224
The Role of Grapefruit Consumption in Cardiometabolic Health in Overweight and Obese Adults Item Type text; Electronic Dissertation Authors Dow, Caitlin Ann Publisher The University of Arizona. Rights Copyright © is held by the author. Digital access to this material is made possible by the University Libraries, University of Arizona. Further transmission, reproduction or presentation (such as public display or performance) of protected items is prohibited except with permission of the author. Download date 06/01/2022 20:07:19 Link to Item http://hdl.handle.net/10150/293398

THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

The Role of Grapefruit Consumption in CardiometabolicHealth in Overweight and Obese Adults

Item Type text; Electronic Dissertation

Authors Dow, Caitlin Ann

Publisher The University of Arizona.

Rights Copyright © is held by the author. Digital access to this materialis made possible by the University Libraries, University of Arizona.Further transmission, reproduction or presentation (such aspublic display or performance) of protected items is prohibitedexcept with permission of the author.

Download date 06/01/2022 20:07:19

Link to Item http://hdl.handle.net/10150/293398

Page 2: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC

HEALTH IN OVERWEIGHT AND OBESE ADULTS

by

Caitlin Dow, M.S.

A Dissertation Submitted to the Faculty of the

DEPARTMENT OF NUTRITIONAL SCIENCES

In Partial Fulfillment of the Requirements

For the Degree of

DOCTOR OF PHILOSOPHY

In the Graduate College

THE UNIVERSITY OF ARIZONA

2013

Page 3: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

2

THE UNIVERSITY OF ARIZONA

GRADUATE COLLEGE

As members of the Dissertation Committee, we certify that we have read the dissertation

prepared by Caitlin Dow, titled The Role of Grapefruit Consumption in Cardiometabolic Health

in Overweight and Obese Adults and recommend that it be accepted as fulfilling the dissertation

requirement for the Degree of Doctor of Philosophy.

4/16/13

Cynthia Thomson, PhD, RD

4/16/13

Scott Going, PhD

4/16/13

Hsiao-Hui (Sherry) Chow, PhD

4/16/13

Zoe Cohen, PhD

4/16/13

Patricia Thompson, PhD

Final approval and acceptance of this dissertation is contingent upon the candidate’s submission

of the final copies of the dissertation to the Graduate College.

I hereby certify that I have read this dissertation prepared under my direction and recommend

that it be accepted as fulfilling the dissertation requirement.

4/16/13

Dissertation Director: Cynthia Thomson, PhD, RD

Page 4: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

3

STATEMENT BY AUTHOR

This dissertation has been submitted in partial fulfillment of requirements for an

advanced degree at The University of Arizona and is deposited in the University Library

to be made available to borrowers under rules of the Library.

Brief quotations from this dissertation are allowable without special permission, provided

that accurate acknowledgement of source is made. Requests for permission for extended

quotation from or reproduction of this manuscript in whole or in part may be granted by

the head of the major department or the Dean of the Graduate College when in his or her

judgment the proposed use of the material is in the interests of scholarship. In all other

instances, however, permission must be obtained from the author.

SIGNED: Caitlin Dow

Page 5: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

4

ACKNOWLEDGEMENTS

The author would like to acknowledge the following groups for their role in making this

research possible.

The funding sources for this study:

o Vegetable and Fruit Improvement Center at Texas A&M and the United

States Department of Agriculture Cooperative State, Research, Education

and Extension Service (USDA-CSREES) (2010-34402-20875)

o United States Department of Agriculture National Needs Fellowship in

Obesity Research (USDA-NNF) (2010-38420-20369)

Rio Queen Citrus Inc. (Mission, TX) for their generous grapefruit donation.

Bayer Healthcare (Morristown, NJ) for their generous multivitamin donation.

Jennifer Ravia, Amy Butalla, Lindsey Diemert, and Julie West for their role in

coordination of the study.

Christina Preece for her work in biosample analysis.

Page 6: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

5

TABLE OF CONTENTS

LIST OF FIGURES…………………………………………………………………8

LIST OF TABLES…………………………………………………………………..9

ABSTRACT………………………………………………………………………...10

CHAPTER 1: INTRODUCTION.……………………………….…………………12

Figure………………………………………………………………………..22

CHAPTER 2: THE EFFECTS OF DAILY CONSUMPTION OF GRAPEFRUIT

ON BODY WEIGHT, LIPIDS, AND BLOOD PRESSURE IN HEALTHY,

OVERWEIGHT ADULTS..............………………………………………….…….23

Introduction………………………………………………………………....23

Methods……………………………………………………………………..26

Results………………………………………………………………………31

Discussion…………………………………………………………………..34

Figures………………………………………………………………………42

Tables……………………………………………………………………….43

CHAPTER 3: DAILY CONSUMPTION OF GRAPEFRUIT FOR SIX WEEKS

DOES NOT REDUCE SVCAM-1, CRP, OR F2-ISOPROSTANES IN

OVERWEIGHT AND OBESE ADULTS………………………..…...……………47

Introduction………………………………………………………………....47

Methods……………………………………………………………………..49

Results………………………………………………………………………52

Discussion…………………………………………………………………..54

Page 7: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

6

TABLE OF CONTENTS – Continued

Tables……………………………………………………………………….59

CHAPTER 4: GRAPEFRUIT INCREASES POSTPRANDIAL INSULIN AND

ATTENUATES F2-ISOPROSTANE AND IL-6 CONCENTRATIONS WHEN

CONSUMED WITH A HIGH-FAT, HIGH-CALORE MEAL.................................62

Introduction………………………………………………………………....62

Methods……………………………………………………………………..64

Results………………………………………………………………………67

Discussion…………………………………………………………………..69

Figures………………………………………………………………………75

Tables……………………………………………………………………….76

CHAPTER 5: Summary and Conclusions………………………………………….78

Obesity and Visceral Adiposity…………………………………………….78

Vascular Reactivity and Blood Pressure……………………………………80

Lipid Response………………………………………….…………………..83

Insulin Response………………………………………….………………...84

Inflammation and Oxidative Stress Modification…………………………..88

Limitations, Future Directions, and Conclusions…………………………..93

Figure……………………………………………………………………….97

APPENDIX A CONSENT FORM…………………………………………………98

APPENDIX B SUPPLEMENTAL MATERIALS

Grapefruit Weights…………………………………………………………106

Page 8: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

7

TABLE OF CONTENTS - Continued

Compliance Logs………………...………………………………………....107

APPENDIX C- ADDITIONAL PUBLICATION #1: A review of evidence based

strategies to treat obesity in adults………………...…………………….......108

APPENDIX D- ADDITIONAL PUBLICATION #2: Associations between

mother’s BMI, fruit and vegetable intake and availability, and child’s body

shape as reported by women responding to an annual survey………..……..144

APPENDIX E- ADDITIONAL PUBLICATION #3: Predictors of

cardiometabolic risk factor improvements with weight loss in women...…..166

APPENDIX F- ADDITIONAL PUBLICATION #4: Use of herbal, botanical, and

natural product supplements in oncology and diabetes populations……..…194

REFERENCES……………………………………………………………………...214

Page 9: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

8

LIST OF FIGURES

FIGURE 1. Pathophysiology of Atherosclerotic Plaque Production and Targets

for Risk Modification …………………………………………………….…22

FIGURE 2. Consort diagram describing the flow of participants through the

grapefruit intervention feeding trial.….……………………………………..42

FIGURE 3. Concentrations of various metabolic, inflammatory, and oxidative

stress markers following consumption of a HFHC meal alone or with ruby

red grapefruit……………………………………………………………….76

FIGURE 4. The Effect of Grapefruit on Pathophysiology of Atherosclerotic

Plaque Production and Targets for Risk Modification……………………...97

Page 10: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

9

LIST OF TABLES

TABLE 1. Baseline characteristics of participants who completed the grapefruit

feeding trial: grapefruit vs. control ……………………………………….……43

TABLE 2. Anthropometric and blood pressure changes in participants who

completed the grapefruit feeding trial: grapefruit vs control.…...………...……44

TABLE 3. Comparison of diet composition in participants who completed the

grapefruit feeding trial: grapefruit vs control……………………………..……45

TABLE 4. Comparison of the lipid profile in those participants who completed

the Grapefruit Feeding Trial: Grapefruit vs Control, before and after the

intervention phase.……………………………………………………………….……46

TABLE 5. Baseline characteristics of participants who successfully completed

all study activities (n=69) ……………………………………………....…….59

TABLE 6. Change in biomarkers (mean ± SD) associated with atherosclerotic

plaque production in participants enrolled in a six-week grapefruit feeding

trial..…………………………………………………………………….…….60

TABLE 7. Associations between common cardiovascular disease (CVD) risk

factors and inflammatory/oxidative stress biomarkers in participants

enrolled in a grapefruit feeding trial (n=69)…………………………….……61

TABLE 8. Baseline characteristics of participants who completed the

grapefruit double meal challenge postprandial trial....……………..…..……77

TABLE 9. Comparison of AUC of biomarkers in response to a HFHC meal

eaten alone or with grapefruit (n=10)…………………………………..……77

Page 11: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

10

ABSTRACT

Atherosclerotic cardiovascular diseases (CVD) are the leading cause of death and often

develop due to obesity-induced complications including hyperlipidemia, elevated blood

pressure (BP), inflammation, and oxidative stress. Epidemiological, animal model, and

cell culture studies indicate that citrus, and grapefruit specifically, exert cardiovascular

health benefits, likely due to the high flavonoid content in citrus fruits. Grapefruit and/or

isolated grapefruit flavonoids elicit cardiovascular benefits via improvements in lipid

metabolism and endothelial reactivity, and by antioxidant and anti-inflammatory actions.

The aim of this work was to determine the role of six-week daily consumption of

grapefruit on weight, lipid, and BP control as well as inflammatory and oxidative stress

markers in overweight/obese adults. Further, we sought to evaluate the acute,

postprandial effects of grapefruit consumption on metabolic, inflammatory, and oxidative

stress markers in response to a high fat, high calorie (HFHC) double meal challenge.

Participants were randomized to either a grapefruit group (n=42) in which they consumed

1.5 grapefruit/day for six weeks or to a control condition (n=32). Ten participants who

completed the feeding trial also participated in the postprandial study. On two test days

participants consumed a HFHC meal for breakfast and again for lunch. A ruby red

grapefruit was consumed with breakfast on the first test day. Blood samples were

collected at baseline and for the subsequent eight hours on each day. In the feeding trial,

grapefruit consumption resulted in reductions in waist circumference (p<0.001), systolic

BP (p=0.03), total cholesterol (p=0.001), and LDL-cholesterol (p=0.021) compared to

baseline values. F2-isoprostanes and hsCRP values were nonsignificantly lower in the

Page 12: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

11

grapefruit vs. control arm following the intervention (p=0.063 and p=0.073, respectively).

In the postprandial evaluation, insulin concentrations were significantly higher 30

minutes (p=0.007) and 2 hours (p=0.025) post HFHC + grapefruit meal consumption vs.

HFHC alone. HFHC + grapefruit intake resulted in lower IL-6 concentrations after two

hours (p=0.017) and lower F2-isoprostanes after 5 hours (p=0.0125). These findings

suggest that regular grapefruit consumption may reduce CVD risk by targeting many of

the risk factors and pathogenic factors involved in endothelial dysfunction. However,

this dietary change alone is unlikely to result in significant CVD risk reduction.

Page 13: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

12

CHAPTER 1

INTRODUCTION

Atherosclerotic cardiovascular diseases (CVD) are the leading cause of death in

the developed and developing world [1, 2], affecting over 79.4 million people in the US

alone [3]. Obesity also poses a significant public health threat; according to the most

recent National Center for Health Statistics report, 69.2% of the adult population in the

United States is overweight, and 35.7% are obese [4]. The link between obesity and

cardiometabolic complications that often result in atherosclerotic plaque formation is

well-characterized [2], and obesity is considered one of the primary modifiable risk

factors in the CVD risk profile [5]. Furthermore, obesity increases the risk of morbidity

and mortality from diseases of endothelial damage including hypertension, coronary

artery disease (CAD), and stroke [6]. Perhaps even more important than the role of

obesity in vascular disease pathogenesis, is the role of central obesity, characterized by an

increased volume of visceral adipose tissue.

Visceral adipose tissue (VAT) functions as a metabolic reservoir that becomes

highly dysregulated with increasing volume and is involved in the pathophysiology of

atherogenesis [2, 7]. Numerous reports indicate that elevated waist circumference, a

surrogate measure of visceral adiposity, is associated with an increased risk of CAD [8-

10]. VAT is pathogenically linked to metabolic issues that impact vascular function

including insulin resistance and secretion of excess triglycerides [7].

Page 14: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

13

Several studies have shown positive associations between waist circumference

and circulating markers of inflammation and oxidative stress, including interleukin-6 (IL-

6), C-reactive protein (CRP), and F2-isoprostanes [11-13]. Evidence also indicates that

VAT is not only associated with circulating atherogenic molecules, but also directly

secretes inflammatory cytokines, growth factors, and vasoactive proteins which are

known to interact with the endothelium and promote atheroma development [12].

Additionally, an analysis of various fat depots in obese women suggested that adhesion

molecule expression and secretion is elevated in the vasculature of VAT [14], thus

providing more evidence for an innate link between endothelial dysfunction and visceral

adiposity.

These vasoactive proteins secreted from VAT interact with the endothelium, a

monolayer of cells that represents the interface between the blood and the vessel wall that

plays a critical role in maintaining vascular homeostasis. The endothelium is highly

reactive to the blood and its constituents and transmits signals to the underlying smooth

muscle cells, leading to activation of pathways involved in vasoreactivity [6]. Injury to

the endothelium results in endothelial dysfunction (ED), the first abnormality in the

process leading to plaque formation [2, 15, 16].

Risk factors for ED and atheroma development include dyslipidemia,

hyperglycemia, insulin resistance, chronic low-grade inflammation, and oxidative stress

[2, 15, 17], biological responses to stimuli such as diet, activity, infection, and

adiposity/adipokines. Collectively, these factors interact with the endothelium, leading to

endothelial cell activation, adhesion molecule expression, white blood cell recruitment, a

Page 15: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

14

hyper-inflammatory response, and eventually, atheroma development [6] (Figure 1).

Over time and with accumulating damage, ischemic events are promoted causing

significant endothelial damage. Furthermore, these risk factors which are cumulatively

damaging to the vasculature typically cluster together as a result of obesity [2, 6, 15].

Given the irrefutable link between obesity and CVD, and the impact of VAT on

endothelial function, strategies that promote healthy weight and cardiovascular health,

particularly in the primary prevention setting, are warranted. Ideally, these strategies

would promote normal lipid values, while simultaneously reducing inflammation and

oxidative stress.

Evidence indicates that citrus, and grapefruit in particular, may be a beneficial

addition to the diet for weight, lipid, and blood pressure control [18-21]. These effects of

citrus are likely attributable to fruit-specific bioactive compounds including the

flavonoids, naringin and hesperidin, which are converted to the aglycones, naringenin and

hesperitin, in the gut [22]. Theses flavonones (a subclass of flavonoids specific to citrus

fruits) are known to have antioxidant, hypolipidemic, and anti-hypertensive properties

[20, 23], suggesting a possible role for these compounds in the prevention of

cardiovascular disease.

A retrospective analysis of the Nurse’s Health Study (NHS) and Health

Professional’s Follow-up Study provides support for the role of citrus in CVD risk

reduction. This analysis of 84,251 women and 42,148 men showed a 19% lower risk of

ischemic stroke in individuals who consumed the highest quintile versus the lowest

quintile of citrus fruits (RR: 0.81, 95% CI: 0.68-0.96) and a 25% lower risk for those

Page 16: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

15

consuming citrus juices (RR: 0.75, 95% CI: 0.62-0.99) [24]. Similar risk lowering

associations were demonstrated in regard to myocardial infarction in 8,087 50-59 year

old men in Northern Ireland and France in the Prospective Epidemiological Study of

Myocardial Infarction (PRIME) study [25]. Additionally, an analysis of more than

34,400 postmenopausal women from the Iowa Women’s Health Study with 16 years of

follow-up suggested that citrus flavonones, quantified using data collected from a Food

Frequency Questionnaire and three USDA databases, were associated with a reduced risk

of total mortality, CAD, and CVD. Grapefruit intake of one serving or more per week

was associated with a 23% and 15% reduced risk of CAD and overall CVD, respectively

[26].

Grapefruit made its first appearance in the health market in the 1930’s as a “fat-

burning” food as part of the 800 to 1000 kcal Hollywood Diet [27]. While dieters

experienced weight loss, it is reasonable to assume that this effect was attributable to the

low energy composition of the diet, as further analyses of grapefruit have revealed that

the fruit has no thermogenic properties [28, 29].

While folklore promotes grapefruit as a weight loss food, clinical trials testing this

effect are limited and results have been inconsistent. A randomized, controlled study in

91 obese subjects by Fujioka, et al. evaluated weight change in participants assigned to

fresh grapefruit, grapefruit juice, a grapefruit capsule, or a placebo treatment. Consuming

fresh grapefruit over 12 weeks resulted in a 1.6 kg weight reduction as compared to a 1

kg weight loss in those randomized to the grapefruit juice or grapefruit capsule, while

those randomized to the placebo arm demonstrated no change in weight [19]. Some

Page 17: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

16

evidence indicates that a food with high fiber/water content consumed before a meal may

induce satiety and reduce caloric intake at the meal [30]. A randomized study by Silver,

et al. investigated the role of grapefruit in weight loss comparing whole grapefruit,

grapefruit juice, or water as meal preloads in addition to 12.5% energy restriction in 85

obese adults. Significant weight loss was observed in all groups as compared to baseline

weight, though no significant difference between the three groups was observed. Further,

energy intake did not differ by preload assignment (water, grapefruit, grapefruit juice),

suggesting that the whole fruit had no additional effects above water or juice in reducing

energy intake [18]. It is possible that these trials may not have had adequate sample sizes

to observe statistically significant weight loss because of the 3-4 arm study designs.

Despite the inconclusive findings regarding grapefruit and weight control,

research does suggest that grapefruit may exert cardiovascular benefits through

modification of other risk factors. Cell culture and animal models support the role of

citrus and grapefruit bioactive compounds in modifying lipid metabolism. In particular,

naringenin has been shown to elicit its hypolipidemic actions by inducing hepatic

peroxisome proliferator-activated receptor (PPAR)-α and –γ expression in rats and

cultured human hepatocytes, thus improving lipid metabolism and fatty acid oxidation

[20, 31]. An in vitro study in HepG2 cells incubated with naringenin or hesperitin

resulted in reduced secretion of ApoB, the primary apoprotein in chylomicrons and low-

density lipoproteins (LDL), and upregulation of LDL receptor (LDL-R) transcription

[32]. To illustrate the bioactivity of a citrus flavonone using a rodent model system, rat

chow was supplemented with naringenin for six weeks at intake levels comparable to 1-4

Page 18: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

17

cups of grapefruit juice per day in humans. Male Long-Evans hooded rats demonstrated

significantly reduced plasma and adipose tissue triglycerides and total plasma cholesterol

concentrations, as well as reductions in parametrial adipose tissue weight [20]. Further,

3% naringenin supplementation of a high cholesterol diet for 12 weeks resulted in

reductions in hepatic steatosis, VLDL overproduction, hyperlipidemia, atherosclerotic

lesion development in LDL-R-/- C57BL/6J mice [33]. These data from animal models

support the hypolipidemic properties of naringenin, though evidence from human studies

is conflicting.

The only human study to date to measure lipid changes in response to grapefruit

consumption as a primary outcome was a study of 57 hyperlipidemic patients who had

recently undergone coronary bypass surgery. After 30 days of daily intake, grapefruit

consumption resulted in significant reductions in total cholesterol, LDL-cholesterol, and

triglycerides in patients with coronary atherosclerosis (p<0.05 for all outcomes) [34].

However, 204 moderately hypercholesterolemic adults consuming either a naringin or a

hesperedin supplement at a dose of 500 mg and 800 mg, respectively, for four weeks

experienced no changes in the lipid profile [35]. These results are not consistent with the

rat study, but suggest that the whole fruit may be required to produce appreciable changes

in lipid parameters. Given the availability of grapefruit crop in the US and the potential

risk-reducing effects of regular consumption there is a need for further research in order

to elucidate the true effects of grapefruit on lipid control, as well as other factors

associated with CVD risk, including blood pressure, inflammation, and oxidative stress.

