donated by Dr.

June Panee. MDA-MB-231 cells were maintain in Minimal essential medium eagle

(MEME) (ATCC, Manassas, VA, U.S.A)) with 10% heat inactivate fetal bovine serum

(FBS) and 1% penicillin/streptomycin.

24
3.3 MTT assay
Cell viability was determined and analyzed by the CellTiter96 Non-Radioactive cell

proliferation assay (MTT) (Promega, Madison, WI, U.S.A.). MCF-7 and MDA-MB-231

cells were cultured and plated on to 96-well plates at a density 100,000 cells/ml for 48 h

prior to the first treatment. The cells were treated with 2.5%, 5%, 10%, 15%, and 20%

fNJ (v/v) for a total of 96 h. Dosages were calculated based on the recommended dose

made by commercial noni juice companies. An average adult is recommended to

consume two ounces twice a day, which equates to 59.14 milliliters twice a day. Since an

average adult has a blood volume of 5 liters, the average adult would be consuming

1.18% of its blood volume per serving and 4.72% over the course of two days.

3.4 Human apoptosis antibody array

The expression profile of apoptosis-related proteins was detected and analyzed using a

human apoptosis array kit (R&D Systems, Minneapolis, MN, U.S.A.) as per

manufacturer’s instructions. This array contains duplicate spots of 35 apoptosis-related

proteins. Briefly, the membrane containing immobilized apoptosis-related antibodies was

blocked with bovine serum albumin for 1 h on a rocking platform at room temperature.

The membrane was then incubated with MCF-7 or MDA-MB-231 cell lysate treated with

or without fNJ along with detection antibody cocktail overnight at 2°C to 8°C on a

rocking platform. The membranes were incubated with streptavidin-horseradish

peroxidase conjugate followed by chemiluminescent detection reagent. The membranes

25
were scanned and pixel density was presented by quantifying the mean spot densities

from two experiments.

3.5 Western Blot

After the in vitro experiments, protein extracts were prepared from the cells using special

solubilizing buffer (SSB) containing 25 mMTris-HCl (pH 7.4), 2 mM Na3VO4, 10

mMNaF, 10 mM Na4P2O7, 1 mM EGTA, 1 mM EDTA, and 1 mM PMSF. After protein

was extracted, its concentration was determined by the Bradford assay (Bio-Rad

Laboratories, Hercules, CA, U.S.A). Equal amounts of protein extracts were separated

on a 4–10% gradient polyacrylamide gel and thenelectrotransferred to polyvinyl

diflouride (PVDF) membrane using a Bio-Rad mini-transfer tank. PVDF membranes

were activated in 100% methanol for 1 minute. Membranes were incubated with primary

antibodies overnight. The protein bands were detected with HRP-conjugated secondary

antibodies and then imaged developed on a film using Pierce enhanced

chemiluminescence (ECL) western blotting substrate (Thermo Scientific, Rockford, IL,

CA, USA). Membranes were stripped with Re-blot (Millipore, Temecula, CA, USA) for

20min minutes at room temperature and reprobed with antibodies to β-tubulin to control

for differences in loading.

The primary antibodies used were survivin, claspin, cytochrome-c, and NFkB (Santa

Cruz Biotechnology, Santa Cruz, CA, U.S.A.). The secondary antibodies used was goat

anti-mouse from Santa Cruz (Santa Cruz Biotechnology,Santa Cruz, CA. U.S.A.).

3.6 Statistical Methods

26
All tabulated data is presented as mean ± SEM. Analysis of statistical significance and

graphing was done using GraphPad Prism 5. P-values of P< 0.05 were considered to be

statistically significant. Means were tested for statistical significance difference using

one-way ANOVA and Tukey’s post-test analysis.

27
Chapter 4: Results

4.1 fNJ decreases cell viability in breast carcinoma cells

MTT assays wereperformed on MCF-7 and MDA-MB-231 cells after treated once

with 2.5%, 5%, 10%, 15%, and 20%fNJ (v/v) over the course of 48 hours. No cell death

was observed in cells that received only one treatment. Therefore two treatments were

used over the course of 96 hours (figure 10 and 11). In both cell lines, cell viability has

been observed to be significantly lower in all the treatments except for 2.5% fNJ (v/v) as

compared to control. For both breast carcinoma cell lines, 40% - 60% cell death was

observed in the 15% fNJ treated as compared to control. 20% fNJ treated cells have been

observed to have 70% - 85% cell death as compared to control. Based on these results,

15% fNJ was chosen for further experiments since higher doses induced cell death by

80%, while lower doses were non-toxic. Although MTT assay results show significant

change in cell viability in treated as compared to control in both cell lines, visual

observation shows that the cell death in treated MDAMB-231 cells are more apparent

than treated MCF-7 cells. In order to determine the toxicity of fNJ against non-

carcinoma cells, MCF10A, a normal breast epithelial cell line was treated under the same

conditions as the breast carcinoma cell lines (figure 12). MTT assay results showed that

there is no significant decrease of cell viability in any of the treatments compared to

control in MCF-10A normal human breast epithelial cells.

28
4.2 Apoptotic proteins identified in fNJ treated breast carcinoma cells
An apoptosis array was used to determine the changes in apoptotic protein levels

in 15% fNJ (v/v) treated breast cancer cells compared to control. The apoptosis array

detects 48 different proteins that are related to the downstream or upstream down-

regulation or up-regulation of apoptosis (figure 13). In 15% fNJ treated MCF-7 cells, a

significant decrease of B-cell lymphoma 2 (Bcl-2), cellular inhibitor of apoptosis protein

1(cIAP-1),survivin, claspin, and phosphorylated Rad 17 (phospho-Rad17) has been

observed compared to control (figure 14). Survivin and claspin resulted in a 60%

decrease, which were the highest amount of decrease compared to other proteins

observed. In addition, a significant 70% increase of heat shock protein 60 (HSP60) has

been observed in treated compared to control.

In 15% fNJ treated MDA-MB231 cells, a significant decrease was observed in

cIAP-1, cellular inhibitor of apoptosis protein 2 (cIAP-2), survivin, claspin, and fibroblast

associated tumor necrosis factor receptor super family member 6 (Fas/TNFRSF6) as

compared to control (figure 15). Fas/TNFRSF6 and cIAP-2 resulted in an 85% and 70%

decrease, respectively, which were the highest amount of decrease compared to the other

proteins. Subsequently, a 20% increase of clusterin has been observed in the treated

samples compared to control.

4.3 fNJ inhibits survivinin MCF-7 (ER+ve) cells


Western blots were used to further verify the change in apoptotic proteins

identified from the Apoptosis Array. In the apoptosis array, 15% fNJ treated breast cells,

MCF-7 displayed significantly lower levels of survivin and cIAP-1 compared to control.

Both survivin and cIAP-1 belong to a class of proteins called the inhibitors of apoptosis

29
(IAP) proteins. Western blot analysis demonstrated that there was a 40 – 60% decrease

in survivin protein levels in 15% fNJ treated breast carcinoma cells as compared to

control (figure 16).

4.4 fNJ up-regulates cytosolic levels of cytochrome c in MCF-7 (ER+ve) cells


In the apoptosis array, Bcl-2 levels were observed to be significantly lowered in

the 15% treated MCF-7 cells compared to control. Since Bcl-2 is known to inhibit the

release of cytochrome c from the mitochrondria to the cytosol, western blots were used to

detect the levels of cytochrome c in cytosolic protein extractions of MCF-7 cells in 15%

fNJ treated and control. Western blot analysis demonstrated a significant 200% increase

in the treated compared to control (figure 17).

4.5 fNJ does not affect cytosolic and nuclear levels of nFκB in MCF-7 (ER+ve) cells
Western blot analysis was used to determine cytosolic and nuclear protein levels

of the pro-inflammatory and pro-survival transcription factor, nFκB (figures 18 and 19).

No significant changes were observed in the 15% fNJ treated MCF-7 cells compared to

control.

4.6 fNJ inhibits survivin in MDAMB-231 (ER-ve) cells


Western blots were used to further verify the change in apoptotic proteins

identified from the Apoptosis Array. In the apoptosis array, 15% fNJ treated breast cells,

MDAMB-231 displayed significantly lower levels of surviving, cIAP-1 and cIAP-2

compared to control. Survivin, cIAP-1 and cIAP-2, all belong to a class of proteins

called the inhibitors of apoptosis (IAP) proteins. Western blot analysis demonstrated that

30
there was a 40 – 50% decrease in survivin protein levels in 15% fNJ treated breast

carcinoma cells as compared to control (figure 20).

4.7 fNJ does not affect cytosolic levels of cytochrome c in MDAMB-231 (ER-ve)
cells
In the apoptosis array, Bcl-2 levels were observed to have no significant difference in the

15% treated MDAMB-231 cells compared to control. Since Bcl-2 is known to inhibit the

release of cytochrome c from the mitochrondria to the cytosol, western blots were used to

detect the levels of cytochrome c in cytosolic protein extractions of MDAMB-231 cells in

15% fNJ treated and control. Western blot analysis confirm the apoptosis array by

showing no significant changes in the treated MDAMB-231 cells compared to control

(figure 21).

4.8 fNJ does not affect cytosolic and nuclear levels of nFκB in MDAM-231 (ER-ve)
cells
Western blot analysis was used to determine cytosolic and nuclear protein levels of the

pro-inflammatory and pro-survival transcription factor, nFκB (figures 22 and 23). No

significant changes were observed in the 15% fNJ treated MDAMB-231 cells compared

to control.

31
4.9Figures of results

Figure 8. Phase contrast photomicrographs of MCF-7 cells treated with fNJ for 96 hours
(magnification x 200)
(A) Untreated control (B) 15% fNJ (v/v) (C) 20% fNJ (v/v)

Figure 9. Phase contrast photomicrographs of MDAMB-231 cells treated with fNJ for 96
hours (magnification x 200)
(A) Untreated control (B) 15% fNJ (v/v) (C) 20% fNJ (v/v)

32
Figure 10. Effects of fNJ on MCF7 cell viability
Two independent experiments were performed in replicates of seven. Data is represented
a, b
as percentages of control and as mean ± SE (n=14). Mean values with common letters

to control do not differ (p<0.05).

33
Figure 11. Effects of fNJ on MDAMB-231 cell viability
Two independent experiments were performed in replicates of seven. Data is represented
a, b
as percentages of control and as mean ± SE (n=14). Mean values with common letters

to control do not differ (p<0.05).

34
Figure 12. Effects of fNJ on MCF-10A cell viability
Two independent experiments were performed in replicates of seven. Data is represented
a, b
as percentages of control and as mean ± SE (n=14). Mean values with common letters

to control do not differ (p<0.05).

35
Figure 13. Human apoptosis array overlay

36
Figure 14. Effects of 15% fNJ (v/v) on apoptotic proteins in MCF-7 cells as compared to
control.
Two independent experiments were performed in triplicate. Values are mean ± SE (n=6).
a, b
Mean values with common letters do not differ (p<0.05).

Figure 15. Effects of 15% fNJ (v/v) on apoptotic proteins in MDAMB-231 cells as
compared to control.
Two independent experiments were performed in triplicate. Values are mean ± SE (n=6).
a, b
Mean values with common letters do not differ (p<0.05).

37
Figure 16. fNJ reduced survivin levels in MCF-7 cells
Figure 16A shows images of the blots.Figure 16B represents the relative intensity of the

bands measured by densitometry units. Two independent experiments were performed in


a, b
triplicate. Values are mean ± SE (n=6). Mean values with common letters do not differ

(p<0.05).

38
Figure 17. fNJ increases cytosolic cytochrome c in MCF-7 cells
Figure 17A shows images of the blots. Figure 17B represents the relative intensity of the

bands measured by densitometry units. Two independent experiments were performed in


a, b
triplicate. Values are mean ± SE (n=6). Mean values with common letters do not differ

(p<0.05).

39
Figure 18. fNJ did not affect changes in cytosolic nFkB levels in MCF-7 cells

Figure 18A shows images of the blots. Figure 18B represents the relative intensity of the

bands measured by densitometry units. Two independent experiments were performed in


a, b
triplicate. Values are mean ± SE (n=6). Mean values with common letters do not differ

(p<0.05).

40
Figure 19. fNJ did not affect changes in nuclear nFkB in MCF-7 cells
Figure 19A shows images of the blots. Figure19B represents the relative intensity of the

bands measured by densitometry units. Two independent experiments were performed in


a, b
triplicate. Values are mean ± SE (n=6). Mean values with common letters do not differ

(p<0.05).

41
Figure 20. fNJ reduced survivin levels MDAMB231

Figure 20A shows images of the blots. Figure 20B represents the relative intensity of the

bands measured by densitometry units. Two independent experiments were performed in


a, b
triplicate. Values are mean ± SE (n=6). Mean values with common letters do not differ

(p<0.05).

42
Figure 21.fNJdid not affect cytosolic levels of cytochrome c in MDAMB-231 cells
Figure 21A shows images of the blots. Figure 21B represents the relative intensity of the

bands measured by densitometry units. Two independent experiments were performed in


a, b
triplicate. Values are mean ± SE (n=6). Mean values with common letters do not differ

(p<0.05).

43
Figure 22.fNJ does not affect cytosolic nFkB in MDAMB-231 cells
Figure 22A shows images of the blots. Figure 22B represents the relative intensity of the

bands measured by densitometry units. Two independent experiments were performed in


a, b
triplicate. Values are mean ± SE (n=6). Mean values with common letters do not differ

(p<0.05).

44
Figure 23. fNJ did not affect nuclear nFkB levels in MDAMB-231 cells
Figure 23A shows images of the blots. Figure 23B represents the relative intensity of the

bands measured by densitometry units. Two independent experiments were performed in


a, b
triplicate. Values are mean ± SE (n=6). Mean values with common letters do not differ

(p<0.05).

45
Chapter 5: Discussion

In both MCF-7 and MDAMB-231 breast carcinoma cell lines, cell death was

observed in fNJ treated cells. Normal cells grown on a culture exhibit a behavior called

contact inhibition where they limit themselves to grow in a single layer[1]. Cancer cells

do not exhibit this behavior. Instead, cancerous cells tend to grow uncontrollably and

overlap each other[1]. Visual observation of the control breast carcinoma cells confirms

that behavior.MCF-7 and MDAMB-231 cells that were treated with fNJ cease to grow

uncontrollably. Rounding of cells was observed in the fNJtreated group, indicating cell

death possibly via apoptosis. 15% fNJ dosage was chosen for further experimentation

because if caused a significant amount of cell death yet it was not severely toxic to the

cells. Since an average adult has about 5 liters of blood, a 15% fNJ treatment would

mean that an average adult would have to consume 375 mililiters of fNJ per day.