Page 19: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

18

Studies on the direct vascular effects of citrus are limited, though promising. Ex

vivo studies of aortic tissue indicate that citrus flavonones induce vasorelaxation due to

inhibition of phosphodiesterases [21] and activation of endothelial nitric oxide synthase

(eNOS), thus inducing nitric oxide (NO) production [36]. Five-week consumption of

sweetie fruit (a hybrid of grapefruit and pummelo) juice resulted in a significant

reduction in diastolic blood pressure of 3.7 mmHg in 12 hypertensive men [37]. In a

randomized crossover study, 24 men consumed 500 mL of orange juice, a beverage with

added hesperidin, or a placebo beverage for four weeks each. Diastolic blood pressure

was lower by 3.2 mmHg and 5.5 mmHg after the hesperidin beverage or orange juice,

respectively, compared to the placebo beverage [38]. Further, a postprandial analysis of

each treatment revealed improved skin blood flow, a measure of microvascular reactivity,

after orange juice and hesperidin beverage intake compared to placebo [38].

Additionally, hesperitin treatment of bovine aortic endothelial cells (BAEC) stimulated

NO production, whereas 3 weeks of 500 mg hesperidin supplementation resulted in

improved flow mediated dilation, a measure of endothelial reactivity, compared to a

placebo treatment in 24 participants with the Metabolic Syndrome [36].

In addition to the effects of grapefruit on classic risk factors of CVD (weight loss,

lipid control, blood pressure), preliminary evidence suggests that citrus/grapefruit may

also play a role in reducing risk of CVD due to effects on novel risk factors including

inflammation and oxidative stress. Given the known roles of flavonoids in the

cardiovascular system, a cross-sectional analysis was performed to test the association

between intake of overall and specific flavonoid subgroups (flavonols, flavones,

Page 20: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

19

flavonones, flavan-3-ols, anthocyanidins, and polymeric flavonoids) and biomarkers

associated with endothelial health (CRP, IL-6, IL-18, soluble TNF Receptor-2, soluble

vascular cellular adhesion molecule-1, soluble intercellular adhesion molecule-1, and E-

selectin) using the NHS prospective cohort of 2,115 women aged 43-70 [39]. In this

analysis, greater intake of grapefruit flavonones was associated with lower circulating

concentrations of CRP and soluble tumor necrosis factor-receptor 2 (sTNF-R2) [39]. A

recent evaluation of consumption of orange and blackcurrant juices in individuals with

peripheral artery disease resulted in reductions in CRP and F2-isoprostanes compared to a

control condition [40]. In vitro work also indicates that LDL-bound naringenin reduces

oxidation and glycation of LDL [41], thus potentially reducing the pro-atherogenic

properties of oxidized LDL.

When evaluating a dietary intervention in the context of CVD health, it is ideal to

determine both the acute and chronic effects. Atherosclerotic plaques develop over

decades [1]; however, it is also well recognized that endothelial activation and

subsequent plaque development are the products of metabolic, oxidative, and

inflammatory perturbations that arise during the postprandial period [42-44]. Indeed,

some data suggest that evaluation of fasting values of various CVD risk factors alone

does not predict future events and that study of the postprandial period provides a more

comprehensive and accurate risk profile [45-47]. Previous postprandial research suggests

that consuming antioxidant rich foods with a high fat, high calorie (HFHC) meal

attenuates the oxidative/inflammatory responses induced by the meal alone [48-51].

Only a few studies have evaluated the postprandial effects of citrus consumption, and the

Page 21: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

20

exposures were either orange juice and/or a beverage containing hesperidin [38, 49, 52].

This is despite the epidemiological data that suggest that grapefruit has a more

pronounced effect on markers of endothelial health than orange juice [26, 39].

Nevertheless, these trials indicate that orange juice consumption reduces the pro-

inflammatory effects induced by a HFHC meal [49], improves serum antioxidant capacity

[52], and improves endothelial reactivity [38] in the postprandial period.

The goal of this research was to expand on the sparse human intervention trials to

prospectively evaluate the effect of regular grapefruit consumption on cardiovascular

disease risk factors, both classic and novel. We chose ruby red grapefruit for our studies

given evidence that suggests that the red grapefruit has higher bioactive content than the

white fruit cultivars [52]. We also targeted overweight and obese adults given the

propensity for the development of CVD and likelihood that baseline CVD biomarkers of

risk would be perturbed prior to participation in the trial. The aims of the current

randomized controlled trial were:

1) Evaluate the role of six weeks of daily consumption of ruby red grapefruit on

classical CVD risk factors including weight loss, blood pressure reduction, and

improvements in the lipid profile (reduced total cholesterol, LDL cholesterol, and

triglycerides and increased HDL cholesterol) in overweight and obese adults;

2) Evaluate the role of six weeks of daily consumption of ruby red grapefruit on

novel risk factors of cardiovascular health including circulating C-reactive

protein, soluble vascular adhesion molecule-1 (sVCAM-1), and F2-isoprostanes

Page 22: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

21

in overweight and obese adults, and in a subsample of participants with the

Metabolic Syndrome;

3) Examine the acute, postprandial effects of ruby red grapefruit consumption when

consumed with a high fat, high carbohydrate meal on metabolic markers including

glucose, triglycerides, and insulin and inflammatory/oxidative markers including

interleukin-6 (IL-6), sVCAM-1, soluble intercellular adhesion molecule-1

(sICAM-1), and F2-isoprostanes in overweight and obese adults.

We expected that regular grapefruit feeding would not significantly modify weight, but

would reduce blood pressure as well as promote improvements in the lipid profile by

reducing total and LDL cholesterols and triglycerides. Further, we hypothesized that

grapefruit intake would reduce markers of inflammation and oxidative stress after six

weeks of feeding as well as in the postprandial setting compared to control conditions.

Page 23: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

22

Page 24: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

23

CHAPTER 2

THE EFFECTS OF DAILY CONSUMPTION OF GRAPEFRUIT ON BODY WEIGHT,

LIPIDS, AND BLOOD PRESSURE IN HEALTHY, OVERWEIGHT ADULTS

Introduction

The current obesity epidemic provides a significant challenge, from both

economical and clinical perspectives. Currently, 68% of the American population is

overweight, while 50% of the overweight population is considered obese [53]. Obese

individuals endure greater health care costs and experience a reduced quality of life, as

well as suffer from a number of obesity-related comorbidities such as diabetes and

cardiovascular disease (CVD) [54, 55]. CVD, specifically, affects 79.4 million

Americans [3], and with the aging population and current trends in obesity prevalence,

this number is expected to increase in the coming years [56]. Strategies that promote

healthy weight and metabolic health are essential, and whole food approaches may

provide this option.

Grapefruit was first introduced as a weight loss food as part of the Hollywood

Diet of the 1930’s, consisting of 800-1000 kcals/day and half of a fresh grapefruit before

every meal for twelve days [27]. It is now known that grapefruit has no thermogenic

properties, and that any weight loss observed was likely attributable to the hypocaloric

diet [28, 29]. Nevertheless, a biologically plausible association between grapefruit intake

and weight control can be suggested given the known effects of select bioactive

constituents in grapefruit on adiposity [20, 57], and the proposed effects of the whole

fruit on satiety [18, 19].

Page 25: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

24

In fact, weight loss (1.6 kg-5.8 kg) has been observed in two clinical trials

evaluating the role of grapefruit in promoting healthy weight [18, 19]. A low-energy

dense preload, such as fruit, is thought to encourage reduced energy consumption at the

subsequent meal. Indeed, a clinical trial considering the form of the preload (solid,

pureed, or liquid) on energy intake showed that a solid preload of apple slices resulted in

a greater reduction in energy intake at the following meal compared to apple sauce or

apple juice. However, a study evaluating the effect of grapefruit preload to reduce energy

consumption before meals as compared to grapefruit juice or water alone showed no

difference between groups [18].

Despite the conflicting results regarding energy intake and fruit, there is a great

deal of evidence supporting citrus, and grapefruit specifically, in promoting

cardiovascular health. These effects are likely attributable to the flavonones found in the

pith of grapefruit, naringin, and hesperidin, which are converted to the aglycones,

naringenin and hesperitin in the gut [22]. These bioactives have demonstrated anti-

oxidant, hypolipidemic, and anti-hypertensive properties in cell culture, animal model,

and clinical trial studies [20, 21, 23, 35, 36, 38].

Naringenin’s hypolipidemic properties have been confirmed in numerous studies

in both rats and mice [20, 58-61]. Many animal studies use dosages of naringenin

unattainable by the human diet; however, one trial that tested naringenin supplementation

at a dose comparable to 1-4 cups of grapefruit juice/day resulted in a significant reduction

in plasma triglyceride and total cholesterol concentrations in rats [20]. While

naringenin’s hypolipidemic effects in animal models have been well characterized,

Page 26: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

25

results from clinical trials are highly heterogenous. Some clinical trials of grapefruit

intake or a grapefruit flavonoid supplement have shown no change in any parameter of

the lipid profile [19, 35]. Conversely, a trial of whole grapefruit and grapefruit juice

demonstrated beneficial changes in HDL, but no other lipid measurements (TC, LDL-C,

and triglycerides) when compared to controls [18]. Another trial in patients with

coronary atherosclerosis showed reductions in triglycerides, LDL, and TC, but no change

in HDL [34] in response to daily grapefruit consumption. Clearly, more research is

necessary in order to elucidate a definitive role of grapefruit in lipid control.

Studies of blood pressure and citrus intake are limited, but suggestive of a

potential protective association. Ex vivo and in vitro models of naringenin/hesperitin

treatment have resulted in vasorelaxation and nitric oxide production [21, 36].

Additionally, clinical trials in adults using hesperitin or orange juice have shown

improvements in flow-mediated dilation [36], increased postprandial endothelial

reactivity, and reduced diastolic blood pressure [38]. Epidemiological data suggests an

inverse association between citrus intake and markers of endothelial dysfunction [39].

Despite these results, the two grapefruit feeding trials to date showed no effect on blood

pressure [18, 19].

Based on the gaps in the literature regarding grapefruit, CVD, and weight control,

the aims of this study were to determine the effects of daily consumption of 1.5 red

grapefruit for six weeks for reducing weight and blood pressure and improving the lipid

profile in overweight and obese adults.

Page 27: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

26

Methods

Study Sample. Two hundred and ninety three overweight and obese men and

premenopausal women who responded to print and electronic advertising were screened

via telephone, and eighty-five were enrolled into this randomized clinical trial (Figure 2).

Postmenopausal women were excluded in order to avoid any potential changes in

hormone therapy that may have affected study outcomes. Study eligible individuals had a

BMI of 25-45 kg/m2, were in a period of weight maintenance (≤10 pound fluctuation for

at least six months), had abstained from use of tobacco products for at least five years,

were willing to maintain their current exercise regimen (not to exceed 10 hours/week),

and were willing to discontinue supplement and anti-inflammatory medication use.

Those individuals with a diagnosis of diabetes, high cholesterol (≥225mg/dL),

cardiovascular disease, cancer, inflammatory disease, or liver or kidney disease, or taking

medications metabolized by the cytochrome P450 3A enzyme were excluded from the

study. Two cohorts completed the study over the course of two grapefruit seasons from

October 2009-March 2011. Written informed consent was obtained from all study

participants prior to enrollment. The University of Arizona Institutional Review Board

approved this study protocol.

Study Design. The study was a randomized, controlled design in which subjects were

enrolled in two separate cohorts (winter 2009 and winter 2010) corresponding to

seasonality of red grapefruit crop production. During the 2009 season randomization was

balanced 2:1 for grapefruit intervention knowing the available crop and to assure

Page 28: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

27

adequate numbers were enrolled in the intervention; during 2010, randomization was

reversed to support > recruitment to the control treatment in order to balance group size.

Subjects made four visits to the clinical research setting: at baseline, randomization

(week 3), three weeks into the intervention phase (week 6), and at the end of the

intervention phase (week 9). Anthropometry was performed at baseline, week 6, and

week 9. Blood biosamples were collected at weeks 3 and 9. Blood pressure was

measured in duplicate at all clinic visits using a ReliOn HEM-780REL.

Diet Intervention. Following the baseline visit, subjects consumed a diet low in bioactive

rich fruits and vegetables for the study duration. The washout was followed to increase

the probability that results could be attributed to grapefruit consumption itself and not

another component of the diet. The diet included fruits and vegetables such as apples,

pears, bananas, iceberg lettuce, cucumbers, white potatoes, and onions, and restricted

fruits and vegetables with high polyphenol and carotenoid content such as citrus, berries,

spinach, carrots, tomatoes, and sweet potatoes. All other food groups were consumed ad

libitum. Subjects were provided with a multivitamin/mineral supplement for the duration

of the study (One-A-Day®, Bayer Healthcare; Morristown, NJ) in order to standardize

supplementation; other dietary supplementation was prohibited for the duration of the

study.

The first three weeks of the trial entailed a washout phase, while the last six

weeks comprised the intervention phase. After the three-week washout, subjects were

randomized to the intervention group (n=42), in which they continued consuming the

Page 29: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

28

washout diet and supplemented their diet with one half of a fresh ruby red grapefruit

(Texas Rio Star® Grapefruit; distributed by RioQueen Citrus, Mission, TX) fifteen

minutes prior to breakfast, lunch, and dinner for six weeks or to the control condition

(n=32), in which they continued consuming the washout diet. Subjects were trained to

peel the grapefruit and eat all portions of the peeled grapefruit, including the pith.

Subjects were instructed to not add sugar to their grapefruit and were provided with a

non-calorie sweetener for use if desired.

Outcome Measurements. Diet composition was determined from repeat 24-hour diet

recalls. Dietary recalls were collected during the washout phase (n=3) and again during

the intervention phase (n=3). A Registered Dietitian unaffiliated with the study collected

the dietary recalls via telephone using the USDA multi-pass protocol [62]. Dietary data

was analyzed using the University of Minnesota Nutrition Data System for Research

software (NDS-R 2009). Energy intake was determined separately for the washout phase

and the intervention phase. Data from the phase-specific 24-hour recalls were combined

and averaged to determine the average intake for each respective phase.

Compliance to the grapefruit diet was measured via self-report. Following

randomization to the grapefruit group, subjects were provided with a log book in which

they were instructed to indicate each time grapefruit was consumed throughout the six

week intervention. Submitted logs were analyzed for compliance (# of times grapefruit

consumed before each meal/# meals)*100.

Page 30: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

29

Bioactive content of the grapefruit was also estimated using the USDA Database

for Flavonoid Content of Selected Foods, as measured by high performance liquid

chromatography at the Beltsville Human Nutrition Research Center. Flavonoid content is

reported in mg/100g. In order to estimate gram weight of grapefruit intake, a random

sample of ten grapefruits were weighed following peeling using an Ohaus CS 5000

digital food scale. Average weight of the grapefruits was calculated and recorded in

grams, and flavonoid content of the fruit was then estimated.

Anthropometry was performed at baseline, week 6, and week 9. Height was

measured without shoes to the nearest quarter inch using a wall-mounted stadiometer

[63]. Weight was measured without shoes or over garments to the nearest half pound

using a calibrated double ruler standing scale [63]. Waist and hip circumferences were

measured using a Gulick II measuring tape to the nearest quarter inch. Briefly, subjects

stood upright in front of a full-length mirror as the measuring tape was extended around

the waist or hip using the umbilicus and the widest part of the hip area as anatomical

reference points for the respective measurements [63]. Body fat was estimated using a

handheld Omron Body Fat Analyzer HBF-306 (Omron Healthcare, Inc.; Vernon Hills,

IL).

Blood pressure was measured in duplicate at baseline, week 3, week 6, and week

9 using a ReliOn HEM-780REL (Omron, Inc.; Bannockburn, IL) automated blood

pressure cuff. Subjects were instructed to place feet flat on the floor and not speak during

the measurement. Systolic, diastolic, and heart rate in beats per minute were recorded

separately and the average of the two measurements was used in analysis.

Page 31: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

30

Following an eight-hour overnight fast, blood was collected from the antecubital

vein into a serum tube with a Z serum clot activator at week 3 (randomization) and week

9 (end of intervention phase). Serum tubes were gently inverted and stored at room

temperature for 45 minutes to allow for clotting. The blood was then centrifuged at 3,000

rpm for 10 minutes at 4oC in a Sorvall RT 6000B Centrifuge. Following centrifugation,

serum was aliquoted into storage cryovials and stored at -80 oC until analysis.

The lipid profile was analyzed using an LDX Cholestech System, a validated

approach for use in assessment of serum lipids in the published literature [64]. At the

time of analysis, serum samples were thawed until reaching room temperature. 35μL

serum was pipetted into an LDX cassette and placed in the LDX Cholestech System.

Outcome measures included total cholesterol, HDL, LDL, and triglycerides. All samples

were run in duplicate and recorded as an average of the two measurements. Variance of

greater than 10% resulted in a third measurement. The means of the two measures that

were within 10% of each other were averaged for final analysis.

Statistical Analysis. Baseline characteristics between each treatment arm (and between

those who did and did not complete the study) were compared using chi-square tests for

categorical variables and independent t-tests for continuous variables. Attrition rates

between treatment arms were compared using t-tests. Change in all anthropometric and

blood pressure measurements were compared from baseline to post-intervention using

paired t-tests. Lipid parameters were compared from post-washout to post-intervention

using paired t-tests. Post-intervention values were compared between the two treatment

Page 32: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

31

arms using linear regression, adjusting for baseline measures; the Wald statistic p-value

for the post-intervention is reported. Measures of dietary intake were compared between

the washout phase and the intervention phase using paired t-tests within each treatment

arm. Intervention-phase values were compared between the two treatment arms using

linear regression, adjusting for washout-phase values; the Wald statistic p-value for the

intervention-phase term is reported. Differences were considered statistically significant

at p < 0.05 for all tests. No adjustments were made for multiple comparisons. Values are

expressed as mean ± standard deviation (SD). All statistical analyses were performed

using Stata 11.1 (StataCorp, College Station, TX).

Results

Subjects. Over the two enrollment periods, seventy-four subjects completed the three-

week washout phase and were randomly assigned to the control group (n=32; 25 women,

7 men) or the grapefruit group (n=42; 32 women, 10 men). Three subjects (all women)

randomized to the grapefruit treatment withdrew from the study during the six-week

intervention phase, resulting in 71 subjects completing the trial. At baseline, there were

no statistically significant differences between groups with regard to demographics,

anthropometrics, or blood pressure (Table 1). No significant differences in baseline

characteristics were observed between non-completers and completers.

Anthropometric and Blood Pressure Changes. Participants in the grapefruit group

demonstrated an average weight loss of −0.61±2.23 kg, a loss that approached

Page 33: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

32

significance (p=0.097). Waist circumference and waist to hip ratio were significantly

reduced in those consuming grapefruit by −2.45±0.60 cm (p<0.001) and −0.01±0.01

(p=0.019), respectively. No significant anthropometric changes were observed in the

control group or between groups following the intervention phase after controlling for

baseline values.

Participants consuming grapefruit, but not the control group, demonstrated a

significant reduction in mean systolic blood pressure of −3.21±10.13 mmHg (p=0.039).

Reductions in diastolic blood pressure or resting heart rate were not observed in either

group. No significant differences were observed between groups with regard to blood

pressure following the intervention phase (Table 2).

Energy and Bioactive Intake and Compliance. Overall energy intake, macronutrient and

micronutrient content of the diet were not different between groups during the washout

phase or during the intervention phase. Additionally, no within group changes were

observed between phases in either study arm with regard to energy intake and

macronutrient distribution. Fruit and vegetable intake changed significantly in both

groups during the intervention phase. Fruit intake increased in the grapefruit group by

1.2 servings/day (p<0.001), while intake decreased in the control group by -0.8

servings/day (p<0.001), an effect that was significant between groups (p<0.001).

Vegetable intake also decreased during the intervention phase in the grapefruit group (-

0.7 servings/day, p=0.03).