Previous studies conducted by West et al. demonstrated that noni juice is clinically safe

even when consumed in amounts as high as 750 mililiters per day[111].Resulting data

from MTT assay demonstrates that cell viability decreases as fNJ treatment increases,

which agrees with visual observations. This suggests that fNJ somehow causes cell death

in MCF-7 and MDAMB-231.

Apoptosis is a form of intrinsic programmed cell death that can be characterized

by several morphological features such as cell shrinkage, membrane blebbing, and

nuclear DNA fragmentation [112]. Apoptosis plays a crucial role in regulating cell

growth and tissue homeostasis. Therefore, an important factor to consider in anticancer

46
research, are the extrinsic and intrinsic signal transduction pathways of apoptosis. The

intrinsic pathway of apoptosis can be activated via the mitochondria or extrinsic receptors

such as TRAIL or FADD (figure 24)[113].The mitochrondrial apoptosis pathway initiates

apoptosis through the release of cytochrome c, apoptosis inducing factor (AIF), or

Smac/DIABLO. The release of these proapoptotic proteins into the cytosol activates cell

death via the caspase cascade [113-115]. In cancer, the uncontrolled growth of abnormal

cells may be induced by anti apoptotic proteins such as survivin, Bcl-2, XIAP, cIAP-1

and cIAP-2. By examining the effects of fNJ on a molecular level, a specific molecular

approach to developing anticancer therapies can be developed.

Figure 24. Extrinsic and intrinsic pathways of apoptosis [source: Fulda, S. and Debatian, K.M.,
Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy.Oncogene, 2006.25: p.
4798-4811.]

47
In order to investigate the apoptotic mechanisms involved in cell death, an

apoptosis array was used to view the changes of apoptotic protein levels between control

and 15% fNJ treated MCF-7 and MDAMB-231 cells. Inbreast cancer cells, significant

reductions in cIAP-1, cIAP-2, Bcl-2, survivin, and claspin were observed in the 15% fNJ

group. A reduction of claspin has been observed in treated cells compared to control.

Claspin is a protein involved cell cycle arrest [116]. When DNA damage occurs, the

cells will either repair the damage or induce cell cycle arrest. ATR phosphorylates

claspin then recruits Plx1 to claspin causing claspin to dissociate from the chromatin.

The dissociation of claspindownregulates Chk1 which inhibits mitosis [116]. Thus, the

reduction of claspin indicates the anti-proliferative effect of fNJ.

A common characteristic seen in cancer is the dysregulation of apoptosis[117].

Therefore proteins involved in pro or anti apoptotic signals are of interest when it comes

to cancer research. Survivin and cIAP-1 belong to a class of proteins call the Inhibitor of

Apoptosis (IAP) proteins, which are known to suppress apoptotic cell death[118]. An

overexpression of IAP proteins is commonly observed in human cancers and has been

associated with tumor progression and treatment failure [119]. Thus, IAP proteins are

potential targets for cancer therapy and drug development.Survivin is an IAP that is

commonly expressed in breast cancer tissues and plays a big role in cell survival and

chemoresistance by inhibiting caspases 3 and 9[120, 121]. Results show that MCF-7 and

MDAMB-231 cells treated with 15% fNJ have approximately 50% less survivin than the

non-treated control. This suggests that fNJ inhibits the expression of survivin in breast

cancer cells. Previous studies have shown that IAPs such as, cIAP-1, cIAP-2, survivin,

and XIAP, block apoptosis byinhibiting caspases 3 and 9 downstream of Bax, Bik, Bak,

48
and cytochrome c[115, 118, 122]. Thus suggesting that fNJ’s pro-apoptotic effects

contribute to the inhibition of IAPs, which in return activates caspases 3 and 9.

Bcl-2 is a protein that is located mainly in the nuclear membrane, endoplasmic

reticulum and mitochondrial membrane [123]. It is known to inhibit the release of

cytochrome c from the mitochondria to the cytosol and therefore inhibiting caspases 3

and 9 induced apoptosis[114, 122, 123]. Results show that MCF-7 cells treated with 15%

fNJ significantly reduces Bcl-2 levels by 40% compared to non-treated control. In

addition, western blot results show an increase in cytosolic cytochrome c levels by 300%

in treated compared to control. These results demonstrate the indirect correlation of Bcl-

2 and cytosolic cytochrome c published by Klucket al., indicating that fNJ inhibits Bcl-2

therefore allowing the release of cytosolic cytochrome c and activating apoptosis through

caspases 3 and 9. These findings suggest the fNJ induces apoptosis in MCF-7 ER+ve

cells through the mitochondrial apoptosis pathway. The same results were not found in

MDAMB231 ER-ve cells. There were no significant changes in cytosolic cytochrome c

levels in 15% fNJ treated MDAMB-231 cells compared to control, suggesting that fNJ

does not induce apoptosis in MDAMB-231 cells through the mitochondrial apoptosis

pathway.

49
Figure 25.Roles of Bcl-2[source: Hengartner, Michael O., The biochemistry of apoptosis. Nature
2000. 407: p. 770-776.]

The anti apoptotic proteins discusses earlier (Bcl-2, cIAP-1, cIAP-2, and survivin)

are regulated by a pro-inflammatory transcription factor, NfκB[124]. NfκB works

upstream and controls the gene expression of certain IAP proteins [118, 125]. Evidence

suggests that NfκBmediate desensitization, chemoresistance, and radio resistance [126].

Consequently, Hirazumiet al. demonstrated that noni-ppt increased levels of TNF-α,

which functionsextrinsically upstream of NfκB[61]. To further verify the role of NfκB,

our results have demonstrated a significant decrease in tumor necrosis factor receptors

(TNFR) in 15% fNJ treated MCF-7 and MDAMB-231 cells compared to control. Since

inactive NfκB remain in the cytosol of cells [124], changes in NfκB levels wereexpected

to be seen in nuclear and cytosolic protein extracts of fNJ treated MCF-7 and MDAMB-

231. However, no changes in cytosolic and nuclear NfκB levels were observed in both

breast carcinoma cells lines. Although previous studies have indicated the early

50
activating properties of NfκB[127, 128]. Since the proteins were extracted 96 hours after

treatment, perhaps NfκB has already activated transcription and left the cell’s nucleus.

Many active chemical constituents in noni have been isolated and identified.

Studies have been conducted to identify the anticancer signal transduction pathways

induced by chemicals found in noni such as anthraquinone, scopoletin and damnacanthal.

For example, damnacanthal isolated from noni have been demonstrated to induced

apoptosis by activating caspase [105]. Our data has indicated that naturally fermented

noni fruit juice as a whole is also as effective as the isolated components.

51
Figure 26. The role of NF-kB in apoptosis.[source: Aggarwal, Bharat B., Nuclear factor-
kB: the enemy within.Cancer Cell, 2006.6: p. 203-208.]
(A) NfκB is activated by inflammatory agents, carcinogens, infection, cytokines, and

tumor promoters. (B) NfκB activates the expression of genes related to cell survival,

proliferation, angiogenesis, and inflammation.

52
Chapter 6: Conclusion

Although several studies have shown the apoptotic effects of fNJ in in vivo

models, to our knowledge, no studies have been conducted to identify the molecular

mechanisms association with the anti-cancer effects of fNJ.Our studies have shown

thatfNJ induces apoptosis in human breast carcinoma, MCF-7 and MDAMB-231 cells

through various intrinsic pathways.In both cell lines, fNJ is able to inhibit anti-apoptotic

proteins thus allowing the breast cancer cells to undergo apoptosis via caspases 3 and 9.

In addition, fNJ inhibited anti-apoptotic protein, Bcl-2 in treated MCF-7 cells. The

inhibition of Bcl-2 allows the release of cytochrome c into the cytosol. High cytosolic

levels of cytochrome c induced apoptosis by activating the caspase cascade.

Studies on the molecular mechanisms of breast cancer offer different and possibly

more selective treatments. Although hormone treatments are available for women with

ER+ve breast carcinoma, women suffering from ER-ve breast carcinoma will have to

turn to non-selective treatments such as chemical or radiation therapy. fNJ delivers

promising results as a selective treatment for ER+ve and ER-ve breast cancer.

6.1 Significance
Currently, the popular methods of cancer treatment are surgery, chemotherapy,

radiotherapy and hormone therapy (www.breastcancer.org). Unfortunately each

treatment has a side effect. More importantly, these treatments, besides hormone therapy,

are systematic and will kill normal healthy cells therefore leaving the patients with a

number of side effects, such as hair loss, lowered immune system, nausea, headache, and

53
possiblychemoresistanceand radioresistance. fNJcan potentially offer a cost friendly and

selective way of battling breast cancer.

6.2 Future studies


Since apoptosis is facilitated through extrinsic and intrinsic pathways [112], it is

important to examine all aspects of it. Therefore, our lab will be not only further

analyzing the intrinsic pathways by looking at the presence of caspase, but also the

extrinsic pathway by looking at the presence of TNF-α and TNFR. In addition, our lab

will be reexamining the pro-survival activites of early activating transcription factor,

nFκB by extracting proteins 1-4 hours post treatment.

Anti-proliferation is an important factor to consider when looking at cancer

therapies. Our studies contain preliminary data demonstrating the anti-proliferative

effects of fNJ. Further studies will be conducted to verify it by looking in other proteins

such as claspin, phosphorylated claspin, and chk1 in treated compared to control.

Many commercial noni fruit juice products are not fermented. Therefore a study

on the comparison of the fermented and non-fermented fruit juices will be beneficial to

their consumers. In addition, chemical fractionation and analysis is warranted to identify

the constituents responsible for the anti-cancerous effects of noni fruit juice.

54
Reference:

1. Cancer Facts and Figures: 2011-2012. American Cancer Society, 2012.


2. United States Cancer Statistics: 2007 Top Ten Cancers Web-based Report. 2007
[cited 2012 March 8]; Available from: http://www.cdc.gov/uscs.
3. Breast Cancer Facts and Figures: 2011-2012. American Cancer Society, 2012.
4. Breast Cancer. American Cancer Society, 2011.
5. Schawartz, Gordon F., Hughes, Kevin S., Lynch, Henry T., Fabian, Carol J.,
Fentiman, Ian S., Robson, Mark E., Domcheck, Susan M., Hartmann, Lynn C.,
Holland, Roland, and Winchester, David J., Proceedings of the International
Conference on Breast Cancer Risk, Genetics, & Risk Management, April, 2007.
The Breast Journal, 2009. 15(1): p. 4-16.
6. Colditz, Graham A., Kaphingst, Kimberly A., Hankinson, Susan E., and Rosner,
Bernard, Family history and risk of breast cancer: nurses' health study. Breast
Cancer Research and Treatment, 2012.
7. Roy, Rohini, Chun, Jarin, and Powell, Simon N., BRCA1 and BRCA2: different
roles in a common pathway of genome protection. Nature Reviews Cancer, 2012.
12: p. 68-78.
8. Cybulski, Cezary, Wkolorczyk, Dominika, Jakubowska, Anna, Huzarski, Tomasz,
Byrski, Tomasz, Gronwalk, Jacek, Debniak, Tadeusk, Gorski, Bohdan, Narod,
Steven A., and Lubinski, Jan, The risk of breast cancer in women with a CHEK2
mutation. Hereditary Cancer in Clinical Practice, 2012. 10(1): p. A5.
9. Masciari, Serena, Dillon, Deborah A., Rath, Michelle, Robson, Mark , Weitzel,
Jeffrey N., Balmana, Judith, Gruber, Stephen B., Ford, James M., Euhus, David,
Lebensohn, Alexandra, Telli, Melinda, Pochebit, Stephen M., Lypas, Georgios,
and Garber, Judy E., Breast cancer phenotype in women with TP53 germline
mutations: a Li-Fraumeni syndrom consortium effort. Breast Cancer Research and
Treatment, 2012.
10. Brennan, Kevin, Garcia-Closas, Montserrat, Orr, Nick, Fletcher, Olivia, Jones,
Michael, Ashworth, Alan, Swerdlow, Anthony, Thorne, Heather, Riboli, Elio,
Vineis, Paolo, Dorronsoro, Miren, Clavel-Chapelon, Francoise, Panico, Salvatore,
Onland-Moret, Charlotte, Trichopoulos, Dimitrios, Kaaks, Rudolf, Khaw, Kay-
Tee, Brown, Robert, and Flanagan, James M., Intragenic ATM methylation in
peripheral blood DNA as a biomarker of breast cancer risk. Cancer Research,
2012.
11. Becker, Karl-Friedrich, Atkinson, Michael J., Reich, Ulrike, Becker, Ingrid,
Nekarda, Hjalmar, Siewert, Jorg R., and Hofler, Heinz, E-Cadherin Gene
Mutations Provide Clues to Diffuse Type Gastric Carcinomas. Cancer Research,
1994. 54: p. 3845-3852.
12. Steindorf, K., Schmidt, M., and Ulurich, C., Effects of physical activity on cancer
risk and disease progression after cancer diagnosis. Bundesgesundheitsbl, 2012.
55: p. 10-16.