Page 34: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

33

During the intervention phase, daily Vitamin C intake increased from 56.0±29.2

mg to 155.4±74.5 mg in the grapefruit group. This increase was statistically significant

within the group when compared to washout phase values (p<0.001) and when compared

to the mean vitamin C intake during the intervention phase by the control group

(54.5±36.9 mg, p<0.001). Sodium intake also increased in the control group during the

intervention phase from 3381.3±1304.5 mg/day to 3747.3±1371.7 mg/day, which did not

reach statistical significance (p=0.079). This result was not observed in the grapefruit

group; however, a significant difference in sodium intake was observed between groups

during the intervention phase (p=0.012).

Thirty-two of the thirty-nine participants who completed the intervention

treatment logged their grapefruit consumption. Of those subjects, compliance was

recorded at 93±11%. Grapefruit weighed, on average, 298.4±22 grams (Appendix I).

Based on the USDA Flavonoid Content of Selected Foods Database, naringenin and

hesperitin content of pink grapefruit is estimated at 32.64 mg/100g and 0.35 mg/100g,

respectively. No estimates have been made regarding the flavonoid content of the Rio

Red fruit, though previous research has shown similarities in flavonoid content between

the Rio Red and the Thompson Pink juices [65]. Thus, we estimated that naringenin

intake in those consuming grapefruit was 146.2 mg/day, and hesperidin intake was

estimated at 1.57 mg/day.

Lipid Profile. The lipid profile was analyzed for all subjects whose blood was

successfully collected at both pre-randomization and post-intervention visits (n=69). At

Page 35: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

34

pre-randomization (post-washout) and following the intervention phase, no significant

difference was observed between groups in regards to any parameter of the lipid profile

(total cholesterol, LDL cholesterol, HDL cholesterol, or triglycerides). However, total

cholesterol decreased significantly in both the intervention and control groups when

compared to their baseline values by -11.7 ± 21.3 mg/dL (p=0.002) and -6.9 ± 17.5

mg/dL (p=0.032), respectively. Additionally, those subjects in the grapefruit group

demonstrated a reduction in LDL cholesterol values of -16.0 ± 41.4 mg/dL (p=0.024)

when compared to their baseline values (Table 4).

Discussion

Several studies have investigated the effects of grapefruit and/or its bioactives for

attenuating the risk for CVD or as a weight loss food, though results are inconclusive.

The present study indicates a beneficial role for daily, pre-meal grapefruit for six weeks

in reducing the risk for CVD by promoting reductions in waist circumference, systolic

blood pressure, and total and LDL cholesterols compared to baseline values, but not

compared to values in those following a control treatment.

Six-week consumption of grapefruit did not result in a statistically significant

reduction in weight in this trial. Energy intake did not change significantly in either the

control or the grapefruit treatments, indicating that grapefruit does not promote a satiety-

induced restriction in energy. Results from previous studies show inconsistent results. In

a four arm study design, Fujioka et al observed a significant reduction in weight in those

consuming whole grapefruit compared to those consuming a placebo pill, while no

Page 36: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

35

difference was observed in those consuming grapefruit juice or a grapefruit pill compared

to the placebo pill [19], suggesting that whole grapefruit does promote weight loss.

Conversely, Silver et al showed no difference in weight loss between those consuming

grapefruit, grapefruit juice, or water as preloads in addition to calorie restriction [18]. In

contrast to both of these studies, only our study observed a significant reduction in waist

circumference, a measure of central adiposity, in those randomized to the grapefruit

treatment. To our knowledge, only animal studies, not studies in humans, have shown

the beneficial effects of naringenin on reducing central adiposity [20]. Naringenin also

has known lipolytic effects in animals and cell culture studies [20, 57]. In the present

study a reduction in waist circumference was observed, though body fat percentage was

not changed, suggesting the effect may be specific to central adiposity. Of course,

circumferences are prone to measurement error even when trained personnel take

measures. Despite this, the literature indicates that central adiposity is more highly

correlated with risk for CVD than body fat percentage, suggesting that a reduction in

waist circumference is of greater relevance for cardiovascular health.

Grapefruit consumption also resulted in a significant reduction in systolic blood

pressure. Though the average BMI of participants in this study was 32 kg/m2, the

average baseline blood pressure was, on average, only 119/81 mmHg, suggestive of

borderline prehypertensive status. While our results were statistically significant, it can

be postulated that these effects may be more pronounced in a hypertensive population,

although this will need to be prospectively tested. In fact, many studies have shown a

beneficial effect of citrus intake on blood pressure, though this is the first to show an

Page 37: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

36

effect of grapefruit, specifically, on blood pressure or of an effect of citrus on systolic

blood pressure. High flavonoid sweetie fruit intake was associated with a reduction in

diastolic blood pressure in hypertensive men [37]. Hesperidin supplementation has also

resulted in improved flow-mediated dilation of the brachial artery [36], as well as a

reduction in diastolic blood pressure and postprandial microvascular reactivity [38].

Naringenin treatment (0.1 mM) of the rat aortic myocytes resulted in inhibition of

calmodulin-associated phosphodiesterases, ultimately leading to vasorelaxation [21].

This concentration of naringenin is unattainable by the diet, even given the high intake in

our population (estimated at 146.2 mg/day). Nevertheless, the aforementioned pathway

may explain the mechanism by which naringenin intake facilitated the systolic pressure

reductions observed in our population.

Micronutrient content of the diet must also be considered when evaluating blood

pressure change. Potassium intake did not change. Vitamin C intake increased in the

grapefruit treatment group, an increase that was significant compared to baseline values

as well as intervention values in the control group. Vitamin C is a known antioxidant and

a potent scavenger of superoxide [66, 67]. Superoxide is known to inhibit eNOS,

ultimately producing a state of vasoconstriction [67]. Thus, vitamin C has been proposed

as a natural anti-hypertensive treatment, with some evidence to support this hypothesis

[66]. Considered in combination with the proposed effects of naringenin on the

vasculature, this may explain the blood pressure benefit observed in our trial.

Fruit and vegetable consumption changed in both the grapefruit and control

groups during the intervention phase when compared to the washout phase. Fruit intake

Page 38: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

37

increased in the grapefruit group, as expected due to the increase in grapefruit

consumption. However, fruit intake decreased in the control group during the

intervention phase, perhaps due to the limited types of fruits and vegetables that they

could consume. Despite this reduction in fruit and vegetable consumption, outcomes

such as weight and blood pressure did not increase as a response in the control group.

Total cholesterol (TC) was significantly lowered in both groups, though this effect

was larger and more physiologically significant in those in the grapefruit group. More

importantly, LDL cholesterol (LDL-C) was reduced by an average of -16.0 mg/dL due to

daily grapefruit consumption. Results from previous research on grapefruit intake and

the lipid profile are extremely variable. Fujioka et al observed no changes in HDL

cholesterol and triglycerides after twelve weeks of treatment [19]. Silver et al observed a

significant increase in HDL in those consuming grapefruit or grapefruit juice as a preload

by 3.0 mg/dL or 4.9 mg/dL, respectively, but observed no changes in any other parameter

of the lipid profile after twelve weeks [18]. However, the present study and all other

studies evaluating citrus and lipid change have not shown this effect on HDL.

Eight-week naringin supplementation (400 mg/day) resulted in a 14% reduction in

TC and a 17% reduction in LDL-C in hypercholesterolemic patients, whereas the same

treatment showed no benefit in individuals with normal cholesterol values [68]. Type of

grapefruit consumed also seems to be an important factor in relation to the cholesterol-

lowering effects of grapefruit, though no mechanism currently exists to explain this

effect. In a study of 57 patients with coronary atherosclerosis, consumption of one ruby

red or white grapefruit for thirty days resulted in reduction in TC and LDL-C compared

Page 39: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

38

to controls, but only those eating the ruby red cultivar observed additional reductions in

triglycerides [34]. Despite consuming red grapefruit, our population showed no

significant change in triglycerides possibly because the participants were overall healthy

and any history of cardiovascular disease was an exclusion criterion for the study. While

the grapefruit bioactives have known hypolipidemic effects as observed in many animal

model studies, 500 mg/day or 800 mg/day of naringin or hesperidin, respectively, showed

no benefit on any parameter of the lipid profile in mildly hypercholesterolemic adults

[35]. These results may suggest that the bioactive constituents must be consumed

simultaneously. In addition, fiber is known to reduce circulating cholesterol levels [69];

however, our population demonstrated no change in overall fiber intake. This suggests

that the lipid lowering effects of grapefruit are due to its high bioactive content, and not

its fiber.

The results of the present study regarding improvements in the lipid profile are

particularly robust when coupled with the change observed in waist circumference.

Dyslipidemia, characterized by elevated TC, LDL-C, and triglycerides, and reduced

HDL-C, is a risk factor for CAD and is associated with obesity [43-46]. Interestingly,

visceral adiposity is a much greater predictor of dyslipidemia than BMI [9] and is thought

to be its primary cause [70]. Visceral adipose tissue is more metabolically active than

subcutaneous adipose tissue, characterized by altered lipid metabolism, resulting in

increased triglycerides and LDL-C concentrations in circulation [70]. Thus, the reduction

observed in waist circumference may have triggered the decrease in TC, and particularly,

LDL-C.

Page 40: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

39

The lipid lowering effects due to grapefruit are of important clinical significance

in that they may provide a primary prevention approach or an early intervention strategy

prior to pharmaceutical approaches. Statins, the primary lipid-lowering therapy, are

extremely effective at lowering lipids as well as reducing risk for cardiovascular events

and mortality [71]. Despite the benefits of statin therapy, compliance is low with

approximately 60-75% of cardiovascular patients discontinuing therapy within five years

due to side effects [71]. In a small, but significant population, statins may cause adverse

reactions including myalgia, muscular tenderness, pain, and in select cases advance to

extreme muscle weakness, known as myositis [72]. While our results do not compare to

the lipid lowering results of short term statin therapy [73], grapefruit may be considered

as an alternative therapy in those who do not require aggressive lipid reductions or in

those who may experience myositis and other complications.

The results of this study must be interpreted with caution, and the limitations

should be noted. Dietary composition was measured by 24 hour recalls, and a dietary

record may have resulted in greater accuracy, though this may have also increased

participant burden and resulted in greater attrition. Measurement of waist circumference

is also known to have a high degree of variability, even when performed by a trained

individual. Individuals performing measurements were not blinded to treatment

condition, which may have introduced bias, though they were blinded to previous

recorded measures. The Australian Risk Factor Prevalence Study, a study of 9, 279

adults, showed that waist circumference is subject to measurement error of approximately

1.84 cm and concluded that a change greater than 2 cm is statistically significant [74],

Page 41: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

40

findings that suggest the change observed in our population is not due to measurement

error alone. Additionally, more precise and objective measures of body composition such

as is provided by dual x-ray absorptiometry, computed tomography, or magnetic

resonance imaging instead of bioelectrical impedance may have enhanced the capacity to

detect significant changes in body composition over time or by treatment group. The

present study was also limited in that physical activity was not measured or recorded.

Participants were instructed to maintain their physical activity regimen, not to exceed ten

hours per week. However, participants may have increased their physical activity, which

may have resulted in the reductions in blood pressure, lipids, and waist circumference.

An increase in fruit intake may also explain the results of this study. Grapefruit intake

was responsible for the increase in fruit consumption in the intervention group. It is

possible that the increase in fruit itself, not grapefruit specifically, may have driven the

positive results observed here. Finally, the present trial may have been limited by time.

Our intervention period lasted only six weeks, as compared to twelve weeks in the two

previous clinical trials evaluating grapefruit in weight and CVD risk factors [18, 19].

Had the trial lasted longer, it is possible that a significant reduction in weight may have

been observed.

Based on the results from the current trial, we conclude that 6 week consumption

of one and a half grapefruit per day is not effective at reducing weight in an overweight

and obese population when compared to a usual dietary intake control group. In spite of

this, grapefruit consumption does elicit beneficial effects compared to baseline values

that are associated with reducing the risk for CVD. We observed a significant decrease in

Page 42: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

41

waist circumference, which may have facilitated a reduction in LDL and total cholesterol.

Finally, systolic blood pressure was significantly lowered, an effect not seen before in a

study of grapefruit consumption. Future research must replicate these findings in a larger

sample in order to confirm our results of grapefruit as a lipid and blood pressure lowering

food. Due to the variability in study design as well as outcomes from previous studies, it

will be necessary to elucidate an optimal dose of grapefruit consumption regarding lipid

and blood pressure control. The findings from the present study as well as from previous

studies regarding grapefruit, its bioactives, and similar citrus fruits suggest that grapefruit

may be a beneficial addition to a heart healthy diet and underscores the need for a larger,

longer duration trial.

Page 43: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

42

FIGURES

Figure 2. Consort diagram describing the flow of participants through the grapefruit

intervention feeding trial.

Screened for Eligibility n=293

Intervention n=42 ½ ruby red grapefruit before

each meal

Control n=32 Maintain low bioactive

diet

Consented n=85 Consume low bioactive diet

Eat ½ low bioactive fruit before each meal

Completed trial

n=39

Completed trial

n=32

Excluded n=208 Not Willing to Participate n=50

Postmenopausal n=34

BMI out of range n=28

Medication n=22

Metabolic/Inflammatory

Smoker n=17

Weight Loss in prev 6 mos n=9

Lost to follow through n=12

Other n=22

Discontinued Trial n=11 Washout Diet too strict n=6

Wanted to take supplements n=2

Personal reasons n=3

Randomized n=74

Lost to follow up n=3

3-W

EE

K W

AS

HO

UT

-

WE

EK

IN

TE

RV

EN

TIO

N

Page 44: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

43

TABLES

TABLE 1. Baseline characteristics of participants who completed the grapefruit feeding trial:

grapefruit vs. control

Total (n = 71) Grapefruit (n = 39) Control (n = 32)

Age (Years) 41.2±11.0 39.4±10.7 44.0±11.0

Sex, n (%)

Female 54 (76.1) 29 (74.3) 25 (78.1)

Male 17 (23.9) 10 (25.6) 7 (21.9)

Race, n (%)

White 44 (62.0) 24 (61.5) 20 (62.5)

Other 27 (38.0) 15 (38.5) 12 (37.5)

Education, n (%)

High School Degree 26 (36.7) 13 (33.3) 13 (40.6)

Undergraduate Degree 23 (32.3) 15 (38.5) 8 (25)

Graduate Degree 5 (7) 2 (5) 3 (9)

Other 1 (1) 0 (0) 1 (3)

Weight (kg) 91.7±13.6 92.4±14.9 90.8±12.8

BMI (kg/m2) 32.1±4.1 32.9±4.2 31.4±3.8

Body Fat (%) 35.7±6.1 36.3±6.2 35±6.0

Waist Circumference (cm) 103.2±10.5 103.2±11.2 103.3±9.7

Hip Circumference (cm) 116.9±9.4 117.2±10.3 115.4±7.3

W:H Ratio 0.88±0.08 0.88±0.08 0.88±0.08

Systolic (mmHg) 119.1±16.6 118.4±16.9 119.1±15.9

Diastolic (mmHg) 80.6±10.5 80±10.2 81.3±11.1

Heart Rate (bpm) 72.8±11.1 72.6±10.4 73.1±12.1

Data shown indicate mean ± standard deviation. No differences existed between groups at baseline.

Page 45: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

44

TABLE 2. Anthropometric and blood pressure changes in participants who completed the grapefruit feeding trial: grapefruit vs control.

Grapefruit (n=39)

Control (n=32)

Change Post-

interventiona

p valueb Change

Post-

interventiona

p valueb p value

c

Weight (kg) −0.61±2.23 90.9 0.098 −0.11±1.1 91.5 0.56 0.302

BMI (kg/m2) −0.23±0.13 31.9 0.09 −0.03±0.06 32.1 0.62 0.362

Body Fat (%) 0.67±1.0 35.8 0.13 0.12±0.3 36.1 0.682 0.307

Waist Circumference (cm) −2.45±0.60 100.0 <0.001 −1.23±0.71 101.8 0.092 0.188

Hip Circumference (cm) −0.77±3.17 116.0 0.135 0.02±0.44 116.8 0.972 0.311

W:H Ratio −0.01±0.01 0.87 0.019 −0.01±0.01 0.87 0.23 0.557

Systolic (mmHg) −3.21±10.13 114.7 0.039 −0.31±12.65 117.6 0.978 0.213

Diastolic (mmHg) −0.90±7.47 79.0 0.367 1.03±8.56 80.0 0.565 0.419

Heart Rate (bpm) −0.72±8.9 72.3 0.529 −2.63±9.62 70.8 0.151 0.425

P values comparing postwashout differences between study arms were insignificant. aPostintervention means are reported after controlling for potential confounders by ANCOVA: BMI, age, sex, and washout-phase values.

bWald statistic from linear regression model comparing post-intervention values within study arms, adjusted for washout-phase values.

cANCOVA comparing postintervention values between study arms, adjusted for washout-phase values, baseline BMI, age, and sex.

Page 46: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

45

TABLE 3. Comparison of diet composition in participants who completed the grapefruit feeding trial: grapefruit vs control.

Grapefruit (n=38) p valuea Control (n = 32) p value

a p value

b

Washout Intervention

Washout Intervention

Energy (kcal) 1784±511 1839±488 0.437 1940±730 1913±536 0.791 0.955

Fruit (servings) 1.8±0.2 3.0±0.2 <0.001 1.9±0.2 1.1±0.2 <0.001 <0.001

Vegetable (servings) 3.3±0.3 2.6±0.2 0.03 2.9±0.3 3.1±0.3 0.562 0.115

Fat (g) 69±23 70±25 0.765 77±38 78±29 0.906 0.47

Saturated Fat (g) 23±8 24±10 0.337 25±13 26±10 0.716 0.743

Carbohydrate (g) 220±78 232±73 0.262 240±84 228±67 0.284 0.231

Protein (g) 73±19 74±16 0.74 77±33 78±25 0.709 0.458

Fiber (g) 22±9 23±9 0.62 23±9 21±7 0.199 0.129

Sodium (mg) 3357±1054 3161±836 0.244 3381±1305 3747±1372 0.079 0.012

Potassium (mg) 2545±819 2506±646 0.715 2637±949 2471±740 0.128 0.465

Folate (mcg) 393±151 395±146 0.917 387±161 441±175 0.139 0.184

Vitamin C (mg) 56±29 155±74 <0.001 60±29 55±37 0.372 < 0.001 aPaired t-test comparing dietary intake during the washout and intervention phases within each study arm.

bWald statistic from linear regression model comparing post-intervention values between study arms, adjusted for washout-phase values.

Page 47: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

46

TABLE 4. Comparison of the lipid profile in those participants who completed the Grapefruit Feeding Trial: Grapefruit vs Control,

before and after the intervention phase.

Grapefruit (n=37)a p value

c Control (n = 32)

a p value

c p value

d

Pre Post Post

Pre Post Post

TC (mg/dL) 192.6±29.7 180.93±22.9 178.8 0.002 185.8±35.4 178.9±27.4 181.2 0.033 0.587

HDL (mg/dL) 43.7 ±11.8 42.8±11.2 43.4 0.493 41.6±14.0 43.9±13.9 43.8 0.493 0.832

LDL (mg/dL) 122.7±44.4 106.7±30.9 110.5 <0.001 110.6±36.3 106.8±27.2 111.2 0.009 0.871

TG (mg/dL) 111.5±73.3 119.9±68.1 127.4 0.346 133.3±91.4 130.4±72.9 126.7 0.644 0.95

TC = total cholesterol; HDL = high density lipoprotein; LDL = low density lipoprotein; TG = triglycerides. P values comparing

postwashout differences between study arms were insignificant. aLipid parameters were undetectable by the Cholestech LDX System in some serum samples. Grapefruit: TC and HDL (n=37), LDL

(n=34), TG (n=33); control: TC (n=32), HDL (n=31), LDL (n=28), TG (n=30). bPostintervention means are reported after controlling for potential confounders by ANCOVA: BMI, age, sex, and washout-phase

values. cPaired t-test comparing measurements of the lipid profile pre- and postintervention within each study arm

dANCOVA model comparing postintervention values between study arms, adjusted for washout-phase values.