55
13. Zhu, Zongjian, Jian, Weiqin, Zacher, Jarrod H., Neil, Elizabeth S., McGinley,
John N., and Thompson, Henry J., Effects of Energy Restriction and Wheel
Running on Mammary Carcinogenesis and Host Systemic Factors in a Rat Model.
Cancer Prevention Research, 2012. 5: p. 414-422.
14. Food, nutrition physical activity and the prevention of cancer: a global
perspective. World Cancer research Fund/American Institute for Cancer Research,
2007: p. 30-46.
15. Romieu, Isabelle, Diet and breast cancer. Salud Publica de Mexico, 2011. 53(5):
p. 430-439.
16. Anderson, Garnet L. and Neuhouser, Marian L., Obesity and the Risk for
Premenopausal and Postmenopausal Breast Cancer. Cancer Prevention
Research, 2012. 5(3).
17. Ogundiran, Temidayo O., Huo, Dezheng, Adenipekun, Adeniyi, Campbell,
Oladapo, Oyesegun, Rasaaq, Akang, Effiong, Adebamowo, Clement, and
Olopade, Olufunmilayo I., Body fat distribution and breast cancer risk: findings
from the Nigerian breast cancer study. Cancer Causes Control, 2012.
18. Calder, Philip C., Ahluwalia, Namanjeet, Brouns, Fred, Buetler, Timo, Clement,
Karine, Cunningham, Karen, Esposito, Katherine, Jonsson, Lena S., Kolb, Hubert,
Lansink, Mirian, Marcos, Ascension, Margioris, Andrew, Matusheski, Nathan,
Nordmann, Herve, O'Brien, John, Warnberg, Julia, Watzl, Bernhard, and Roob-
Winklhofer, Brigitte M., Dietary factors and low-grade inflammation in relation
to overweight and obesity. British Journal of Nutrition, 2011. 106: p. S1-S78.
19. Baumgarten, Sarah C. and Frasor, Jonna, Inflammation: An Instigator of More
Aggressive Estrogen Receptor (ER) Positive Breast Cancers. Molecular
Endocrinology, 2012. 26(3): p. 360-371.
20. Ouchi, Noriyuki, Parker, Jennifer L., Lugus, Jesse J., and Walsh, Kenneth,
Adipokines in inflammation and metabolic disease. Nature Reviews Immunology,
2011. 11: p. 85-97.
21. Subbaramaiah, Kotha, Howe, Louise R., Bhardwaj, Priya, Du, Baoheng,
Gravaghi, Claudia, Yantiss, Rhonda K., Zhou, Xi Kathy, Blaho, Victoria A., Hla,
Timothy, Yang, Peiying, Kopelovich, Levy, Hudis, Clifford A., and Dannenberg,
Andrew J., Obesity Is Associated with Inflammation and Elevated Aromatase
Expression in the Mouse Mammary Glad. Cancer Prevention Research, 2011.
4(3): p. 329-346.
22. Morris, Patrick G., Hudis, Clifford A., Giri, Dilip, Morrow, Monica, Falcone,
Domenick J., Zhou, Xi Kathy, Du, Baoheng, Brogi, Edi, Crawford, Carolyn B.,
Kopelovich, Levy, Subbaramaiah, Kotha, and Dannenberg, Andrew J.,
Inflammation and Increase Aromatase Expression Occur in the Breast Tissue of
Obese Women in Breast Cancer. Cancer Prevention Research, 2011. 4(7): p.
1021-1029.
23. Ray, Ratna B., Raychoudhuri, Amit, Steele, Robert, and Nerurkar, Pratibha, Bitter
Melon (Momordica charantia) Extract Inhibits Breast Cancer Cell Proliferation
by Modulating Cell Cycle Regulatory Genes and Promotes Apoptosis. Cancer
Research, 2010. 70(5): p. 1925-1931.

56
24. Shehzad, Adeeb, Khan, Salman, and Lee, Young Sup, Curcumin molecular
targets in obesity and obesity-related cancers. Future Oncology, 2012. 8(2): p.
179-190.
25. Casanova, Fabiana, Quarti, J., da Costa, D.C., Ramos, C.A., da Silva, J.L., and
Flalho, E., Resveratrol chemosensitizes breast cancer cells to melphalan by cell
cycle arrest. Journal of Cellular Biochemistry, 2012.
26. Montales, Maria Theresa E., Rahal, Omar M., Kang, Jie, Rogers, Theodore J.,
Prior, Ronald L., Wu, Xianli, and Simmen, Rosalia C.M., Repression of
mammosphere formation of human breast cancer cells by soy isoflavone genistein
and blueberry polyphenolic acids suggests diet-mediated targeting of cancer
stem-like/progenitor cells. Carcinogenesis, 2011. 33(3): p. 652-660.
27. Li, Qian, Holford, Theodore R., Zhang, Yawei, Boyle, Peter, Mayne, Susan T.,
Dai, Min, and Zheng, Tongzhang, Dietary fiber intake and risk of breast cancer
by menopausal and estrogen receptor status. European Journal of Nutrition, 2012.
28. Choudhary, Shambhunath, Sood, Shilpa, Donnell, Robert L., and Wang, Hwa-
Chain R., Intervention of human breast cell carcinogenesis chronically induced
by 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine. Carcinogenesis, 2012.
0(0): p. 1-10.
29. Fu, Zhenming, Deming, Sandra L., Fair, Alecia M., Shrubsole, Martha J., Wujcik,
Debra M., Shu, Xiao-Ou, Kelley, Mark, and Zheng, Wei, Well-done meat intake
and meat-derived mutagen exposures in relation to breast cancer risk: the
Nashville Breast Health Study. Breast Cancer Research and Treatment, 2011. 129:
p. 919-928.
30. Nelson, David L. and Cox, Michael M., Principles of Biochemistry2005, New
York, NY: W.H. Freeman and Company.
31. Kleinsmith, Lewis J., Kerrigan, Donna, and Kelly, Jeanne, Understanding Cancer
and Related Topics: Understanding Estrogen Receptors, Tomaxifen, and
Raloxifene, N.C. Institute, Editor 2010.
32. Yager, James D. and Davidson, Nancy E., Estrogen Carcinogenesis in Breast
Cancer. The New England Journal of Medicine, 2006. 354: p. 270-282.
33. Yue, W., Santen, R.J., Wang, J.P., Li, Y., Verderame, M.F., Bocchinfuso, W.P.,
Korach, K.S., Devanesan, P., Todorovic, R., Rogan, E.G., and Cavalieri, E.L.,
Genotoxic metabolites of estradiol in breast: potential mechanism of estradiol
induced carcinogenesis. Journal of Steroid Biochemistry and Molecular Biology,
2003. 86(3-5): p. 477-486.
34. Clamp, A., Danson, S., and Clemons, M., Hormonal and genetic risk factors for
breast cancer. Surgeon, 2003. 1(1): p. 23-31.
35. Howe, Louise R. and Brown, Powel H., Targeting the HER/EGFR/ErbB Family
to Prevent Breast Cancer. American Association for Cancer Research, 2012.
36. Rugo, H. S., The breast cancer continuum in hormone-receptor-positive breast
cancer in postmenopausal women: evolving management options focusing on
aromatase inhibitors. European Society for Medical Oncology, 2008. 19: p. 16-
27.
37. Gucalp, Ayca and Traina, Tiffany A., Triple-Negative Breast Cancer: Adjuvant
Therapeutic Options. Chemotherapy Research and Practice, 2011. 2011.

57
38. The Use of Complementary and Alternative Medicine in the United States.
National Center for Complementary and Alternative Medicine, 2002.
39. Barnes, Patricia, Bloom, Barbara, and Nahin, Richard, Complementary and
Alternative Medicine Use Among Adults and Children: United States, 2007.
National Health Statistics Report, 2008. 12: p. 1-24.
40. Nahin, Richard, Barnes, Patricia, Stussman, Barbara J., and Bloom, Barbara,
Costs of Complementary and Alternative Medicine (CAM) and Frequency of
Visits to CAM Practitioners: United States, 2007. National Health Statistics
Report, 2009. 18.
41. Complementary and Alternative Medicine in the United States, ed. I.o.
Medicine2008, Washington, D.C.: The National Academies Press.
42. The Use of Complementary and Alternative Medicine in the United States.
National Center for Complementary and Alternative Medicine, 2008.
43. Exploring the Sciences of Complementary and Alternative Medicine. National
Center for Complementary and Alternative Medicine, 2005.
44. Jacobson, Judith, Workman, Sara B., and Kronenberg, Fredi, Research on
Complementary/Alternative Medicine for Patients With Breast Cancer: A Review
of the Biomedical Literature. Journal of Clinical Oncology, 2000. 18(3): p. 668-
683.
45. Olaku, Oluwadamilola and White, Jeffrey D., Herbal therapy use by cancer
patients: A literature review on case reports. European Journal of Cancer, 2011.
47: p. 508-514.
46. Molassiotis, A., Fernadez-Ortega, P., Pud, D., Ozden, G., Scott, J.A., Panteli, V.,
Margulies, A., Browall, M., M., Magri, Selvekerova, S., Madsen, E., Milovics, L.,
Bruyns, I., Gudmundsdottir, G., Hummerston, S., Ahmad, A.M.A., Platin, N.,
Kearney, N., and Patiraki, E., Use of complementary and alternative medicine in
cancer patients: a European survey. Annals of Oncology, 2005. 16: p. 655-663.
47. Elkins, Rita M.H., Hawaiian Noni (Morinda citrofolia)1998, Pleasant Grove, UT:
Woodland Publishing.
48. McClatchey, Will, From Polynesian Healers to Health Food Stores: Changing
Perspectives of Morinda citrifolia (Rubiaceae). Integrative Cancer Therapies,
2002. 1(2): p. 110-120.
49. Pawlus, Alison D., Kinghorn, Douglas A., Review of the ethnobotany, chemistry,
biological activity and safety of the botanical dietary supplement Morinda
citrifolia (noni). Pharmacy and Pharmacology, 2007. 59: p. 1587-1609.
50. Nelson, Scot C. and Elevitch, Craig R., Noni: The Complete Guide for
Consumers and Growers2006, Holualoa, Hawaii: Permanent Agriculture
Resources.
51. McClatchey, Will C., Diversity of Growth Gorms and Uses in the Morinda
citrifolia L. Complex, in Proceedings of the 2002 Hawai'i Noni Conference2003,
University of Hawaii Manoa: Honolulu, Hawaii.
52. Morton, J., The ocean-going noni, or Indian Mulberry (Morinda citrifolia,
Rubiaceae) and some of its "colorful" relatives. Econ Bot, 1992. 46: p. 241-256.
53. Wang, Mian-Ying , West, Brett J., Jensen, Jarakae, Nowicki, Diane, Chen, Su,
Palu, Afa K., and Anderson, Gary, Morinda citrifolia (Noni): A literature review

58
and recent advances in Noni research. Acta Pharmacol Sin, 2002. 12: p. 1127-
1141.
54. McBride, L.R., Practical Folk Medicine of Noni1975, Hilo, HI: Petroglyph Press.
55. Chun, M.N., Native Hawaii Medicines. 1994.
56. Morton, J. F., The ocean-going Noni, or Indian mulberry and some of its 'colorful'
relative. Economic Botany, 1992. 46: p. 241-256.
57. Dixon, A. R., McMillen, H., and Etkin, H.L., Ferment this: the transformation of
Noni, a traditional Polynesian medicine. Economic Botany, 1999. 53: p. 51-68.
58. McBride, L. R., Practical Folk Medicine of Hawaii1975, Hilo, Hawaii: The
Petroglyph Press.
59. Heinicke, R., The pharmacologically active ingredient of Noni. Bulletin of the
National Tropical Botanical Garden, 1985.
60. Furusawa, Eiichi, Hirazumi, Anne, Story, Stephen, and Jensen, Jarakae,
Antitumour Potential of a Polysaccharide-rich Substance from the Friut Juice of
Morinda citrifolia (Noni) on Sarcoma 180 Ascites Tumour in Mice. Phytotherapy
Research, 2003. 17: p. 1158-1164.
61. Hirazumi, Anne and Furusawa, Eiichi, An Immunomodulatory Polysaccharide-
Rich Substance from the Fruit Juice of Morinda citrifolia (Noni) with Antitumor
Activity. Phytotherapy Research, 1999. 13: p. 380-387.
62. Nualsanit, T., Rojanapanthu, P., Gritsanapan, W., Lee, S. H., Lawson, D., and
Baek, S. J., Damnacanthal, a noni component, exhibits antitumorigenic activity in
human colorectal cancer cells. J Nutr Biochem, 2011.
63. Nayak, B. S., Marshall, J. R., Isitor, G., and Adogwa, A., Hypoglycemic and
Hepatoprotective Activity of Fermented Fruit Juice of Morinda citrifolia (Noni) in
Diabetic Rats. Evid Based Complement Alternat Med, 2011. 2011: p. 875293.
64. Nayak, S. and Mengi, S., Immunostimulant activity of noni (Morinda citrifolia) on
T and B lymphocytes. Pharm Biol, 2010. 48(7): p. 724-731.
65. Youngken, H.W., A study of the root of Morinda citrifolia. Journal American
Pharmacy Association, 1958. 47: p. 162-165.
66. Atkinson, N., Antibacterial substances from flowering plants. Australian Journal
of Experimental Biology 1956. 34: p. 17-26.
67. Duncan, S.J. , Flint, H.J. , and Stewart, C.S., Inhibitory activity of gut bacteria
against Escherichia coli 0157 mediated by dietary plant metabolites. FEMS
Microbiology Letter, 1998. 164: p. 283-258.
68. Locher, C. P. , Burch, M. T. , Mower, H. F. , Berestecky, J. , Dabis, H. , B., Van
P., Lasure, A., Vanden, B. D., and Vlietinck, A. J., Anti-microbial activity and
anti-complement activity of extract obtained from selected Hawaiian medicinal
plant. Journal Ethnopharm, 1995. 49: p. 23-32.
69. Brown, Amy C., Anticancer Activity of Morinda citrofolia (Noni) Fruit: A
Review. Phytotherapy Research, 2012.
70. Chan-Blanco, Yanine, Vaillan, Fabrice, Perez, Ana Mercedes, Reynes, Max,
Brillouet, Jean-MArc, and Brat, Pierre, The noni fruit (Morinda citrofolia L.): A
review of agricultural research, nutritional and therapeutic properties. Journal of
Food Composition and Analysis, 2006. 19(2006): p. 645-654.