Page 48: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

47

CHAPTER 3

DAILY CONSUMPTION OF GRAPEFRUIT FOR SIX WEEKS

DOES NOT REDUCE SVCAM-1, CRP, OR F2-ISOPROSTANES IN

OVERWEIGHT AND OBESE ADULTS

Introduction

Atherosclerotic diseases, characterized by endothelial dysfunction, are the global

leading cause of death [2]. Atherosclerosis is the manifestation of damage to the

endothelium due to a dysregulated cardiometabolic state (elevated lipids, glucose, and

insulin), inflammation and oxidative stress [15]. Individuals with Metabolic Syndrome

(MetS) are at particularly high risk of developing atherosclerosis due to hyperlipidemia

and elevated inflammatory/oxidative processes that develop as a result of visceral

adiposity [2, 15]. Methods to reduce atherosclerotic plaque production via promotion of

a healthy cardiometabolic state and a reduction in inflammatory/oxidative processes are

warranted.

Large epidemiological trials suggest that citrus may be an important component

of a heart healthy diet. The Prospective Epidemiological Study of Myocardial Infarction

(PRIME) in 50-59 year old men found that citrus intake, but not intake of other fruits,

was associated with a reduction in incidence of ischemic events [25]. Data from the

Nurses’ Health Study (NHS) indicate that citrus and grapefruit intake specifically is

associated with lower levels of fasting pro-inflammatory, cardiovascular disease (CVD)-

associated cytokines [39]. Further, an analysis of dietary flavonoid intake in NHS

suggested that high flavonone (flavonoids in citrus) intake, but not other flavonoids,

Page 49: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

48

reduced the risk of ischemic stroke [75]. Together, these data suggest both citrus and

grapefruit may promote cardiovascular health.

In addition to epidemiological evidence supporting a role for flavonones in CVD

health, experimental evidence has shown that flavonones exert significant antioxidant,

anti-inflammatory, and antihypertensive properties in cell culture and animal model

studies [21, 36, 76]. Randomized human trials are limited but suggest that flavonones

have endothelial protective properties, though flavonones from orange juice have been

the most widely tested [36, 38]. To date, no randomized human trials have been

conducted to assess the effect of whole grapefruit, rather than flavonone supplements or

orange juice, on outcomes associated with CVD in an at-risk population. Our group

recently demonstrated that six weeks of daily ruby red grapefruit intake significantly

reduced total and low-density lipoprotein (LDL) cholesterol, systolic blood pressure, and

waist circumference (WC) in healthy, overweight and obese adults [77].

The purpose of this secondary analysis was to determine the effect of six-week

consumption of ruby red grapefruit on biomarkers associated with atherosclerotic plaque

production: soluble vascular adhesion molecule-1 (sVCAM-1), high-sensitivity C

reactive protein (hsCRP), and F2-isoprostanes. Further, we sought to examine

associations between classic demographic and anthropometric cardiovascular risk factors

(age, sex, BMI, and WC) and atherogenic biomarkers at baseline and following the

intervention. Lastly, we assessed the role of grapefruit consumption in participants with

elevated risk for cardiometabolic diseases by performing a post hoc analysis on

participants classified as having MetS.

Page 50: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

49

Methods

Study Population. The methods and primary results from this randomized, controlled

trial have been reported previously [77]. Briefly, 85 non-smoking healthy men and

premenopausal women over the age of 18 who were free of inflammatory diseases or

history of cancer, heart disease, hypercholesterolemia (≥225 mg/dL), hepatic or renal

disease, or diabetes were enrolled. Eligible participants had a body mass index (BMI)

between 25 and 45 kg/m2. Individuals taking medications metabolized by the

cytochrome P450-3A enzyme were excluded due to potential adverse interaction with

grapefruit [78]. The University of Arizona Institutional Review Board approved this

protocol, and all participants provided informed consent.

Study Design. This study was completed over two grapefruit seasons (winter 2009-10

and winter 2010-11). Participants were enrolled in the trial for a total of nine weeks and

made visits to the clinic at four time points (baseline, week 3, week 6, and week 9).

Following the baseline visit, participants consumed a washout diet, restricted in

bioactive-rich fruits and vegetables and devoid of all citrus fruit and juices. Seventy-four

participants completed the washout diet and were randomized either to the control group

(n=32), in which they continued consuming the low bioactive diet, or to the grapefruit

group (n=42), in which they consumed the low bioactive diet plus half of a fresh ruby

red grapefruit (Texas Rio Star Grapefruit; grown by Rio Queen Citrus, Mission, TX)

before each meal (3x daily) for six weeks. Participants were trained to peel the grapefruit

Page 51: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

50

and eat all portions of the fruit, including the innermost white membrane of the fruit (the

albedo), without adding caloric sweeteners to the fruit.

Anthropometry and Blood Pressure Measures. Height, weight, and WC were measured

at baseline, week 6, and week 9. Height and weight were measured to the nearest quarter

inch and the nearest half-pound, respectively. WC was measured at the umbilicus using a

Gulick II (Elmsford, NY) measuring tape. Blood pressure was measured at baseline,

week 3, week 6, and week 9 using a ReliOn HEM 780REL (Omron; Bannockburn, IL)

automated blood pressure cuff following standard procedures [77].

Biosample Collection and Processing. Urine and blood samples were collected at the end

of the washout phase (week 3) and the end of the intervention phase (week 9).

Participants collected a first morning urine sample on the three days prior to their clinic

visit. Urine was stored in ice chests with ice packs until arrival at the research clinic.

The three collections were then pooled and centrifuged at 3,000 rpm for 10 minutes at

4°C. Following an eight-hour overnight fast, blood was collected into serum and plasma

tubes. Plasma and serum were processed following standard procedures [77]. After

centrifugation, plasma, serum, and urine were aliquoted into storage cryovials and stored

at -80°C until analysis.

Biosample Analysis. Serum samples were analyzed for high-density lipoprotein (HDL),

triglyceride, and glucose concentrations using an LDX Cholestech System (Hayward,

Page 52: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

51

CA). Samples were run in duplicate and recorded as an average of the two

measurements.

Plasma samples were analyzed for hsCRP and sVCAM-1 concentrations using

enzyme linked immunosorbency assay (ELISA) kits (United States Biological, San

Antonio, TX and R&D Systems, Minneapolis, MN; respectively). The mean inter- and

intraassay coefficients of variation (CVs) were 3.3% and 6.3% (hsCRP) and 13.4% and

6.3% (sVCAM-1), respectively.

F2-isoprostanes were measured in 3-day, first morning, pooled urine samples.

Urine samples were purified by adding three volumes of ethanol to one volume of

sample, vortexed, and cooled to 4°C. Ethanol-sample mixtures were centrifuged for 20

minutes at 1,500xg. The supernatant was then decanted and the ethanol was evaporated

off using a centrifugal evaporator. F2-isoprostanes were quantified in the purified

samples in duplicate via a competitive ELISA kit (Cayman Chemical, Ann Arbor, MI).

Mean inter- and intraassay CVs were 10.8% and 2.5%, respectively. F2-isoprostane

values were corrected for creatinine concentration (Cayman Chemical, Ann Arbor, MI).

Metabolic Syndrome (MetS) Classification. Participants were classified as having MetS

at study entry, post hoc, based on guidelines set by the National Cholesterol Education

Program Adult Treatment Panel III (NCEP:ATPIII) [79]. MetS classification requires the

presence of at least three of the following five criteria: elevated triglycerides (>150

mg/dL), reduced HDL (<40 mg/dL in men, <50 mg/dL in women), elevated waist

Page 53: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

52

circumference (>102 cm in men, >88 cm in women), elevated blood pressure (≥130/≥85

mmHg), or elevated fasting glucose (≥100 mg/dL).

Statistical Methods. Baseline characteristics of participants in each treatment arm (or for

participants with and without MetS) were compared using χ2

tests for categorical

variables and t-tests for continuous variables. Within each treatment arm (total enrolled

and only those with MetS), inflammatory and oxidative stress markers were compared

from pre- to post-intervention using paired t-tests. Change values (total enrolled and only

those with MetS) were compared between the two arms using linear regression,

controlling for baseline values.

Linear regression models were conducted to explore the relationship between risk

factors of cardiometabolic diseases and levels of inflammatory/oxidative stress markers

in the entire cohort at baseline and in each arm separately post-intervention. Potential

interactions between cardiometabolic risk factors and treatment arm on each post-

intervention biomarker outcome were tested using likelihood ratio tests. Analyses were

performed using Stata 12.1 (StataCorp, College Station, TX).

Results

Three participants randomized to the grapefruit group were lost to follow up

during the intervention phase, and blood collections were unsuccessful for two additional

participants, leaving 37 and 32 participants randomized to the grapefruit and control

groups, respectively. Participants had a mean ± standard deviation (SD) age of 41.8±10.7

Page 54: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

53

years and BMI of 32.1±4.1 kg/m2 (Table 5). Of the 69 participants who successfully

completed the study, 29 (42%) were classified as having MetS at baseline. There were

no statistically significant differences at baseline for any measurement, including

inflammatory/oxidative stress measurements between randomization groups.

Changes in inflammation and oxidative stress were analyzed within and between

randomization arms. No significant difference was detected with regard to change in

sVCAM-1 values from baseline to post-intervention between groups (Table 6a); changes

in sVCAM-1 were also not significant over time within treatment groups. Though not

statistically significant, hsCRP values decreased in in the grapefruit group and increased

in the control group; the difference in hsCRP change score between groups was

marginally significant (p=0.073). Similarly, F2-isoprostanes decreased in the grapefruit

group and increased in the control group, demonstrating a difference that approached

significance in F2-isoprostane change between groups (p=0.063).

To further explore the effect of grapefruit consumption on cardiometabolic risk

factors, post hoc analyses of participants with MetS were performed (n=14 in the

grapefruit arm, n=15 in the control arm). At baseline hsCRP values averaged 2.7±1.6 in

participants with MetS compared to 2.0±1.8 in participants without MetS a difference

that was marginally significant (p=0.076). In an analysis restricted to those with MetS,

there was no significant change in sVCAM-1 regardless of group assignment nor were

there any differences between groups at the post-intervention timepoint (Table 6b).

Similar null results were shown for hsCRP values. Similar to the results from the entire

cohort, F2-isoprostane values in participants with MetS were marginally significantly

Page 55: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

54

lower in the grapefruit intervention group compared to the control group, post-

intervention (11.9±4.7 vs. 18.3±10.9, respectively; p=0.058).

Associations between CVD risk factors (BMI, waist circumference, age, and sex)

and inflammatory/oxidative stress markers (sVCAM-1, hsCRP, and F2-isoprostanes)

were tested at baseline in the entire cohort and post-intervention in each arm separately.

At baseline, sVCAM-1 values were associated with age, hsCRP values were associated

with BMI and waist circumference, while F2-isoprostanes were only associated with WC

(p<0.05 for all associations, Table 7). WC and post-intervention F2-isoprostanes were

significantly inversely related in the grapefruit arm, but they were positively associated in

the control arm (test for waist-by-assignment interaction, p=0.004). There was also a

nonsignificant inverse association between male sex and F2-isoprostanes in the

grapefruit, but not the control, arm (test for sex-by-assignment interaction, p=0.015).

Discussion

In this first randomized trial assessing grapefruit consumption on biomarkers

associated with inflammation/oxidative stress and, more specifically, atherosclerotic

plaque production, we found that six weeks of daily grapefruit consumption did not

reduce sVCAM, hsCRP and F2-isoprostane levels. However, in a subsample of

participants with MetS, those who consumed grapefruit demonstrated marginally

significantly lower F2-isoprostane values compared to those assigned to the control

condition following the intervention.

Page 56: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

55

VCAM-1 is an adhesion molecule expressed by endothelial cells and vascular

smooth muscle cells in response to inflammatory/oxidative stimuli [80], and circulating

values correlate with risk of coronary death [81]. In vitro work also indicates that

flavonoids may attenuate inflammatory-induced adhesion molecule expression [82]. To

date, two studies have evaluated the effects of citrus flavonoids (in the form of orange

juice or its main flavonoid, hesperidin) on sVCAM-1 levels. Daily consumption of

orange juice or a hesperidin enriched beverage for four weeks nonsignificantly lowered

sVCAM-1 levels in overweight men [38]. In line with our results, a study of 3 week of

daily hesperidin supplementation (500 mg/day) in participants with MetS did not change

sVCAM-1 levels [36]. Our results are also supported by a similar study using high

polyphenol grape powder for 30 days wherein no change in sVCAM-1 levels in men with

MetS was demonstrated. In that trial other outcomes associated with endothelial health

did improve (e.g. flow mediated dilation, soluble intercellular adhesion molecule-1),

though these measures were not assessed in our study [83]. Epidemiological evidence

from NHS indicated that women with the highest flavonol intake had a 4% reduction in

sVCAM-1 levels compared to women in the lowest quintile of intake [39]. These data,

coupled with our own, suggest that sVCAM-1 levels are not highly responsive to short-

term exposures of flavonoid intake, but long term, habitual intake may promote healthy

sVCAM-1 values.

A significant body of evidence suggests that CRP is associated with risk of CVD

[84] [80]. CRP was once considered a pathologically insignificant hepatic marker of a

larger inflammatory cascade, but more recent evidence demonstrates that CRP is also

Page 57: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

56

produced by the endothelium in response to injury [80] and has proatherogenic properties

[2]. In the epidemiological NHS, Landberg et al. showed that grapefruit intake (≥1

serving/day) was inversely associated with CRP values [39], in contrast to findings of our

randomized, controlled trial that showed no significant reduction in CRP in response to

eating 3 servings/day for a period of six weeks. These inconsistent results may reflect

differential periods of exposure or could suggest our study was underpowered to detect a

significant change. [39]. Alternately, our dose may have been inadequate to induce short-

term changes in inflammatory markers. A recent randomized crossover study by Dalgard

et al. resulted in an 11% reduction in CRP values after four-week consumption of orange

and blackcurrant juices compared to a placebo drink in participants with peripheral artery

disease (PAD) [40]. Of note, a study in healthy adults who consumed 500 mL orange

juice for 14 days demonstrated a nonsignificant reduction in CRP [85]. These findings

suggest that the health status of the cohort may explain differential effects demonstrated

across the published studies. Patients with vascular diseases may benefit more from

consumption of a high polyphenol/flavonoid diet than overall healthy individuals.

F2-isoprostanes are a validated marker of lipid peroxidation, a key mechanism

leading to atheroma formation [86]. Previous studies have shown a reduction in plasma

F2-isoprostanes following orange juice consumption in healthy adults [85] as well as in

patients with PAD consuming orange and blackcurrant juices [40]. Our intervention

resulted in a nonsignificant reduction in F2-isoprostanes in those consuming grapefruit,

compared to a rise in F2-isoprostanes in the control participants, consistent with results

observed in the orange and blackcurrant juices study [40].

Page 58: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

57

Data from the Framingham Heart study indicate that visceral adiposity is highly

correlated with isoprostane values [13]. At baseline, WC was positively associated with

isoprostane values. Grapefruit consumption modulated this relationship and resulted in

an inverse association between baseline WC and F2-isoprostane values, a result not seen

in the control group. However we cannot ascertain if the reduction in isoprostanes was

directly due to grapefruit consumption or if it was an outcome secondary to WC

reduction, as we had previously shown a significant reduction in WC following six weeks

of grapefruit consumption [77].

Further, individuals with MetS have an increased WC and present with greater

levels of oxidative stress [87]. Thus, it is not surprising that the subpopulation of MetS

individuals consuming grapefruit seemed to be particularly protected from increases in

F2-isoprostanes compared to MetS participants in the control group. Though not

statistically significant, these results are particularly striking given the very small

subsample size.

This study has both strengths and limitations. This randomized controlled trial

evaluated the anti-inflammatory and antioxidant effects of ruby red grapefruit, a widely

available whole food, in overweight/obese individuals, a population that is subject to high

rates of CVD [15]. Outcomes were also evaluated in a subsample of participants with

MetS, a population with a 2 to 5-fold increased risk of CVD compared to metabolically

healthy controls [88]. The original trial was powered for change in weight [77], not

inflammatory/oxidative biomarkers. Nevertheless, grapefruit consumption did lead to

reductions in these markers compared to the control condition, suggesting these

Page 59: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

58

interventions should be tested in a larger sample before definitive conclusions can be

drawn. In our sample baseline sVCAM-1 values were more similar to those of a healthy

population [39], while previous studies in patients with MetS [83] or coronary artery

disease [81] show that circulating sVCAM-1 values may be as much as 2-3-times greater

in at-risk populations. This may explain why sVCAM-1 values did not change in relation

to the grapefruit intervention. A similar intervention in a high-risk population may be

beneficial given previous findings demonstrating sVCAM-1 responsiveness to flavonoids

[82, 89].

Atherosclerosis develops in response to multiple stimuli and signaling pathways

and the cardioprotective response to grapefruit could potentially be detected using a

variety of biomarkers. Here we evaluated biomarkers with the most robust preliminary

data in relation to the pathogenesis of atheroma formation and responsiveness to dietary

flavonoids. Overall, our results cannot support daily citrus and/or grapefruit to

significantly modify CVD risk associated with the selected biomarkers.

Coupled with previous research, our trial suggests future research in the field of

grapefruit/citrus consumption and cardioprotection should focus on populations at high

risk for cardiovascular disease, including MetS individuals, in a sufficiently large sample

size to test these important hypotheses.

Page 60: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

59

Tables

TABLE 5. Baseline characteristics of participants who successfully completed all study activities (n=69)

Total (n=69) Grapefruit (n=37) Control (n=32)

Age (y) 41.8±10.7 40.6±10.8 43.2±10.6

Female, n (%) 48 (69.6) 25 (67.6) 23 (71.9)

Non-Hispanic white

race/ethnicity, n (%) 43 (62.3) 23 (62.1) 20 (62.5)

Weight (kg) 91.5±13.9 92.1±15.0 90.8±12.8

Body mass index (kg/m2) 32.1±4.1 32.8±4.2 31.4±3.8

Waist circumference (cm) 103.0±10.2 102.8±10.7 103.3±9.7

Systolic blood pressure (mmHg) 119.7±16.5 120.5±17.7 118.7±15.2

Diastolic blood pressure (mmHg) 80.9±10.5 81.9±10.9 79.7±10.1

Metabolic Syndrome, n (%) 29 (42.0) 14 (37.8) 15 (46.9)

Blood Pressure not available for one participant: n=31 in control group. No differences were

found between groups at baseline for any measure.

Page 61: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

60

TABLE 6. Change in biomarkers (mean ± SD) associated with atherosclerotic plaque production in participants enrolled in a six-week

grapefruit feeding trial

2a. Entire cohort (n=69)

Biomarker

Grapefruit (n=37)

Control (n=32)

Difference

between arms

P-valueb

Pre Post

Paired t-test

p-valuea

Pre Post Paired t-

test p-valuea

sVCAM-1 (ng/mL) 627.6 ± 117.0 614.3 ± 122.5 0.114 623.0±125.1 610.5±110.5 0.261 0.987

hsCRP (mg/L) 2.2±1.5 2.1±1.5 0.596 2.4±2.0 2.8±2.0 0.136 0.073

F2-isoprostanes

(ng/mg creatinine) 14.1±7.7 12.4±6.4 0.092 14.7±6.5 15.9±9.0 0.452 0.063

2b. Participants with Metabolic Syndrome (MetS) at baseline (n=29)

Biomarker

Grapefruit (n=14) Control (n=15)

Difference

between arms

P-valueb

Pre Post

Paired t-test

p-valuea

Pre Post Paired t-

test p-valuea

sVCAM-1 (ng/mL) 665.5±133.7 650.1±144.8 0.540 603.1±86.3 594.1±93.6 0.303 0.840

hsCRP (mg/L) 2.3±1.7 2.0±1.0 0.380 3.1±1.5 3.1±1.6 0.966 0.106

F2-isoprostanes

(ng/mg creatinine) 14.3±6.7 12.0±4.5 0.239 15.0±6.9 18.3±10.9 0.257 0.058

No differences were found between groups at baseline for any biomarker. A definitive value for biomarkers was not determined in all

samples: sVCAM-1: n=36, n=30; hsCRP: n=36, n=30; F2-isoprostanes: n=34, n=32. aChange in biomarker within each arm using paired t-tests.

bDifference between arms post-intervention, adjusting for baseline value of biomarker, using linear regression.