59
71. Mandukhail, S. U., Aziz, N., and Gilani, A. H., Studies on antidyslipidemic effects
of Morinda citrifolia (Noni) fruit, leaves and root extracts. Lipids Health Dis,
2010. 9: p. 88.
72. Dussossoy, E., Brat, P., Bony, E., Boudard, F., Poucheret, P., Mertz, C., Giaimis,
J., and Michel, A., Characterization, anti-oxidative and anti-inflammatory effects
of Costa Rican noni juice (Morinda citrifolia L.). J Ethnopharmacol, 2011.
133(1): p. 108-115.
73. Raj, R.K., Screening of indigenous plants for anthelmintic action against human
Ascaris Lmbricoids: Part-II. Indiana Journal of Physiology Pharmacology, 1975.
19: p. 47-49.
74. Hirazumi, A. and Furusawa, E., An immunomodulatory polysaccharide-rich
substance from the fruit juice of Morinda citrifolia (noni) with antitumour
activity. Phytother Res, 1999. 13(5): p. 380-387.
75. Hirazumi, A., Furusawa, E., Chou, S. C., and Hokama, Y., Immunomodulation
contributes to the anticancer activity of morinda citrifolia (noni) fruit juice. Proc
West Pharmacol Soc, 1996. 39: p. 7-9.
76. Taskin, E. I., Akgun-Dar, K., Kapucu, A., Osanc, E., Dogruman, H., Eraltan, H.,
and Ulukaya, E., Apoptosis-inducing effects of Morinda citrifolia L. and
doxorubicin on the Ehrlich ascites tumor in Balb-c mice. Cell Biochem Funct,
2009. 27(8): p. 542-546.
77. Li, J., Stickel, S. L., Bouton-Verville, H., Burgin, K. E., Yu, X., Wong, D. K.,
Wagner, T. E., and Wei, Y., Fermented Noni exudate (fNE): a mediator between
immune system and anti-tumor activity. Oncol Rep, 2008. 20(6): p. 1505-1509.
78. Hirazumi, A., Furusawa, E., Chou, S. C., and Hokama, Y., Anticancer activity of
Morinda citrifolia (noni) on intraperitoneally implanted Lewis lung carcinoma in
syngeneic mice. Proc West Pharmacol Soc, 1994. 37: p. 145-146.
79. Younos, C and Rolland, A., Analgesic and behavioural effect of Morinda
citrifolia. Planta Med, 1990. 56: p. 430-434.
80. Pachauri, S. D., Tota, S., Khandelwal, K., Verma, P. R., Nath, C., Hanif, K.,
Shukla, R., Saxena, J. K., and Dwivedi, A. K., Protective effect of fruits of
Morinda citrifolia L. on scopolamine induced memory impairment in mice: a
behavioral, biochemical and cerebral blood flow study. J Ethnopharmacol, 2012.
139(1): p. 34-41.
81. Nerurkar, P. V. , Nishioka, A. , Eck, P. O. , Johns, L. M. , Volper, E. , and
Nerurkar, V. R., Regulation of glucose metabolism via hepatic forkhead
transcription factor 1 (Fox01) by Morinda citrifolia (noni) in high-fat diet-
induced obese mice. British Journal of Nutrition, 2011. 20: p. 1-11.
82. Mahattanadul, S., Ridtitid, W., Nima, S., Phdoongsombut, N., Ratanasuwon, P.,
and Kasiwong, S., Effects of Morinda citrifolia aqueous fruit extract and its
biomarker scopoletin on reflux esophagitis and gastric ulcer in rats. J
Ethnopharmacol, 2011. 134(2): p. 243-250.
83. Prapaitrakool, S. and Itharat, A., Morinda citrifolia Linn. for prevention of
postoperative nausea and vomiting. J Med Assoc Thai, 2010. 93 Suppl 7: p.
S204-209.

60
84. Singh, D.R. , Singh, S. , and Salim, K.M., Estimation os phytochemicals and
antioxidant activity of underutilized fruits of Andaman Islands (India). Int J Food
Sci Nutr, 2011.
85. Jang, B.C., The fruit juice of Morinda citrifolia (noni) downregulates HIF-1a
protein expression through inhibition of PKB, ERK-1/2, JNK-1 and S6 in
manganese-stimulated A549 human lung cancer cells. Int J Mol Med, 2012.
29(3): p. 499-504.
86. Brito, D. R., Fernandes, R. M., Fernandes, M. Z., Ferreira, M. D., Rolim, F. R.,
and da Silva Filho, M. L., Anthelmintic activity of aqueous and ethanolic extracts
of Morinda citrifolia fruit on Ascaridia galli. Rev Bras Parasitol Vet, 2009. 18(4):
p. 32-36.
87. Muto, J., Hosung, L., Uwaya, A., Isami, F., Ohno, M., and Mikami, T., Morinda
citrifolia fruit reduces stress-induced impairment of cognitive function
accompanied by vasculature improvement in mice. Physiol Behav, 2010. 101(2):
p. 211-217.
88. Harada, S., Hamabe, W., Kamiya, K., Satake, T., Yamamoto, J., and Tokuyama,
S., Preventive effect of Morinda citrifolia fruit juice on neuronal damage induced
by focal ischemia. Biol Pharm Bull, 2009. 32(3): p. 405-409.
89. Okusada, K., Nakamoto, K., Nishida, M., Fujita-Hamabe, W., Kamiya, K.,
Mizushina, Y., Satake, T., and Tokuyama, S., The antinociceptive and anti-
inflammatory action of the CHCl3-soluble phase and its main active component,
damnacanthal, isolated from the root of Morinda citrifolia. Biol Pharm Bull,
2011. 34(1): p. 103-107.
90. Liu, Q. , Kim, S. B., Ahn, J. H., Hwang, B. Y., Kim, S. Y., and Lee, M. K.,
Anthraquinones from Morinda citrifolia roots enhance adipocyte differentiation
in 3T3-L1 cells. Nat Prod Res, 2011.
91. Masuda, M., Itoh, K., Murata, K., Naruto, S., Uwaya, A., Isami, F., and Matsuda,
H., Inhibitory Effects of Morinda citrifolia Extract and Its Constituents on
Melanogenesis in Murine B16 Melanoma Cells. Biol Pharm Bull, 2012. 35(1): p.
78-83.
92. Lishuang, L. V., Chen, H. , and Ho, C., Chemical components of the roots of Noni
(Morinda citrifolia) an their cytotoxic effects. Fitoterapia, 2011. 82: p. 704-708.
93. Pazos, D. C., Jimenez, F. E., Garduno, L., Lopez, V. E., and Cruz, M. C.,
Hypolipidemic effect of seed oil of noni (Morinda citrifolia). Nat Prod Commun,
2011. 6(7): p. 1005-1008.
94. Nelson, Scot C.The Noni Website. 2006 [cited 2012 April 3].
95. Wang, Cheng-Hsin, Lai, Phoency, Chen, Mei-En, and Chen, Hsiao-Lin,
Antioxidative capacity produced by Bifidobacterium- and Lactobacillus
acidophilus-mediated fermentations of konjac glucomannan and glucomannan
oligosaccharides. Journal of Science of Food and Agriculture, 2008. 88(7): p.
1294-1300.
96. Kaikainahaole, M., Hawaiian uses of herbs - past and present. Hawaiian
Botanical Society, 1968. 7(5): p. 31-38.
97. Handy, E.S.C., Pukui, M.K., and Livermore, K., Outline of Hawaiian physical
therapeutics. Bernice P. Bishop Museum Bulletin, 1934. 126: p. 51.