Page 62: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

61

TABLE 7. Associations between common cardiovascular disease (CVD) risk factors and inflammatory/oxidative stress biomarkers

in participants enrolled in a grapefruit feeding trial (n=69)

Biomarker

CVD risk

factor

Predicted Effect on

Biomarker at

Baseline (95% CI)a Predicted change in biomarker (95% CI)

b P interaction

c

Control Arm Grapefruit Arm

sVCAM-1

(ng/mL) BMI (kg/m

2) −0.84 (-8.21, 6.54) −0.26 (-5.75, 5.23) −1.61 (-5.77, 2.55) 0.681

Waist (cm) −0.16 (-3.12, 2.79) −0.29 (-2.42, 1.83) −0.81 (-2.42, 0.81) 0.661

Age (y) 3.15 (0.49, 5.80)* −0.12 (-2.08, 1.84) 1.0 (-0.71, 2.71) 0.184

Sex (male) 26.47 (-89.89, 36.95) −36.17 (-6.16, 78.50) −2.41 (-34.95, 39.77) 0.161

hsCRP (mg/L) BMI (kg/m2) 0.15 (0.05, 0.25)* 0.02 (-0.11, 0.16) 0.02 (-0.08, 0.12) 0.662

Waist (cm) 0.06 (0.02, 0.10)* −0.03 (-0.08, 0.02) 0.02 (-0.02, 0.06) 0.220

Age (y) 0.01 (-0.02, 0.05) −0.02 (-0.06, 0.03) 0.003 (-0.032, 0.039) 0.486

Sex (male) 0.01 (-0.94, 0.92) -0.22 (-0.83, 1.23) 0.28 (-1.10, 0.54) 0.425

F2-isoprostanes

(ng/mg creatinine) BMI (kg/m

2) 0.10 (-0.33, 0.53) 0.50 (-0.29, 1.28) −0.20 (-0.59, 0.18) 0.076

Waist (cm) 0.19 (0.02, 0.36)* 0.32 (0.02, 0.63) −0.16 (-0.32, −0.01) 0.004

Age (y) 0.10 (-0.06, 0.26) −0.07 (-0.36, 0.22) −0.06 (-0.21, 0.10) 0.914

Sex (male) 2.16 (-5.95, 1.64) 5.78 (-12.30, 0.74) −2.68 (-0.75, 6.11) 0.015 aLinear regression models testing associations between risk factors and biomarkers in the entire cohort at baseline and

bwithin each

arm post-intervention, adjusting for baseline values. .

cLikelihood ratio test for interactions between intervention arm and each CVD risk factor on each biomarker, post-intervention.

*Significant association between risk factor and biomarker at baseline, p<0.05.

Page 63: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

62

CHAPTER 4

GRAPEFRUIT INCREASES POSTPRANDIAL INSULIN AND ATTENUATES

THE RISE IN F2-ISOPROSTANE AND IL-6 CONCENTRATIONS ASSOCIATED

WITH CONSUMPTION OF A HIGH-FAT, HIGH-CALORIE MEAL

Introduction

There is a large body of evidence indicating that each meal incites an inflammatory

response that promotes the development of atherosclerosis [42, 90]. Meal consumption

results in a postprandial period which lasts 4-8 hours, depending on meal size and

macronutrient composition. With the Westernization of food and dietary behaviors, meal

size and frequency have increased; thus, humans spend the majority of each day in the

postprandial state [44, 91]. The postprandial period represents a dynamic phase of metabolic

trafficking and transport of macronutrients, micronutrients, and phytochemicals [42], all of

which come into direct contact with the endothelium via vascular transport. As compared to

prudent meals low in fat and refined carbohydrates, a Western-style diet, high in refined

carbohydrates and fat, is associated with greater postprandial dysmetabolism (hyperglycemia

and hypertriglyceridemia) and is recognized as a pro-inflammatory and pro-oxidative pattern

of eating [92]. Due to the exaggerated metabolic and inflammatory perturbations caused by

this type of diet, a Western pattern of eating is considered causal in the development of

endothelial dysfunction and atherosclerotic plaques [92-95]. Further, spikes in glucose and

triglycerides induced by Western meal consumption that are repeated multiple times daily

may eventually contribute to an increased risk of coronary artery disease (CAD) [92].

Page 64: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

63

Ultimately, the purpose of studying the postprandial period is to determine how this

detrimental short, but repetitive exposure to metabolic dysregulation may be modified to

reduce cardiovascular disease (CVD) risk. One plausible approach would be to introduce

foods into the diet with some regularity that would attenuate the endothelial “damage”

promoted during the postprandial period. While data are limited, evidence from a few

postprandial studies indicate that consuming an antioxidant-rich food (e.g. orange juice,

blackcurrant juice, raisins) with a high-fat, high-calorie (HFHC) meal significantly reduces

the pro-inflammatory and pro-oxidative processes that foster atherosclerotic plaque

development induced by the rest of the meal [49, 50, 96].

Large epidemiological trials also suggest that citrus, specifically grapefruit, may

promote cardiovascular health [25, 39]. As a citrus fruit, grapefruit contains appreciable

amounts of the flavonoids naringenin and hesperitin. These bioactive compounds may

explain grapefruit’s cardioprotective properties as these flavonoids have been shown to exert

hypoglycemic, hypolipidemic, antioxidant, and anti-inflammatory properties in cell culture

[97], animal models [20, 98], and randomized human trials [35, 36, 38, 77].

While results from postprandial studies suggest that eating an antioxidant rich food

with an HFHC meal attenuates the oxidative/inflammatory stress induced by the meal and

improves endothelial function [38, 49-51], there are still significant gaps in this field of

research. The beneficial role of grapefruit as a cardioprotective food has been

demonstrated across the spectrum of studies from large population trials to cell culture

studies [20, 25, 35, 36, 38, 39, 52, 77, 97]. However, there has yet to be a trial examining

the acute, postprandial effects of grapefruit when consumed with an HFHC meal on

Page 65: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

64

biomarkers associated with cardiovascular health and disease in humans. Furthermore,

traditional American breakfast meals are typically high in calories and laden in fat, and

grapefruit is a common breakfast food. Thus, it is reasonable to study grapefruit

consumption with an HFHC breakfast meal, as it is a feasible meal option when

considering the human condition. Additionally, trials to date have failed to evaluate the

effects of any bioactive-rich food beyond a single meal. The purpose of this crossover,

double meal challenge, postprandial pilot trial was to evaluate the potential endothelial

protective role of grapefruit on HFHC meal-induced dysmetabolism, inflammation, and

oxidative stress in healthy, overweight and obese adults.

Methods

Participants. Five men and five women who participated in a 6-week grapefruit feeding

trial aimed at evaluating the impact of daily consumption of ruby red grapefruit (Texas

Rio Star Grapefruit, grown by Rio Queen Citrus; Mission, TX) on anthropometry, blood

pressure, lipids, and inflammation/oxidative stress [77] were enrolled in this ancillary

postprandial study. All volunteers completed the feeding trial at least two weeks prior to

participation in this study and abstained from consumption of citrus fruits and juices and

the use of anti-inflammatory drugs while enrolled in the postprandial study. Eligible

participants included healthy, nonsmoking men and premenopausal women with a body

mass index (BMI) between 25-45 kg/m2. Potential participants were excluded if they

reported a history of chronic conditions including cancer, inflammatory disease, hepatic

or renal disease, diabetes, hypercholesterolemia (>225 mg/dL), or heart disease.

Page 66: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

65

Individuals using medications to control lipids or inflammation or drugs metabolized by

the cytochrome P450-3A enzyme were excluded due to potential interactions with

grapefruit [78]. The University of Arizona Institutional Review Board approved this

study protocol, and all participants provided written informed consent.

Study design. On the day prior to the postprandial investigation, participants were asked

to abstain from vigorous physical activity and alcohol consumption. Participants reported

to the laboratory following a 12-hour overnight fast on two individual test days, separated

by at least one week. Upon arrival, a flexible indwelling catheter was fixed to the

antecubital region of one arm where it remained for the subsequent 8.5 hours, and a

fasting blood sample was collected. Participants were then given an HFHC breakfast

meal consisting of scrambled eggs, a biscuit with butter, and a sausage patty (740 kcal, 28

g protein, 48 g fat, 17 g saturated fat, 51 g carbohydrate, 2 g fiber), which was consumed

within 20 minutes. On test day one, all participants ate one ruby red grapefruit (68 kcal,

1 g protein, 0 g fat, 16 g carbohydrate, 2.5 g fiber) with the breakfast meal. Four hours

after breakfast a second HFHC meal was provided, which included a double

cheeseburger and French fries (670 kcal, 28 g protein, 34 g fat, 12.5 g saturated fat, 63 g

carbohydrate, 5 g fiber) and was consumed within 20 minutes. Participants were

permitted to drink water ad libitum for the duration of each test day.

In addition to the baseline blood collection, samples were taken 30 minutes and 2,

5, and 8 hours after consumption of the breakfast meal. Blood was collected into serum

Page 67: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

66

and heparin-coated plasma tubes and processed following standard procedures [77].

After centrifugation, samples were aliquoted into storage cryovials and stored at -80°C.

Biosample analysis. Blood samples from all time points collected on each test day were

analyzed for metabolic, inflammatory, or oxidative stress biomarkers. Glucose and

triglycerides were measured in plasma samples using colorimetric assay kits (Cayman

Chemical, Ann Arbor, MI). Insulin concentrations were quantified in plasma samples

using a monoclonal antibody enzyme linked immunosorbency assay (ELISA) kit (Dako

Ltd., Carpinteria, CA). sVCAM-1, sICAM-1, and IL-6 concentrations were measured in

serum samples via ELISA (R&D Systems, Minneapolis, MN). All samples for each

subject were analyzed in duplicate on the same plate, and inter- and intra-assay

coefficients of variation (CVs) were less than 10% for all analytes.

To measure F2-isoprostanes, plasma was mixed with 15% potassium hydroxide in

equal volumes and incubated at 40C for 60 minutes. The samples were then neutralized

by addition of three volumes of 1M potassium phosphate buffer to one volume of sample.

The prepared samples were purified by adding 3 volumes of ethanol to one volume of

sample, followed by vortexing and incubation at 4C for 5 minutes. Ethanol-sample

mixtures were centrifuged at 1500xg for 10 minutes. The supernatant was then decanted

and the ethanol was evaporated off using a centrifugal evaporator. Finally, F2-

isoprostanes were quantified in the purified samples using a competitive ELISA kit

(Cayman Chemical, Ann Arbor, MI). Inter- and intra-assay CVs were below 10%.

Page 68: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

67

Statistical Methods. Median biomarker concentrations (glucose, insulin, triglycerides,

IL-6, sVCAM-1, sICAM-1, and F2-isoprostanes) were compared between test days at

each time point using Wilcoxon sign-rank tests. The median area under the curve (AUC)

for each biomarker was calculated using the trapezoidal rule [99] for each participant on

each test day and compared between test days using Wilcoxon sign-rank tests. Findings

were verified using student’s t-tests. Wilcoxon rank-sum tests were used to evaluate the

relationship between CVD risk factors, such as BMI (overweight and obese) and gender

(male and female), and the AUC of each metabolic/inflammatory/oxidative stress

outcome. Spearman correlation coefficients were performed to evaluate the relationship

between age and the AUC of each outcome. Analyses were performed using Stata 12.1

(StataCorp, College Station, TX).

Results

Baseline demographics and compliance. Participants included five men and five women

with mean age of 47.5 ± 11.1 years. Participants had a mean BMI of 31.3 ± 2.6 kg/m2

(Table 8). All participants reported compliance with the dietary and activity restrictions

while enrolled in the trial, and all blood draws were completed according to study

protocol. However, only plasma, and not serum, was collected from one participant at

the final time point on both days; thus, final concentrations of IL-6, sVCAM-1, and

sICAM-1 were not available for this participant.

Page 69: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

68

Impact of HFHC meal consumption, with and without grapefruit, on cardiometabolic

biomarkers. As expected, no differences were detectable in baseline values of any

biomarkers between test days. Median insulin concentrations were significantly higher

30 minutes and 2 hours after HFHC+grapefruit (GF) consumption compared with HFHC

consumption alone (63.2 versus 24.2 μIU/mL, P=0.007 and 31.7 versus 23.2 μIU/mL,

P=0.025, respectively; Figure 3). Additionally, serum IL-6 concentrations were

significantly lower 30 minutes after the HFHC+GF meal compared with HFHC alone

(0.077 versus 0.862 pg/mL, P=0.017). Interestingly, median sVCAM-1 was significantly

higher at hour 5 following HFHC+GF compared with HFHC alone (498 versus 482

ng/mL, P=0.017). HFHC+GF meal consumption resulted in lower F2-isoprostane values

after 5 hours versus HFHC alone (45.5 versus 77.0 pg/mL, P=0.013), though no

significant differences were observed by 8 hours. Furthermore, glucose, triglyceride, and

sICAM-1 concentrations were not significantly different at any time point between test

days.

There were no differences in median AUC for most biomarkers between test days

(Table 9). Median AUC insulin concentration was higher following HFHC+GF meal

consumption as compared to the HFHC alone (371.5 versus 247.7 μIU/mL, P=0.059).

Conversely, F2-isoprostanes were lower when the meal was consumed with grapefruit as

compared to when the meal was consumed without grapefruit (403.7 versus 556.0 pg/mL,

P=0.114), though this difference was not statistically significant.

Page 70: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

69

Cardiometabolic biomarkers and potential confounders. Finally, the relationship

between the AUC of each biomarker and risk factors associated with CVD were

evaluated. BMI category (overweight versus obese) was associated with AUC IL-6

values. When the HFHC+GF meal was consumed, the median AUC IL-6 was higher in

overweight than obese individuals (19.3 versus 2.8 pg/mL, P=0.087). Likewise, when

the meal was consumed alone, the median AUC IL-6 was higher in overweight than

obese individuals (18.1 versus 7.3 pg/mL, P=0.139). All other biomarker outcomes were

not different based on BMI status, and none of the biomarkers were associated with

gender (data not shown). Spearman’s correlation coefficients were calculated for the

relationship between age and AUC of each biomarker. F2-isoprostane values were

inversely correlated with age on both test days (HFHC: rho= -0.585, P=0.080;

HFHC+GF: rho= -0.659, P=0.039), though no significant correlations were found

between age and other biomarkers (data not shown).

Discussion

In this first double meal, postprandial trial evaluating the effects of grapefruit on

endothelial health, grapefruit consumption resulted in a significant increase in insulin and

a reduction in IL-6 during the first two hours following a meal, and a reduction in F2-

isoprostanes following the second, pro-inflammatory meal. No differences were detected

in glucose, triglycerides, or sICAM-1 when an HFHC meal was eaten alone or with

grapefruit, while sVCAM-1 values were higher following HFHC meal consumption with

grapefruit.

Page 71: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

70

Insulin is well characterized for its role in glucose metabolism. Insulin signaling also

plays a vital role in the maintenance of endothelial health and vasoreactivity as it induces

nitric oxide (NO) production via activation of the phosphatidylinositol-3 kinase-protein

kinase B-endothelial NO synthase (PI3K-PKB-eNOS) pathway [100]. Insulin resistance

leads to the loss of vascular tone and endothelial reactivity and ultimately increases the

risk of endothelial dysfunction and atherogenesis [100]. In this study, serum insulin

concentrations were significantly higher 30 minutes and two hours after eating the HFHC

meal with grapefruit than when the meal was eaten alone. This response is considered

favorable in that it reflects an immediate and necessary biological response to maintain

glucose homeostasis within cells. Ghanim, et al. observed a similar pattern in

postprandial insulin concentrations in a study assessing the effects of HFHC meal

consumption with orange juice. Insulin concentrations were significantly higher in

response to an HFHC meal+orange juice (OJ) compared with an HFHC meal consumed

with an isocaloric glucose beverage [49]. Interestingly, in the Ghanim trial, beverages

were matched for glucose intake (75 g), though one-hour postprandial glucose values

were suppressed following HFHC+OJ consumption compared with HFHC alone. In the

current study, participants consumed an extra 16 g carbohydrate with the HFHC+GF

meal, though circulating glucose values were not significantly different between test days

at any time point, suggesting the grapefruit supported greater insulin sensitivity in

response to an HFHC meal. These results also suggest that citrus foods may influence

beta cell secretion of insulin, thus increasing glucose uptake in the cells and reducing

postprandial blood glucose concentrations, though we cannot ascertain if the observed

Page 72: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

71

increase in serum insulin concentrations had any direct effect on endothelial function. It

is possible that the hyperinsulinemic response to the HFHC+GF diet may have been due

to the extra 16 g of carbohydrate delivered by the grapefruit and that the results observed

here were not due to activity of grapefruit-specific flavonoids. However, 16 g is a

relatively modest carbohydrate load, and may not have had any notable effect on insulin

secretion. Further exploration is warranted given that this immediate hyperinsulinemic

response to citrus in the postprandial period has now been demonstrated in two studies.

Our results related to postprandial inflammatory response are intriguing. Evidence

indicates that flavonoids and other polyphenols reduce inflammatory processes linked to

CVD, in part by reducing IL-6 [101, 102], a cytokine known to promote inflammatory

processes in atherosclerotic plaques and vascular smooth muscle cells [103]. IL-6 was

significantly reduced two hours after HFHC+GF meal consumption in this study. These

findings are supported by previous studies, which have demonstrated reductions in IL-6

in response to high-fat meals consumed with tomatoes [104] or strawberries [49, 105].

Surprisingly, IL-6 concentrations were also nonsignificantly higher in overweight

compared to obese individuals in this study. These findings disagree with reports that

show that IL-6 is positively associated with BMI and waist circumference in men and

women [106, 107] and may suggest an adaptive response in people with prolonged

exposure to adipokine-associated inflammatory response. It is also possible that this

difference was observed by chance due to the small sample size. Of note, IL-6 was more

responsive to grapefruit in obese individuals as compared to overweight subjects wherein

IL-6 was relatively unchanged in response to the addition of GF to the meal. These

Page 73: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

72

findings may suggest a potential underlying inflammatory pathology in overweight

individuals that is less responsive to dietary change.

Adhesion molecules are intrinsically involved in the promotion of endothelial

dysfunction and atheroma formation, as they facilitate pavementing of monocytes to the

endothelium [108]. sICAM-1 is produced by the monocytes themselves, whereas

sVCAM-1 is expressed by the endothelium. The majority of work evaluating endothelial

responses during the postprandial period has focused on the effects of dietary fat, and

numerous reports indicate that these adhesion molecules are responsive to varying dietary

fat loads and types in the postprandial period [109-112]. In this study, grapefruit

consumption did not significantly impact the effect of an HFHC meal on sICAM-1

values. Very little research has evaluated sICAM-1 or sVCAM-1 in response to a

flavonoid or polyphenol-rich food in the postprandial period, though a study with a

similar design in overweight individuals indicate that neither adhesion molecules were

significantly lower when a high-fat meal was consumed with polyphenol-rich raisins

[96]. Interestingly, in our study, significantly higher levels of sVCAM-1 were observed

five hours post breakfast meal + GF consumption compared to the HFHC meal. The few

studies that have evaluated the postprandial effects of citrus (in the form of orange juice)

have not evaluated sVCAM-1 [49, 95, 113], limiting the ability to make comparisons.

However, in vivo work suggests that flavonoids can inhibit inflammatory-induced

sVCAM-1 expression [82, 114], though the effect of citrus flavonoids, specifically, has

not been evaluated. Given the evidence that suggests that citrus is cardioprotective and

promotes endothelial health, it is possible that the effects on sVCAM-1 were observed by

Page 74: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

73

chance due to the small sample size. Nevertheless, these results indicate that grapefruit is

unlikely to significantly decrease sICAM-1 or sVCAM-1 in response to HFHC meal

feeding.

Flavonoids benefit biological systems through a variety of functions, though the most

well-known and characterized mechanisms are through antioxidant activities [42, 115].

Lipid peroxidation is particularly important in the context of endothelial dysfunction, as

oxidized lipids or lipoproteins are often involved in the primary insult to the endothelium,

thus propagating inflammatory processes and eventual atheroma formation [6, 44].

Previous postprandial evaluations of the effect of flavonoid-rich foods such as

strawberries or cocoa on oxidative stress have suggested that these foods are effective at

reducing lipid peroxidation (oxidized low-density lipoprotein or F2-isoprostanes) induced

by meal consumption [48, 116]. In this study, the AUC of F2-isoprostanes was

marginally lower when the HFHC meal was eaten with grapefruit versus alone, and F2-

isoprostanes were significantly lower 5 hours after HFHC meal consumption with

grapefruit. Importantly, this reduction in F2-isoprostanes was apparent after the second

meal was consumed in this double meal challenge, suggesting an increased serum

antioxidant capacity that accumulated during the first four hours of the challenge. In

agreement with the results shown here, a 3-hour postprandial study in 16 healthy young

adults resulted in higher serum antioxidant capacity and reduced lipoprotein oxidation

following intake of a citrus flavonone mixture or orange juice compared with a placebo

beverage [52]. Ghanim, et al. observed reductions in p47phox

expression, nuclear factor

κB binding, and reactive oxygen species generation in polymorphonuclear cells following

Page 75: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

74

HFHC meal +OJ compared with meal alone [49], indicating that, postprandially, citrus

constituents may suppress cellular mechanisms involved in oxidative stress. The data

shown here, taken in consideration with previous work, suggest that citrus foods improve

postprandial oxidative metabolism, and these benefits may extend beyond a single meal.

There are strengths and limitations of this study. This is the first trial to evaluate

grapefruit as a cardioprotective dietary agent in the postprandial period, despite

epidemiological and basic science studies that indicate that grapefruit may be an

important constituent of a heart healthy diet. Additionally, to our knowledge, this is the

first study to evaluate a flavonoid or polyphenol rich food associated with cardiovascular

health in a double meal challenge design. The findings of this trial suggest that grapefruit

consumption with a HFHC meal may modify insulin and IL-6 response nearly

immediately, while the antioxidant effects of the fruit may be observed after a second

HFHC challenge. However, this trial had a small sample size, which likely explains the

nonsignificant results in some of the outcomes. Furthermore, there was wide variability

in response to the meals, and a larger sample size may resolve this issue. Regardless,

significant outcomes of increased insulin concentrations and reduced IL-6 and F2-

isoprostanes are particularly robust.

In conclusion, the results of this trial suggest that grapefruit consumption with an

HFHC meal modifies some of the selected biomarkers associated with endothelial health

and disease. These findings, coupled with previous research of the postprandial response

to citrus fruits and their flavonoids, highlight the role of citrus as a potential vascular

protective food. These results also underscore the need for studies with larger samples

Page 76: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

75

and the potential to study cellular processes in order to elucidate the exact mechanisms by

which citrus benefits the cardiovascular system.

Page 77: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

76

FIGURE 3. Concentrations of various metabolic, inflammatory, and oxidative stress markers

following consumption of a HFHC meal alone or with ruby red grapefruit

Page 78: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

77

Tables

TABLE 8. Baseline characteristics of participants who completed the grapefruit double meal

challenge postprandial trial.

Subject Gender Race Age (y) Weight (kg) BMI (kg/m2)

1 female H 40.5 76.8 30.9

2 female NHW 48.5 90.5 27.0

3 male NHW 53.0 102.3 27.9

4 female H 47.0 83.1 29.1

5 male NHW 40.5 93.5 32.4

6 female NHW 53.0 77.5 30.3

7 male AA 29.0 109.2 33.7

8 male NHW 70.5 104 33.6

9 male H 40.5 105.2 34.0

10 female NHW 52.0 91.8 33.7

Total 50% female 60% NHW 47.5 ± 11.1 93.4 ± 11.7 31.3 ± 2.6

Abbreviations: BMI, body mass index; H, Hispanic; NWH, non-Hispanic white;

AA, African American. Totals for age, weight, and BMI represent mean ± SD.

TABLE 9. Comparison of AUC of biomarkers in response to a HFHC meal eaten alone or with

grapefruit (n=10)

Biomarker Test day Median AUC IQR P value

Glucose (mg/dL) HFHC 809.1 (680.7, 867.1)

0.575 HFHC+GF 777.1 (717.8, 881.5)

Insulin (μIU/mL) HFHC 247.7 (176.9, 445.2)

0.059 HFHC+GF 371.5 (189.4, 493.5)

Triglycerides (mg/dL) HFHC 179.9 (716.4, 2102.5)

0.800 HFHC+GF 1393.6 (1087.7, 1501.6)

IL-6 (pg/mL) HFHC 9.34 (1.52, 18.13)

0.721 HFHC+GF 10.70 (2.22, 19.33)

sVCAM-1 (ng/mL) HFHC 3851.1 (3668.0, 4209.5)

0.139 HFHC+GF 3909.3 (3743.9, 4097.9)

sICAM-1 (ng/mL) HFHC 1219.0 (1074.9, 1324.5)

0.959 HFHC+GF 1256.9 (1072.3, 1350.3)

F2-isoprostanes

(pg/mL)

HFHC 556.0 (401.1, 758.3) 0.114

HFHC+GF 403.7 (344.1, 403.7)

Abbreviations: AUC, area under the curve; HFHC, high-fat, high-calorie; GF, grapefruit;

IQR, interquartile range

Page 79: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

78

CHAPTER 5

SUMMARY AND CONCLUSIONS

The current research indicates a beneficial effect of daily ruby red grapefruit

intake on risk factors for CVD such as reducing central adiposity, blood pressure, various

parameters of the lipid profile, and, potentially, inflammation and oxidative stress. Six-

week consumption of grapefruit resulted in significant reductions in waist circumference,

total cholesterol, LDL-cholesterol, systolic blood pressure compared to baseline values.

Additionally, marginally significant reductions in hsCRP and F2-isoprostanes were

observed compared to the control condition, post-intervention. Grapefruit consumption

with an HFHC meal reduced IL-6 and F2-isoprostane concentrations in the postprandial

period, while insulin levels were significantly higher during the eight-hour postprandial

period after HFHC+GF consumption (Figure 4, pg. 97).

Obesity and Visceral Adiposity

Obesity is not only a disease in and of itself, but also significantly increases the

risk of cardiovascular events and, together, obesity and CVD impose a substantial burden

on individual and community health [117]. The literature suggests that modest weight

loss of 5-10% results in cardiovascular benefit [6, 117], in part by promoting vasodilation

and endothelial health [6]. Additionally, the favorable effects of weight loss on

endothelial health can be observed in as little as six weeks [118]. In the present study,

those randomized to daily grapefruit consumption for six weeks demonstrated a slight

Page 80: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

79

reduction in weight, though this effect was not statistically significant. Based on self-

reported energy intake, grapefruit did not promote appetite suppression and lead to a

reduction in energy consumption, as has been hypothesized [18]. Previous research

evaluating grapefruit as a weight loss food has yielded conflicting results. A four-arm

study comparing whole grapefruit, grapefruit juice, a grapefruit capsule, and a placebo

resulted in weight loss of 1.6 kg, 1.5 kg, 1.1 kg, and 0.3 kg, respectively [19]. A

significant difference in weight loss was detected between those randomized to the fresh

grapefruit group and those in the placebo group [19]. However, another study comparing

preloads of fresh grapefruit, grapefruit juice, and water in addition to 12.5% calorie

restriction over twelve weeks showed non-significant differences in weight loss between

groups [18], indicating that grapefruit had no further effects on promoting weight loss

than calorie restriction alone. Taken together, the results from the present trial and

previous research suggest that grapefruit is unlikely to stimulate weight loss robust

enough to promote metabolic or vascular health.

High VAT volume, as measured by waist circumference, is also a major risk

factor for cardiovascular disease [2, 117], and is more predictive of CAD than BMI [8].

Visceral fat secretes a slew of pro-atherogenic molecules that are known to activate

endothelial cells [2, 6], and reductions in waist circumference may result in more

beneficial effects on vascular health than overall weight loss alone [6, 7, 117]. In this

six-week grapefruit feeding trial, waist circumference and waist-to-hip ratio were

significantly reduced in those consuming grapefruit compared to baseline values, though

these results were not significantly different from those randomized to the control

Page 81: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

80

condition. Conversely, studies by Fujioka, et al. and Silver, et al. showed no effect of

grapefruit consumption on waist circumference in obese adults [18, 19].

Cell culture and animal model studies that have explored the effects of citrus

flavonoids on lipolysis may explain the findings observed in our study. Studies in Long

Evans Hooded rats and LDL-Receptor (LDL-R) -/- mice fed high fat diets supplemented

with naringenin found reductions in parametrial adipose tissue volume [20] and reduced

adipocyte size [33] compared to control conditions. The exact mechanisms that may

have caused these results were not explored. Some research suggests that citrus

flavonoids impact the cAMP cascade, which may further explain the results observed in

our trial and in the aforementioned rodent studies. cAMP activates protein kinase A

(PKA), which in turn activates lipases, whereas phosphodiesterases (PDE) breakdown

cAMP, thus preventing lipolysis. Human adipocytes (ex vivo) treated with SINETROL (a

flavonoid-rich supplement made of an extract of the juice, peels, and seeds of blood,

sweet, and bitter oranges and grapefruit) demonstrated increased lipolysis and free fatty

acid release due to SINETROL inhibition of cAMP-PDE [57]. These results suggest that

citrus flavonoids may promote lipolysis by promoting activity of the cAMP signaling

cascade.

Vascular Reactivity and Blood Pressure

Hypertension is one of the most important contributing factors for atherosclerotic

CVD risk, and a recent American Heart Association: Heart Disease and Stroke Statistics

2013 Update indicates that 33% of American adults (78 million) are hypertensive [119].

Page 82: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

81

Lifestyle interventions that promote a diet rich in fruits and vegetables, such as the

Dietary Approaches to Stop Hypertension (DASH) plan are effective at promoting

systolic and diastolic blood pressure reduction of 6 and 3 mmHg, respectively, in pre-

hypertensive adults [120]. These results are comparable to use of angiotensin converting

enzyme (ACE) inhibitors, one of the primary pharmacotherapies utilized for treatment of

pre-hypertension or hypertension [120]. In this trial, daily consumption of grapefruit

resulted in a significant reduction in SBP of 3 mmHg, compared to baseline values.

Though these results are not as pronounced as that seen when following the DASH diet,

they do suggest that a simple dietary change can significantly improve blood pressure.

Of note, though this population had an average BMI categorized in the obese range, their

baseline blood pressure averaged 119.1/80.6, indicative of borderline pre-hypertension.

While the reduction in SBP was significant, this effect may be more marked in a

hypertensive population, though this hypothesis will need to be prospectively tested.

To our knowledge, this is the first study showing a reduction in blood pressure as

a result of grapefruit intake. However, studies in grapefruit bioactives or similar citrus

fruits have indicated blood pressure benefit and explored potential mechanisms

explaining these effects. Daily supplementation of 500 mg hesperidin for three weeks in

patients with MetS resulted in a significant improvement in flow-mediated dilation

(FMD) of the brachial artery, a measure of endothelial reactivity [36]. Furthermore, a

study exploring the role of orange juice (OJ) compared to a control drink with added

hesperidin (CDH, both containing approximately 292 mg hesperidin) or a control drink

alone resulted in reduced diastolic blood pressure after four week daily consumption of

Page 83: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

82

OJ or the CDH as well as improved postprandial microvascular endothelial reactivity

compared to the control drink [38].

The results from cell culture and animal model trials may explain the effect of

citrus on endothelial function and promotion of healthy blood pressure. Acute hesperitin

treatment of bovine aortic endothelial cells (BAEC) resulted in eNOS phosphorylation,

thus promoting NO production in endothelial cells [36], the primary mechanism involved

in vasodilation. Hesperidin consumption in our population was extremely low (1.57

mg/day) compared to the previously mentioned studies on hesperidin and vascular

function [36, 38]. Due to their similar structure, it is plausible that naringenin induces

similar results on these various enzymes, though there has yet to be a study testing this

hypothesis. The mechanisms underlying the results observed here and in other studies

may be due to naringenin’s effect on phosphodiesterases. Phosphodiesterases are

involved in the degradation of cGMP, a molecule that inhibits calcium influx into the

vascular smooth muscle cell, and ultimately inhibits vasoconstriction. Similar to the

effects of SINETROL on cAMP-PDE, naringenin (0.1mM) treatment of rat aortic

myocytes resulted in inhibition of cGMP-PDE, thus promoting vasodilation [21].

Though the concentration of naringenin explored in the aforementioned study is

unattainable by the human diet, even in our population who consumed a great deal of

naringin (146.2 mg/day), this evidence may explain the mechanism by which grapefruit

consumption elicited its blood pressure lowering effects in our population. Additionally,

intake of Vitamin C, a potent superoxide scavenger, also significantly increased in those

consuming grapefruit, as expected. Superoxide, a reactive oxygen specie (ROS), couples

Page 84: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

83

with NO to form peroxynitrite, leading to the uncoupling of the eNOS homodimer and

subsequently preventing NO formation [67]. As a scavenger of superoxide, Vitamin C

prevents peroxynitrite formation and the uncoupling of eNOS, and promotes

vasorelaxation and the maintenance of a healthy endoethelium [66, 67]. Therefore, the

proposed roles of Vitamin C, naringenin, and hesperidin in the endothelium may

collectively explain the effect of grapefruit consumption on blood pressure as observed in

this study.

Lipid Response

An important aspect of cardiometabolic health that contributes heavily to CVD

risk is dyslipidemia. Elevated levels of TC, LDL-C, triglycerides, and suppressed HDL-

C are all associated with risk of CAD [117]. LDL and triglyceride-rich lipoproteins both

facilitate endothelial dysfunction and atheroma formation by activating the endothelial

cells, disrupting the endothelial layer, and migrating into the subendothelial space [121].

Thus, reducing TC, LDL-C, and triglyceride values are important targets in CVD risk

reduction.

TC decreased in both the grapefruit and control groups, though a more marked

reduction was observed due to grapefruit intake (approximate reductions of 12 mg/dL and

7 mg/dL, respectively). These results are interesting given that the baseline values of TC

were already within a normal range in this sample. A striking decrease in LDL-C of

approximately 19 mg/dL was observed in those consuming grapefruit, which is of

particular significance given the short time frame of the study. The effects of 8-week

Page 85: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

84

naringin supplementation (400 mg/day) resulted in a 14% reduction in TC and a 17%

reduction in LDL-C in hypercholesterolemic patients, while those with healthy

cholesterol values observed no benefit [68]. A differential effect of the type of grapefruit

consumed was found in patients with coronary atherosclerosis. Those consuming either

one ruby red or one white grapefruit (which differ in lycopene and flavonoid content)

daily for thirty days demonstrated reductions in both TC and LDL-C. Only those eating

ruby red grapefruit observed reductions in triglycerides [34], suggesting an increased

benefit of consuming the ruby red cultivar, though no change was observed in

triglycerides in this trial. In spite of the positive results in many studies, four-week

supplementation of 800 mg/day or 500 mg/day of hesperidin or naringin, respectively,

revealed no benefit in any lipid parameter in mildly hypercholesterolemic adults [35].

This may suggest that the bioactives in grapefruit need to be consumed simultaneously.

Additionally, fiber intake did not change in our population, indicating that the role of

grapefruit in lipid control was likely a result of the bioactives and potentially other

micronutrients, and not the fiber content of the fruit.

Insulin Response

Insulin is well characterized for its essential role in glucose metabolism. In the

last twenty years, research has shed light on the effect of insulin’s signal transduction

mechanisms that impact cardiometabolic health outcomes [100]. Insulin signaling is of

particular importance in hepatic tissue, and insulin sensitivity promotes healthy lipid

metabolism by inhibiting microsomal triglyceride transfer protein and subsequent

Page 86: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

85

apolipoprotein-(apo)B100 lipoprotein assembly and secretion [122]. Insulin is also

intrinsically involved in maintenance of vascular health through signaling of the PI3K-

Akt-eNOS cascade. Activation of this pathway leads to vasodilation and also promotes

cardiovascular cell survival, reduces inflammatory processes, and inhibits oxidative stress

[100]. Thus, insulin sensitivity is an important goal for cardiometabolic disease risk

reduction.

Pre-clinical and in vitro studies suggest that citrus flavonones improve insulin

sensitivity. Naringenin increases hepatic expression of PPAR-γ and -α [20, 31, 123],

receptors involved in glucose homeostasis and lipid metabolism, and activation of which

improves insulin sensitivity [123]. In a study of LDL-R -/- mice fed a high fat, high

cholesterol diet, naringenin supplementation reduced fasting insulin values, as well as

reduced plasma glucose levels in a 60-minute glucose tolerance test, suggesting enhanced

insulin sensitivity [33]. Male Wistar rats fed a high fat diet developed hyperinsulinemia,

insulin resistance, and hyperglycemia, though naringin supplementation significantly

reduced all of these deleterious metabolic outcomes [123]. However, in the only clinical

trial to evaluate the effects of grapefruit on parameters associated with metabolic

(dys)function, 12-weeek grapefruit consumption in pill, juice, or whole fruit form had no

significant impact on fasting insulin concentrations [19].

Insulin was not evaluated before and after the six-week grapefruit feeding trial,

though the effects of acute grapefruit consumption on the postprandial insulin response

were assessed. Fasting insulin concentrations were not different between test days,

though addition of grapefruit to a HFHC meal resulted in elevated serum insulin

Page 87: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

86

concentrations 30 minutes and 2 hours post feeding. Of note, these effects were no

longer present after the second HFHC meal was consumed, indicating an immediate

effect of grapefruit on insulin secretion. These results are supported by a study by

Ghanim and colleagues, in which a HFHC meal was consumed with OJ, a glucose

beverage, or water. HFHC+OJ resulted in elevated postprandial insulin values compared

to the other two treatments, though glucose values were suppressed [49]. The results of

both of these trials suggest that citrus fruits or the bioactives therein induce pancreatic

beta cell secretion of insulin and concomitant tissue uptake of glucose. This hypothesis is

supported by a study in which pancreatic histology of male Wistar rats fed a high fat diet

supplemented with naringin for 28 days revealed reduced injury of islet cells and

increased numbers of insulin secretory granules [123]. Importantly, this effect was

observed after chronic naringin supplementation and at levels unattainable by the human

diet, limiting the ability to compare the results from this animal study and the human

postprandial condition. Nevertheless, these results, taken in consideration with the results

from the present trial and the Ghanim, et al. trial suggest that citrus flavonones may

preserve beta cell function in the context of a high fat diet.

There are inconsistencies in the literature regarding the effect of dietary

flavonoids on insulin secretion, as postprandial evaluations have demonstrated reduced

insulin concentrations in response to flavonoid-rich berries consumed with a high fat

meal [105] or alone [124]. Conversely, black tea consumed with a 75 g glucose load and

cranberry juice both elicit a hyperinsulinemic response within 60 or 90 minutes of

consumption, respectively, compared to control conditions (caffeinated beverage or a

Page 88: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

87

high fructose beverage, respectively) [125, 126]. Of note, the postprandial evaluations of

insulin response to dietary flavonoids to date have all been performed in healthy adults

with no history of cardiovascular diseases or diabetes. In order to elucidate the effects of

dietary flavonoids on postprandial insulin concentrations, similar evaluations should be

completed in a population with known insulin resistance.

Inflammatory and Oxidative Stress Modification

Oxidative stress and inflammation are two of the key underlying causes of CVD,

and obese individuals, particularly those with central obesity, present with high

circulating values of inflammatory and oxidative stress markers [6, 127]. Obesity is

characterized by chronic, low-grade inflammation/oxidative stress and metabolic,

oxidative, and inflammatory pathways are heavily interconnected and function in a

reciprocal, positive feedback fashion, often leading to dysregulation of one another [127].

These pathways converge at the endothelium and induce endothelial dysfunction,

characterized by increased activity of proatherogenic factors [127], including

inflammatory cytokines, adhesion molecules, and ROS, amongst others [2, 6].