61
98. Figueroa-Gonzalez, Ivonne, Quijano, Guillermo, Ramirez, Gerardo, and Cruz-
Guerroro, Alma, Probiotics and prebiotics - perspectives and challenges. Journal
of Science of Food and Agriculture, 2011. 91: p. 1341-1348.
99. Heller, Knut J., Probiotic bacteria in fermented foods: product characteristics
and starter organisms. American Journal Clinical of Nutrition, 2001. 73: p. 374S-
379S.
100. Wang, Chung-Yi, Ng, Chang-Chai, Su, Hsuan, Tzeng, Wen-Sheng, and Shyu,
Yuan-Tay, Probiotic potential of noni juice fermented with lactic acid bacteria
and bifidobacteria. International Journal of Food Sciences and Nutrition, 2009.
60(S6): p. 98-106.
101. Yang, Shu-Chuan, Chen, Tsu-I, Li, Ken-Yuon, and Tsai, Tsun-Chung, Change in
Phenolic Compound Content, Reductive Capacity and ACE Inhibitory Activity in
noni Juice during Traditional Fermentation. Journal of Food and Drug Analysis,
2007. 15(3): p. 290-298.
102. Arpornsuwan, Teerakul and Tadsanee, Punjanon, Tumor cell-selective
antiproliferative effect of the extract from Morinda citrifolia fruits. Phytotherapy
Research, 2006. 20(6): p. 515-517.
103. Hornick, Conrad A., Myers, Amy, Sadowska-Krowicka, Halina, Anthony,
Catherine T., and Woltering, Eugene A., Inhibition of angiogenic initiation and
disruption of newly established human vascular networks by juice from Morinda
citrifolia (noni). Angiogenesis, 2003. 6(2): p. 143-149.
104. Hiramatsu, Tomonori, Imoto, Masaya, Koyano, Takashi, and Umezawa, Kazuo,
Induction of normal phenotypes in ras-transformed cells by damnacanthal from
Morinda citrifolia. Cancer Letters, 1993. 73(2-3): p. 161-166.
105. Nualsanit, Thararat, Rojanapanthu, Pleumchitt, Gritsanapan, Wandee, Lee,
Seong-Ho, Lawson, Darunee, and Baek, Seung Joon, Damnacanthal, a noni
component, exhibits antitumorigenic activity in human colorectal cancer cells.
Journal of Nutritional Biochemistry, 2011.
106. Liu, Guangmin, Bode, Anne, Ma, Wei-Ya, Sang, Shengmin, Ho, Chi-Tang, and
Dong, Zigang, Two Novel Glycosides from the Fruits of Morinda Citrifolia (Noni)
Inhibit AP-1 Transactivation and Cell Transformation in the Mouse Epidermal.
Cancer Research, 2001. 61(5749-5756).
107. Takashima, Junko, Ikeda, Yoshitaka, Komiyama, Kanki, Hayashi, Masahiko,
Kishida, Akio, and Ohsaki, Ayumi, New Constituents from the Leaves of Morinda
citrifolia. Chemical and Pharmaceutical Bulletin, 2007. 55(2): p. 343-345.
108. Li, Jinhua, Stickel, Sara L., Bouton-Verville, Hilary, Burgin, Kelly E., Yu,
Xianzhong, Wong, Desmond K.W., Wagner, Thomas E., and Wei, Yanzhang,
Fermented Noni Exudate (fNE): A mediator between immune system and anti-
tumor activity. Oncology Reports, 2008. 20: p. 1505-1509.
109. Stoner, Gary D., Wang, Li-Shu, Seguin, Claire, Rocha, Claudio, Stoner, Kristen,
Chiu, Steven, and Kinghorn, A. Douglas, Multiple Berry Types Prevent N-
nitrosomethylbenzylamineInduced Esophageal Cancer in Rats. Pharmacology
Research, 2010. 27: p. 1138-1145.
110. Jang, Byeong-Churl, The fruit juice of Morinda citrifolia (noni) downregulates
HIF-1α protein expression through inhibition of PKB, ERK-1/2, JNK-1 and S6 in

62
manganese-stimulated A549 human lung cancer cells. International Journal of
Molecular Medicine, 2012. 29(3).
111. West, Brett J., White, Leland D., Jensen, Jarakae, and Palu, 'Afa K., A double-
blind clinical safety study of noni fruit juice. Pacific Health Dialog, 2009. 15(2):
p. 21-32.
112. Hengartner, Michael O., The biochemistry of apoptosis. Nature 2000. 407: p. 770-
776.
113. Fulda, S. and Debatian, K.M., Extrinsic versus intrinsic apoptosis pathways in
anticancer chemotherapy. Oncogene, 2006. 25: p. 4798-4811.
114. Bender, Cheryl E., Fitzgerald, Patrick, Tait, Stephen W.G., Llambi, Fabien,
McStay, Gavin P., Tupper, Douglas O., Pellettieri, Jason, Alvarado, Alejandro
Sanchez, Salvesen, Guy S., and Green, Douglas R., Mitochondrial pathway of
apoptosis is ancestral in metazoans. Cell Biology, 2012: p. 1-6.
115. Luo, Xu, Budihardojo, Imawati, Zou, Hua, Slaughter, Clive, Wang, Xiaodong,
and Hughes, Howard, Bid, a Bcl2 interacting protein, mediates cytochrome c
release from mitochondria in response to activation of cell surface death
receptors. Cell, 1998. 94: p. 481-490.
116. Lupardus, Patrick J. and Cimprich, Karlene A., Checkpoint Adaptation:
Molecular Mechanisms Uncovered. Cell, 2004. 117: p. 555-559.
117. Hanahan, Douglas and Weinberg, Robert A., Hallmarks of Cancer: The Next
Generation. Cell 2011. 144(5): p. 646-674.
118. Deveraux, Quinn L. and Reed, John C., IAP family proteins- suppressors of
apoptosis Genes & Development, 1999. 13: p. 239-252.
119. Fulda, Simone and Vuviv, Domagoj, Targeting IAP proteins for therapeutic
intervention in cancer. Nature Reviews Drug Discovery, 2012. 11(109-123).
120. Shimizu, Yoshiko, Luk, Hugh, Horio, David, Miron, Penelope, Griswold,
Michael, Iglehart, Dirk, Hernandez, Brenda, Killeen, Jeffrey, and Elshamy, Wael
M., BRCA1-IRIS Overexpression Promotes Formation of Aggressive Breast
Cancers. PLoS One, 2012. 7(4).
121. Ambrosini, Grazia, Adida, Collette, Sirugo, Giorgio, and Altieri, Dario C.,
Induction of apoptosis and inhibition of cell proliferation by survivin gene
targeting. The Journal of Biological Chemistry, 1998. 273: p. 11177-11182.
122. Delivani, P. and Martin, SJ, Mitochondrial membrane remodeling in apoptosis:
an inside story. Cell Death and Differentiation, 2006. 13: p. 2007-2010.
123. Kluck, Ruth M. and Bossy-Wetzel, Ella, The release of cytochrome c from
mitochrondria: a primary site for Bcl-2 regulation of apoptosis. Science, 1997.
275(5303).
124. Aggarwal, Bharat B., Nuclear factor-kB: the enemy within. Cancer Cell, 2006. 6:
p. 203-208.
125. Jeyasuria, Pancharatnam, Subedi, Kalpana, Suresh, Arvind, and Condon, Jennifer
C., Elevated levels of uterine anti-apoptotic signaling may activate NfkB and
potentially confer resistance to caspase 3-mediated apoptotic cell death during
pregnancy in mice. Biology of Reproduction, 2011. 85: p. 417-424.
126. Wang, Cun-Yu, Cusack, James C., Liu, Rong, and Baldwin, Albert S. Jr., Control
of inducible chemoresistance: enhanced anti-tumor therapy through increased

63
apoptosis by inhibition of NF-kB. Emergining Microbes & Infections, 1999. 5: p.
412-417.
127. Chandel, Navdeep S., Trzyna, Wendy C., McClintock, David S., and Schumacker,
Paul T., Role of Oxidants in NF-κB Activation and TNF-α Gene Transcription
Induced by Hypoxia and Endotoxin. Journal of Immunology, 2000. 165(2): p.
1013-1021.
128. Fitzgerald, D.C., Meade, K.G., McEvoy, A.N., Lillis, L., Murphy, E.P.,
MacHugh, D.E., and Baird, A.W., Tumour necrosis factor-α (TNF-α) increases
nuclear factor κB (NFκB) activity in and interleukin-8 (IL-8) release from bovine
mammary epithelial cells. Veterinary Immunology and Immunopathology, 2006.
116(1-2): p. 59-68.

64

You might also like