Inflammatory Cytokines

IL-6 and CRP are inflammatory cytokines involved in the promotion and

progression of atherosclerotic plaques [103]. IL-6 is secreted by adipocytes, immune

cells, and endothelial cells and induces hepatic CRP synthesis [2, 106]. Previous reports

indicate that CRP and IL-6 are detectable in atherosclerotic lesions, and both are involved

Page 89: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

88

in endothelial cell-induced expression of adhesion molecules, and other pro-inflammatory

cytokines [80, 128]. Further, CRP induces secretion of monocyte chemoattractant

protein-1 (MCP-1) by endothelial cells [129], whereas IL-6 stimulates macrophages to

secrete MCP-1 [130], thus facilitating the homing of monocytes to the endothelial cells

(Figures 1 & 4).

In this study, six-week daily consumption of ruby red grapefruit resulted in

marginal reductions in hsCRP compared to the control condition, an effect that was more

pronounced (though not statistically significant) in individuals with MetS. These

nonsignificant results are likely due to the small sample size in this study, and may be

significant in a study adequately powered to demonstrate changes in CRP. In the acute

setting, HFHC meal consumption with grapefruit resulted in 2-hour post meal reductions

in IL-6 compared to HFHC alone. However, this effect was short-lived, suggesting that

anti-inflammatory foods may need to be consumed at regular time intervals in order to

prevent increases in IL-6 concentrations. No human clinical trial has evaluated the

potential role of grapefruit in attenuation of inflammatory cytokines. There is extensive

evidence that supports naringenin’s ability to prevent inflammatory processes. Ex vivo

work indicates that naringenin treatment of cultured human macrophages reduces

secretion of IL-6 [131]. Additionally, IL-6 production and secretion was reduced by

dietary naringenin supplementation in diabetic mice [132]. Basic science evaluations

have repeatedly demonstrated naringenin’s ability to prevent nuclear factor kappa B

(NFκB) expression and activity in both rodent [123, 132] and cell culture models [133,

134]. NFκB is a transcription factor that promotes the transcription of multiple

Page 90: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

89

inflammatory molecules (including IL-6 and CRP), oxidative stress enzymes, and

adhesion molecules (including ICAM-1 and VCAM-1). Therefore, the potent anti-NFκB

effects of naringenin may explain the responses of CRP and IL-6 to grapefruit in this

study.

Adhesion Molecules

Adhesion molecules are intrinsic in the pathogenesis of atherosclerotic plaque

development. Endothelial cells express adhesion molecules in response to inflammation,

oxidative stress, or endothelial injury and mediate the binding of circulating monocytes to

the endothelium [6, 81]. ICAM and VCAM are the most well characterized of the

adhesion molecules, and soluble values correlate with risk of coronary death [81].

Studies in rabbits indicate that naringin or naringenin supplementation of a high

cholesterol diet results in lower circulating values of both sICAM-1 [135] and sVCAM-1

[136]. Additionally, hesperitin treatment of BAEC resulted in reduced sVCAM-1

expression in response to TNF-alpha stimulation [36], all of which suggests that citrus

flavonones have a role in promoting endothelial health by suppressing adhesion molecule

expression.

In this study, sVCAM-1 levels were not modified by six weeks of daily ruby red

grapefruit consumption. In a study of overweight men, sVCAM-1 levels were

nonsignificantly lower following 4 weeks of orange juice or a hesperidin enriched

beverage compared to placebo [38]. Of note, sVCAM-1 levels in our sample were

similar to that of healthy individuals [39]. Previous evaluations in participants with MetS

Page 91: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

90

[83], pre-hypertension [38] or CAD [81] have shown that sVCAM-1 levels may be as

much as 2-3 fold higher in at-risk populations, which could explain why sVCAM-1 levels

were not modified in this trial. Furthermore, 3 weeks of hesperidin supplementation (500

mg/day) in MetS individuals resulted in no change is sVCAM-1, though E-selectin values

were reduced compared to placebo [36], suggesting that E-selectin may be more

responsive to citrus flavonoids than sVCAM-1.

In the acute setting, sICAM-1 levels were not affected by grapefruit, whereas

sVCAM-1 levels were significantly higher five hours post HFHC+GF feeding compared

to HFHC alone. Postprandial evaluations of citrus intake have not assessed these

adhesion molecules, though previous work indicates that both sICAM-1 and sVCAM-1

are responsive to various types of dietary fat in the postprandial period [110-112]. The

response of sVCAM-1 to HFHC+GF meal consumption was unexpected, but may have

happened by chance simply due to the small sample size. Also, it is important to

recognize that, like the individuals studied in the larger feeding trial, this sample

presented with relatively low fasting adhesion molecule concentrations. Therefore, the

slight increase in sVCAM-1 values 5 hours post HFHC+GF meal may not be

pathologically significant. However, the results from both the six-week and postprandial

feeding trials indicate that ruby red grapefruit intake is unlikely to significantly modify

circulating sVCAM-1 and sICAM-1 values.

Page 92: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

91

Lipid Peroxidation

Free radicals and ROS are involved in the development and progression of

atherosclerosis due to activation of inflammatory processes, cellular remodeling, and

lipid peroxidation [86, 116]. F2-isoprostanes are the result of non-enzymatic free-

radical-mediated lipid peroxidation of polyunsaturated fatty acids [116, 137], and are

validated markers of lipid peroxidation [86]. F2-isoprostanes can be released from cell

membranes via phospholipases and circulate freely in the blood, though isoprostanes are

also found associated with lipoproteins [86, 137]. Regardless of whether they are

transported in the blood freely or bound to lipoproteins, isoprostanes can interact with the

endothelium and promote endothelial dysfunction. Flavonoids are known for their free-

radical scavenging behavior [116], and flavonoid-rich foods have been shown to reduce

lipid peroxidation [116].

In this trial, six weeks of grapefruit consumption resulted in nonsignificant

reductions in F2-isoprostanes, and this reduction was more pronounced in participants

with MetS. Previous work indicates that orange juice consumption in a healthy

population [85] or those with peripheral artery disease [40] results in reductions in F2-

isoprostanes, though this is the first study to measure F2-isoprostanes in response to

grapefruit, specifically.

In the postprandial evaluation, F2-isoprostane values were significantly lower five

hours post breakfast meal (1 hour post lunch meal) consumption. In a study by Gopaul

and colleagues, F2-isoprostanes increased 2 hours post meal consumption in nine healthy

adults who ate a single fast-food meal, similar to what was consumed in this study, [137].

Page 93: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

92

However, samples were not collected longer than 2 hours postprandially, limiting the

ability to compare the studies further. Nevertheless, the Gopaul study and this trial

indicate that F2-isoprostanes are sensitive to dietary changes in the postprandial period.

This is the first postprandial evaluation to measure F2-isoprostanes in response to citrus

consumption, though previous work indicates that orange juice consumption improves

serum antioxidant capacity [52]. The results from the studies conducted here along with

previous work on orange juice consumption suggest that regular citrus intake reduces

lipid peroxidation in an acute setting and potentially in response to chronic intake. The

results from this trial also suggest that oxidative outcomes in individuals with MetS may

be more responsive to citrus consumption, though this hypothesis will need to be tested in

an adequately powered trial.

Inflammatory and oxidative processes are strongly connected and highly

reciprocal, and their role in atheroma development is irrefutable [2, 6, 127]. The

strongest mechanistic link that influences all of these processes is NFκB activity [138].

Inhibition of NFκB reduces inflammatory and oxidative cascades, which translates into

improved cardiometabolic health [138]. Given the basic science evidence that supports a

role of naringenin in NFκB inhibition, we can postulate that the results observed in the

trials described here may be due to NFκB downregulation by grapefruit. However,

peripheral blood was not collected in these trials, thus preventing our ability to test NFκB

expression or activity, as NFκB is not secreted into circulation. Additionally, the effect

of grapefruit consumption on oxidative stress, inflammation, and adhesion molecule

Page 94: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

93

concentrations as described here was modest, suggesting that if NFκB modulation was in

fact the mechanism underlying these changes, regular grapefruit intake is unlikely to

significantly inhibit this transcription factor’s expression/activity.

Limitations, Future Directions, and Conclusions

The research described here is not without limitations, many of which could be

addressed in future work. In some instances, the sample size may not have been adequate

to observe statistically significant changes in the outcomes evaluated, though the

magnitude of these changes were quite striking in many instancesn ju . The primary aim

of this work, and the outcome for which this study was powered, was evaluation of the

effects of grapefruit on weight change. The application of these findings is also limited

to individuals who are not using pharmacotherapies that are metabolized by the CYP3A4

enzyme, as grapefruit is known to inhibit its activity, thus increasing the bioavailability of

drugs that are metabolized by CYP3A4. Many cardiovascular medications, including

statins and calcium channel blockers, are substrates of CYP3A4 [78]. Taking these

interactions into consideration along with the findings from the present study, grapefruit

could be recommended in the primary prevention setting in those who have not

developed overt CVD.

The overweight and obese population for this study was selected as the link

between elevated BMI and risk of CVD is well established [5, 117]. Overweight/obese

individuals are typically at higher risk for CVD due to the presence of factors that

influence endothelial function including dyslipidemia, hypertension, chronic low-grade

Page 95: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

94

inflammation and oxidative stress [2, 6, 117, 127]. The population enrolled in this trial

may have been too healthy to significantly modify many outcomes of CVD risk, as

baseline values of almost all clinical CVD risk factors were within normal ranges.

Results would likely be more robust and significant in a population at higher risk for

CVD, and future trials should enroll individuals with MetS and/or who are insulin

resistant, as these individuals are more likely to present with an elevated atherogenic risk

profile [79, 88] that would be responsive to a dietary citrus intervention.

In this trial, we showed significant improvements in total cholesterol and LDL-

cholesterol. However, the current literature indicates that particle size of lipoproteins

may be more predictive of future CVD events than only evaluating the type of lipoprotein

(LDL versus HDL) [7]. Individuals may present with normal LDL and HDL cholesterol,

but be at higher risk for cardiovascular events [139] due to increased concentrations of

small, dense LDL particles and suppressed concentrations of large, HDL particles that

result due to remodeling of the lipoproteins by cholesteryl ester transfer protein and

hepatic triglyceride lipase [7]. Due to the effects of citrus flavonoids on hepatic lipid

metabolism, an evaluation of the effect of dietary citrus on lipoprotein particle size is

warranted in order to provide a more comprehensive perspective of the role of citrus on

CVD risk.

Fasting insulin values were also not assessed in the six-week feeding trial. In

light of animal model studies that suggest a beneficial role of dietary naringenin

supplementation on beta cell function, and evaluations of flavonoid-rich foods that have

resulted in increased postprandial insulin secretion, further research is needed. A study

Page 96: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

95

evaluating both the chronic effects of citrus consumption on fasting insulin as well as the

acute effects of citrus on postprandial insulin in a population at risk for cardiometabolic

diseases should be implemented in order to elucidate the relationship between citrus

consumption and insulin secretion/sensitivity. Additionally, the effect of grapefruit on

markers of oxidative stress/inflammation was relatively modest. These results may be

due to biomarker selection, as endothelial dysfunction and atheroma development

develops in response to multiple stimuli, and a variety of different biomarkers could have

been used. However, the biomarkers evaluated in this study were chosen because of the

preliminary data regarding the role of atheroma pathogenesis and responsiveness to

dietary flavonoids in preclinical or clinical trials. Lastly, a functional measure of

endothelial reactivity and vascular tone such as flow mediated dilation was not evaluated

in this trial. FMD is a mechanical measure of a vessel’s ability to react to changes in

blood flow, and has been validated as a prognostic indicator of vascular disease [140]. A

comprehensive assessment of the effects of citrus consumption on endothelial health

should include an analysis of vascular tone in conjunction with measures of metabolic

health, oxidative stress, and inflammation.

There is a significant body of evidence that suggests that citrus fruits are an

important addition to a heart healthy diet. Animal models, cell culture, and

epidemiological data continually demonstrate that grapefruit and its primary flavonoids,

naringin and hesperidin, are cardioprotective. Previous clinical trials evaluating

grapefruit consumption have all evaluated limited numbers of outcomes related to

cardiometabolic health, reducing the ability to compare study results and draw

Page 97: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

96

conclusions. The research described here expands on previous work by testing the effects

of regular grapefruit consumption in a semi-chronic setting as well as in the postprandial

period on a wide range of risk factors that are strongly associated with cardiometabolic

diseases, and specifically, endothelial (dys)function and atheroma development. Based

on the results of this six-week feeding trial and the postprandial evaluation, grapefruit

could be recommended as part of a heart healthy diet, though this dietary change alone is

unlikely to result in significant CVD risk reduction.

Page 98: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

97

Page 99: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

98

APPENDIX A

SUBJECT'S CONSENT FORM

Project Title: Efficiency of daily grapefruit exposure in reducing body weight and

inflammatory markers

You are being asked to read this form so that you know about this research study.

Federal regulations require that you know about the study and the risks involved. If

you decide that you want to participate in this study (consent), signing this form will

say that you know about this study. If you decide you do not want to participate,

that is okay. There will be no penalty to you and you will not lose any benefit which

you would normally have.

WHY IS THIS STUDY BEING DONE? You have the choice to participate in this research project. The purpose of this project is

to determine if grapefruit will produce a greater reduction in oxidative stress (common

damage to cells), inflammation (low level swelling of cells in your body) and weight loss

in overweight adults.

WHY ARE YOU BEING ASKED TO BE IN THIS STUDY? The Principal Investigator or a member of her study staff will discuss the requirements

for participation in this study with you. To be eligible to participate, you should be over

18 years old and if you are female you should be premenopausal. You must be in general

good health with no diagnosis of diabetes, liver, renal disease or cardiovascular disease

and not taking any over the counter or physician prescribed drugs to treat inflammation

(swelling). You must not be taking medications that interact with grapefruit such as

Felodipine, Diltiazem, Nisoldipine, Amlodipine, Nitrendipine and Verapamil. You will

be asked to consume the study provided multivitamins.

You must be willing to discontinue all citrus fruits and be randomized to either a daily

grapefruit or a control diet for a period of 9 weeks. If randomized to the daily grapefruit

diet, you must be willing to consume the assigned amount of Ruby Red grapefruit; 1/2 a

grapefruit, 3x daily before meals for a period of 6 weeks. Sweeteners will be provided by

the study (3 pack/day) which you can use with your grapefruit. Adding sweeteners to

your grapefruit is optional. You must be willing to adhere to a low bioactive fruit and

vegetable diet (such as iceberg lettuce, white potatoes, cucumbers, corn, bananas etc.)

and stop all dietary self-prescribed supplements except for those provided to you by the

study investigator for the entire duration of the study. Additionally, you must discontinue

the consumption of all other citrus fruits for the duration of the study. No other changes

to your diet will be required. You will be asked to fill out a hunger/satiety scale on days

1, 21, and 39 of the study. If you are randomized to the grapefruit group you will be

Page 100: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

99

asked to fill out the hunger/satiety scale 15min before and 15 min after consuming the

grapefruit and eat your meal 15 min after the grapefruit consumption. If you are

randomized to the control group you will be asked to fill out the hunger satiety scale

15min before each meal during the above specified days.

You will not be eligible to participate if it is determined by study investigators that you

have a body mass index (BMI) of greater than 40 or less than 25 kg/m2. These values

will be calculated by the study coordinator using your height and weight. Additionally,

you will not be eligible if you have a body fat percentage of less than 25%, as measured

by electrical impedance. You will not be eligible to participate if you have a history of

smoking in the past 10 years. You are also not eligible if you participate in planned

exercise for more than 10 hours per week.

You must be available for clinic visits, telephone contact and be able to complete study

questionnaires.

A total of approximately 110 individuals will be enrolled in this study locally.

WHAT ARE THE ALTERNATIVES TO BEING IN THIS STUDY? This is not a medical treatment study. The alternative is not to participate; you may want

to talk to your personal doctor to discuss your participation.

WHAT WILL YOU BE ASKED TO DO IN THIS STUDY? Your participation in this study will last up to 10 weeks and includes 4 visits. Each visit is

described below.

Visit 1 (Consent Visit, Week 1)

This visit will last about 60 to 75 minutes. During this visit you will be asked to

complete questionnaires to describe information such as your age, gender,

education, and medical history. The questionnaires should take approximately 30

to 60 minutes to complete. You will also have you height, weight, blood pressure

and body measurements taken at this clinic visit.

You will be advised to complete a ‘run-in’ diet during this time. The purpose of a

run-in is to determine if you can routinely consume a given fruit every day, three

times a day, for several weeks. You may pick any fruit from the designated study

fruit/vegetable list, such as a banana, and are required to eat 1/2 serving of that

fruit three times daily for the 3 week period and to record your intake in the study

provided food log.

You will also be asked to limit your fruit and vegetable intake for the 3 week run-

in/wash out period, selecting only those fruits and vegetables on the study

fruit/vegetable list. The “run-in/wash out” phase is a period of time in the study

to clean out the body of bioactive compounds (nutrients) so a baseline value can

be set to measure against.

You will also be asked to complete three diet recalls during the run-in/wash out

Page 101: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

100

period. A certified, trained study personnel from the University Of Arizona

Department Of Nutritional Sciences will contact you by telephone on 3 random

days during this 3 week “run-in/wash out” period to collect a 24 hour recall of

food intake. You will be excluded from study participation if you fail to meet

requirements of the “run-in/wash out” period.

You will be asked to eat half of a grapefruit to evaluate palatability (taste) and

tolerance.

Visit 2 (Week 4)

This visit will last about 30 to 45 minutes. At the end of the three week “run

in/wash out” period you will be asked to come back to the study clinic where

inflammatory (swelling) markers will be assessed by the collection of

approximately 2 tablespoons of blood. In addition you will be asked to collect

your first morning urine during the final 2 days of the “run-in/wash out” period

and the morning of your clinic visit. Urine will need to be stored in the containers

provided by the study investigator and kept on ice in the study provided cooler

throughout the collection (3 days). You will bring the urine samples (3) to the

study clinic on the final morning (day 3) of urine collection. This urine sample

will be used to measure oxidative stress (cell damage) in the body. Your blood

pressure will be measured at this visit. You will then be randomized to one of the

following groups: the grapefruit intervention group (½ grapefruit eaten three

times daily before meals and discontinue all other citrus fruits) or the control

group which requires you to discontinue all citrus fruit intake for 6 weeks.

If you are randomized to the grapefruit group, you will be asked to eat ½ of a

grapefruit 3x daily before meal, continue the low bioactive fruit and vegetable

consumption, and discontinue all other citrus fruit intake for 6 weeks. At the end

of this visit a one week supply of grapefruits will be provided. A schedule will be

provided to you for weekly grapefruit pick up scheduled at your convenience.

During this clinic visit, you will be given written instructions on how to prepare

grapefruits for consumption. During this period a diary will be provided to you to

record your grapefruit intake and record your satiety. If you are randomized to

the control group, you will be asked to continue with the low bioactive fruit and

vegetable consumption and discontinue the consumption of all citrus fruits for 6

weeks.

During the intervention phase (6 weeks) of the study you will be asked to fill out a

satiety scale on days 1, 21, and 39.

During the 6 week intervention phase of the study a certified, trained study

personnel from the University Of Arizona Department Of Nutritional Sciences will

contact you by telephone on 3 random days to collect a 24 hour recall of food

intake.

Visit 3 (Week 6)

This visit will last about 20 minutes. At this visit weight, blood pressure, and body

Page 102: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

101

measurements will be collected.

Visit 4 (End of Week 9)

This visit will last about 30 to 45 minutes. At the end of the six week grapefruit

intervention you will be asked to return to the study clinic where inflammatory

(swelling) markers will be assessed by the collection of approximately 2 tablespoons

of blood. In addition you will be asked to collect your first morning urine during the

final 2 days of the “run in/wash out” period and the next morning of your clinic visit.

Urine will need to be stored in the containers provided by the study investigator and

kept on ice in the study provided cooler throughout the collection (3 days). You will

bring the urine samples (3) to the study clinic on the final morning (day 3) of urine

collection. This urine sample will be used to measure oxidative stress (cell damage) in

the body. You will also have your weight, body measurements and blood pressure

taken at this clinic visit.

Upon completion of the study, participants will be offered a one-time weight loss

counseling session with a registered dietitian (valued at $100).

PHARMACOKINETIC TRIAL

In addition to the grapefruit feeding trial, we will also be performing a two day ancillary

investigation to evaluate the effects of grapefruit on attenuating the oxidative stress and

inflammation that is known to follow the consumption of a high fat, high carbohydrate

meal. A sub sample of twenty participants from the feeding trial will take part in these

additional study activities. This multi-meal feeding component will be scheduled 2-8

wks post completion with at least 7 days between feeding days.

You will be asked to report to the CATS research facility at the Arizona Cancer Center

(3838 N. Campbell Avenue) on two separate occasions. You will arrive at 7 a.m. and

will be required to be in the fasting state for the previous 12 hours. Upon arrival, a nurse

will fit a port onto the ante-cubital region of your arm. This will remain in place for the

entire day (about 10 hours). A fasting blood draw will be collected directly following port

placement. Participants will have eat a high fat, high carbohydrate breakfast style meal

consisting of biscuits, gravy, sausage, and scrambled eggs (approximately 800 kcals).

You will be asked to consume all food provided within a 20 minute time period. Ten

participants will be randomly selected to eat one ruby red grapefruit along with this meal.

Four hours after consuming the breakfast, a second high fat, high carbohydrate meal will

be provided to all participants. This meal will consist of a cheeseburger and french fries

(approximately 800 kcals). You will be permitted to drink water throughout the day.

Blood will be drawn from the port and urine will be collected on multiple occasions

following the consumption of the meals (see timeline below for schedule of day’s

events).

7 am: Report to clinic, have port fixed to arm, blood collection #1, urine collection #1

7:30 am: Eat breakfast

Page 103: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

102

8:00 am: Blood collection #2

9:00 am: Blood collection #3

10:00 am: Blood collection #4, Urine Collection #2

11:00 am: Blood collection #5

12:00 pm: Eat lunch

12:30 pm: Blood collection #6, Urine Collection #3

2:30 pm: Blood collection #7

4:30 pm: Blood collection #8, Urine Collection #4

You will remain at the CATS facility for a total of ten hours on each of the feeding days.

The CATS facility will have space and equipment available for you to work on

computers, read, and watch movies. Upon completion of the first feeding day, you will

be asked to return the following week for another day that replicates the first feeding day.

Those who were not selected to eat grapefruit on the first day will eat grapefruit with

their breakfast meal on the second day. Those who ate grapefruit the first day will not eat

grapefruit with their breakfast meal on the second feeding day.

There are risks to participating in the pharmacokinetic trial. The main risk involves the

port, which will be fixed to the arm for use in blood collections. Port placement could

lead to pain, swelling, bruising, or infection at the blood draw site, similar to a standard

blood draw. A trained, experienced Registered Nurse will place this port in order to

minimize these risks.

Those who choose to take part in this pharmacokinetic trial will be compensated. You

will receive a $75 Target gift card upon completion of each feeding day, for a total

compensation of $150.

Consent to participate in the pharmacokinetic feeding is not a necessary component for

participation in the grapefruit feeding trial. Do you agree to participate in the

pharmacokinetic feeding? Please indicate your response below and sign:

_____ YES ______ NO Participant Signature: ________________________

ARE THERE ANY RISKS TO ME?

The collection of blood may cause pain, swelling, bruising, or infection at the site of the

needle puncture. Subjects may experience stomach upset related to daily intake of

grapefruit.

ARE THERE ANY BENEFITS TO ME? Your participation may help establish a better understanding of bioactive food

compounds in the body in relation to weight loss and inflammation (swelling) in

overweight adults. You will also receive information about your body measurements.

WILL I BE PAID TO BE IN THIS STUDY?

Page 104: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

103

You will receive $25 gift card at the completion of the wash out diet, at the 3 week clinic

visit, and at the end the grapefruit intervention for a total compensation of $75.

WILL I HAVE TO PAY ANYTHING TO BE IN THIS STUDY? Costs for your participation in this study include your cost of transportation to and from

the study clinic and your personal time to complete the study questionnaires, clinic

measurements and blood draws. You will be provided all grapefruits for the duration of

the study.

WILL I WILL HAVE TO PAY ANYTHING IF I GET HURT IN THIS STUDY? Medical treatment is available if you have an injury. You can get more information about

these treatments by calling Cynthia Thomson at (520) 626-1565.

You may need to pay for immediate and necessary care for side effects if you are injured because of being in this research project. The University of Arizona will not provide for the costs of further treatment beyond immediate necessary care or provide monetary compensation for such injury.

Bad things can happen in any research study even with the use of high standards of care.

These events may not be your fault or the fault of the researcher involved. Known side

effects have been described in this consent form. However, unforeseeable harm may

occur and require care. You do not give up any of your legal rights by signing this form.

If you believe you are injured because of the research or are billed for medical care for

injuries that you feel has been caused by the research, you should contact the Principal

Investigator Cynthia Thomson, PhD, RD at (520) 626-1565.

WILL INFORMATION FROM THIS STUDY BE KEPT CONFIDENTIAL? Information about you will be stored in locked file cabinet; computer files protected with

a password. This consent form will be filed in an official area.

Information about you will be kept confidential to the extent permitted or required by

law. People who have access to your information include the Principal Investigator and

research study personnel. Representatives of regulatory agencies such as OHRP or the

FDA and entities such as the University of Arizona Human Subjects Protection Program

may access your records to make sure the study is being run correctly and that

information is collected properly. The agency that funds this study (Vegetable and Fruit

Improvement Center – Texas A&M University) and the institution(s) where study

procedures are being performed (Nutritional Research clinic at 1601 N. Tucson Blvd

suite 23) may also see your information. However, any information that is sent to them

will be coded with a number so that they cannot tell who you are. Representatives from

these entities can see information that has your name on it if they come to the study site

to view records. If there are any reports about this study, your name will not be in them.

Page 105: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

104

Both urine and blood samples will be stored for up to 10 years for use in ancillary

research studies, but samples will only be identified by the study participant

identification number, not by name and/or birth date or identifying information.

The type of research to be performed using stored urine and blood will focus on

additional measures of oxidative stress and/or inflammation. In the event that new

and/or better methods for measuring oxidative stress and/or inflammation and/or

additional research funds become available the research team would like to be

able to re-test the stored samples.

Agreement to have your samples stored for future analysis is not necessary to participate

in this study. Do you agree to have your urine and blood samples stored for these

ancillary studies should additional funding for these measures be secured? Please initial

your response below:

_______YES ______NO Participant

Initials:_________________

WHO CAN BE CONTACTED ABOUT THIS STUDY? You can call the Principal Investigator to tell her about a concern or complaint about this

research study. The Principal Investigator Cynthia Thomson, PhD, RD can be called at

(520) 626-1565. If you have questions about your rights as a research subject you may

call the University of Arizona Human Subjects Protection Program office at (520) 626-

6721. If you have questions, complaints, or concerns about the research and cannot reach

the Principal Investigator; or want to talk to someone other than the Investigator, you can

contact the Human Subjects Protection Program via the web (this can be anonymous),

please visit http://orcr.vpr.arizona.edu/irb.

STATEMENT OF CONSENT

The procedures, risks, and benefits of this study have been told to me and I agree to

be in this study and sign this form. My questions have been answered. I may ask

more questions whenever I want. I can stop participating in this study at any time

and there will be no bad feelings. My medical care will not change if I quit. The

researcher can remove me from the study at any time and tell me why I have to

stop. New information about this research study will be given to me as it is available.

I do not give up any legal rights by signing this form. A copy of this signed consent

form will be given to me.

___________________________________

____________________________________

Subject's Signature Date

INVESTIGATOR'S AFFIDAVIT:

Either I have or my agent has carefully explained to the subject the nature of the above

project. I hereby certify that to the best of my knowledge the person who signed this

Page 106: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

105

consent form was informed of the nature, demands, benefits, and risks involved in his/her

participation.

___________________________________

________________________

____________

Signature of Presenter Date

Page 107: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

106

APPENDIX B

SUPPLEMENTAL MATERIALS

Grapefruit Weights

Average grapefruit weights, separated by shipment.

Shipment Date Weight±SD (g)

12/14/09 410.0±18.0

1/25/10 399.0±34.7

2/22/10 387.4±37.3

3/8/10 438.6±37.4

3/22/10 380.4±22.0

11/5/10 344.4±13.8

3/22/2010* 298.4±13.8

All grapefruit weighed with peel. *Indicates fruit without peel.

Weights are reported as the average of ten randomly selected

grapefruits.

Page 108: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

107

APPENDIX B continued

Compliance Logs

PPT

#times GF

eaten

Rate of

Compliance PPT

#times GF

eaten

Rate of

Compliance

1 N/A N/A 20 126 1.0

2 126 1.0 21 126 1.0

3 N/A N/A 22 73 0.58

4 83 0.66 23 121 0.96

5 126 1.0 24 126 1.0

6 126 1.0 25 116 0.92

7 126 1.0 26 116 0.92

8 N/A N/A 27 103 0.82

9 122 0.97 28 N/A N/A

10 125 0.99 29 126 1.0

11 120 0.95 30 99 0.79

12 125 0.99 31 124 0.98

13 120 0.95 32 122 0.97

14 124 0.98 33 116 0.92

15 126 1.0 34 126 1.0

16 N/A N/A 35 85 0.67

17 126 1.0 36 104 0.82

18 122 0.97 37 126 1.0

19 121 0.96

PPT=participant.

Thirty-seven of the thirty-nine participants who completed the grapefruit

treatment returned their compliance logs. Five were not filled out properly, denoted by

“N/A” in the table.

Participants had the opportunity to eat grapefruit a total of 126 times. Thus,

compliance was calculated as (# times GF eaten/126)*100. The average rate of

compliance was calculated as 93±11%.

Page 109: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

108

APPENDIX C

ADDITIONAL PUBLICATION #1:

A REVIEW OF EVIDENCE BASED STRATEGIES TO TREAT OBESITY IN

ADULTS

Authors: Deepika Laddu, Caitlin Dow, Melanie Hingle, Cynthia Thomson, Scott Going

as published in Nutrition in Clinical Practice. 2011; 26: 512-525.

Abstract

Obesity, with its comorbidities, is a major public health problem. Population-based

surveys estimate 2 of every 3 U.S. adults are overweight or obese. Despite billions of

dollars spent annually on weight loss attempts, recidivism is high and long-term results are

disappointing. In simplest terms, weight loss and maintenance depends on energy balance,

and a combination of increased energy expenditure by exercise and decreased energy

intake through caloric restriction is the mainstay of behavioral interventions. Many

individuals successfully lose 5-10% of body weight through behavioral approaches, and

thereby significantly improve health. Similar success occurs with some weight loss

prescriptions although evidence for successful weight loss with over the counter

medications and supplements is weak. Commercial weight loss programs have helped

many individuals achieve their goals although few programs have been carefully evaluated

and compared, limiting recommendations of one program over another. For the very

obese, bariatric surgery is an option which leads to significant weight loss and improved

health although risks must be carefully weighed. Lifestyle changes, including regular

Page 110: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

109

physical activity, healthy food choices and portion control, must be adopted, regardless of

the weight loss approach, which requires ongoing support. Patients can best decide the

appropriate approach working with a multi-disciplinary team including their health care

provider and experts in nutrition, exercise and behavioral intervention.

Introduction

Obesity is the result of a chronic surplus in energy intake relative to expenditure.

While the specific causes remain unclear, obesity is thought to arise from a dynamic

interaction between an organism’s biology, behavior, and the increasingly obesogenic

environment. In the United States, the prevalence of obesity is estimated to be 34% in

adults1 and an alarming 17% in children and adolescents.

2

Obesity is associated with a wide variety of comorbidities, including diabetes,

hyperlipidemia, fatty liver disease, obstructive sleep apnea, GERD, vertebral disk disease,

osteoarthritis, and increased risk of post-menopausal breast, endometrial, colon, kidney,

and esophageal cancers.3 Obesity also has significant economic consequences. Recent

figures suggest the increased need for medical care and lost productivity due to higher

rates of disease, disability, and death now exceeds $270 billion annually in the U.S.4

Even small reductions in body weight of 5 to 7% usual weight have been

demonstrated to significantly decrease obesity-related co-morbidities.5 However, once

established, obesity is highly refractory to treatment. Successful weight loss requires

significant lifestyle modifications. Sustaining weight control for the longer term will likely

Page 111: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

110

require individual behavioral change as well as social support and policy shifts to help

maintain lost weight.

The purpose of this review is to provide clinicians and health care professionals

with an update of the current evidence supporting effective interventions for weight

control and the treatment of obesity. As described, a variety of treatment options exist,

including dietary programs, medical nutrition therapy, physical activity, behavior therapy,

pharmacotherapy, bariatric surgery, or a combination of approaches.6 There are, of

course, some individuals who are overweight or obese and who do not suffer from obesity

comorbidities and for whom weight loss attempts may be unnecessary. These individuals

may be better served if they are counseled regarding size acceptance.

Dietary Programs for Weight Loss

Caloric restriction combined with exercise or alone in order to create a daily

energy deficit is the foundation of most weight loss programs. In the absence of precise

measurements of energy expenditure (i.e., direct calorimetry or doubly labeled water), the

resting energy expenditure (REE) of an overweight or obese individual may be estimated

using standard prediction equations based on age, gender, height, and weight7,8

and a

calculation of ideal body weight (IBW) (versus actual body weight). The estimated REE

can then be multiplied by an appropriate physical activity level (PAL) factor to yield an

estimate of total energy expenditure (TEE), which may be further adjusted by the clinician

to help the patient achieve a target energy deficit that promotes weight loss. Low calorie

diets (LCD) (goal of 500-1000 kcal deficit/day) are often used to promote weight loss in

Page 112: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

111

obese individuals and overweight (300-500 kcal deficit/day) individuals.9 This type of low

calorie diet followed for 3-12 months can elicit up to an 8% loss of body weight.9 Very

low calorie diets (VLCD) (800 kcals/day or less) induce rapid weight loss in obese

individuals, although these diets are not recommended for long term use and should only

be attempted under the supervision of a physician.9-11

Notably, individuals on VLCD’s

have experienced the same degree of weight loss as those on LCD’s over the long term

due to decreased adherence to the diet.9,11

The impact of dietary macronutrient composition on weight loss has been

extensively investigated.12

Low carbohydrate (≥40-60 g CHO/d) macronutrient patterns

remain a popular option since participants on these diets can consume highly palatable

foods while adhering to a hypocaloric regimen. A 2008 systematic review that compared

the efficacy of low carbohydrate diets to low fat diets demonstrated an overall greater

weight loss with a low carbohydrate diet at 6 months, although this difference was not

evident at 12 months.12

Low CHO diets are associated with slightly higher protein intake,

and the additional protein intake has been hypothesized to have satiating effects, perhaps

leading to an overall decrease in energy intake.13,14

The Mediterranean Diet (MED),

consisting of high intakes of fruits and vegetables, olive oil, fish, and whole grain, has also

been proposed as an effective dietary intervention for weight loss, although only if it is

also hypocaloric.15

A recent two-year intervention study showed that the low carbohydrate

energy-restricted diet and the energy-restricted Mediterranean diet led to more weight loss

than a similar energy-restricted diet administered with a low fat macronutrient distribution

Page 113: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

112

(Table 1).16

Ultimately, these data suggest that weight loss depends more on restricting

total energy intake than modification of the macronutrient distribution.

Commercial weight loss programs are commonly employed as a weight loss

strategy, though randomized controlled trials evaluating their efficacy have been limited.

A study by Rock et al assessed the efficacy of a two-year commercial weight loss program

in 446 overweight/obese adult women randomized to a center-based program, a web and

telephone-based program, or usual care (counseling with a Registered Dietitian).17

The

results suggested that all three interventions were associated with significant weight loss at

12 and 24 months, although women in the commercial program demonstrated greater

weight loss than usual care. Of importance, this is one of the few studies to show

participants’ maintenance of weight loss at 24 months.(Table 1)17

The retention rate of

this study was high, averaging 94%, likely because of various incentives, no fee for

participation, and frequent contact by study staff. A study of the same commercial weight

loss program completed in 2002 showed that of the 60,164 women who voluntarily

enrolled, only 7% remained in the program at one-year.18,19

Those who completed the

study achieved the greatest weight loss,19

suggesting that participation with good

adherence is the strongest predictor of weight loss (a consistent finding in any type of

weight loss program). Very few studies have compared commercial weight loss

programs, and those reported here demonstrate high attrition rates, ranging from 27% to

93%.18,20

One study performed in the UK compared four popular weight loss programs

(low carbohydrate, low fat, high fiber using a point system, or meal replacements) and

found equal weight loss (approximately 5.9 kg, see Table 1) across treatment approaches

Page 114: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

113

at 6 months.20

Currently, evaluations across commercial weight loss program are lacking

and more studies are needed.

Advances in technology have supported increased access to weight loss programs

at a lower cost than face-to-face programs.21

Results from randomized clinical trials of

online weight loss programs have had mixed results, with very few showing clinically

significant weight loss beyond the Rock study described earlier.21

Tate et al found that

participants who received online behavioral therapy and frequent virtual contact with a

registered dietitian over a 6 month period demonstrated greater weight loss than those who

received education only (-4.1kg versus 1.7 kg).21,22

In contrast, Womble et al reported that

education alone was more effective in promoting weight control than the virtual contact

approach.21,23

Despite the rationale that online weight loss is user-friendly and may

increase study retention rates due to greater flexibility with activities and meetings,

attrition rates are high in online weight loss studies, ranging from 20-40%.21

These

programs should continue to undergo evaluation especially as the population advances in

the frequency and understanding of computer use.

There is also a question of intervention ‘dose.’ In the Shape Up Rhode Island

study, increased feedback with regard to behavioral strategies for weight and self

monitoring during an online weight loss intervention resulted in weight loss of more than

5% body weight compared to those who received no feedback about behavior.24

The

DIAL (Drop It At Last) Study tested different amounts of telephone coaching with obese

adults and found that those who received 20 sessions lost more weight than those who

only received 10 coaching session (4.9 kg versus 3.2 kg, respectively).25

This data

Page 115: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

114

suggests that using distance methods (including online methods) may be effective, but

amount of contact, feedback, and social support should be considered.

Physical Activity

Physical activity is a mainstay of behavioral intervention for weight management.

Evidence continues to grow suggesting that physical activity should be combined with

dietary energy restriction for optimal outcome in the setting of weight loss. First, the dose

of exercise necessary to elicit significant weight loss is relatively large.26,27

Second,

caloric restriction is more efficacious for rapid weight loss than exercise alone.

Importantly, combining exercise with energy restriction helps to counter the common

detrimental effects of caloric restriction alone including loss of lean tissue and lowered

resting metabolic rate. Finally, exercise can improve metabolic fitness, even in the absence

of significant weight loss.26,28,29

A combination of moderate caloric restriction and a

moderate increase in exercise energy expenditure is typically recommended since for

many, modest changes in dietary and exercise behaviors are easier to achieve than a major

change in either behavior alone.

Current physical activity recommendations call for approximately 30 minutes of

moderate to vigorous physical activity 5-7 days per week. The health benefits of this level

of physical activity are well documented and reasonable for most people to attain,

particularly if completed in multiple, short bouts. 30,31

. When weight loss is the primary

goal, a greater volume of exercise is needed, especially in the absence of significant

caloric restriction. Based on analyses of studies of energy expenditure using doubly

labeled water, Schoeller et al suggest a minimum of 60 minutes of daily MVPA is

Page 116: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

115

necessary for weight loss.32

Similarly, Jakicic and others33-35

have shown that a weekly

exercise energy expenditure upward of 2,000 kcals/week is necessary for significant

weight loss through exercise alone.36-39

Even at this level of expenditure, average weight

loss is modest, which can be discouraging and likely limits adherence, although the health

benefits are significant. It is important to note, however, that percent of weight lost as fat,

as well as reduction in abdominal and visceral fat is greater with exercise than diet.26,28,29

and regular exercise is the primary predictor of maintenance of weight after weight loss,38-

40

Dynamic, moderate-intensity, aerobic activity (e.g., walking, cycling, swimming,

etc.) is most commonly recommended for weight loss and maintenance since aerobic

activity elicits significant energy expenditure and improves cardiovascular and metabolic

fitness.40

Strength training activities also have significant benefits, promoting lean mass

and maintaining resting metabolic rate, and improving muscle strength and endurance,

thereby supporting further increases in physical activity.41,42

For persons with limitations

in mobility, resistance exercise may be a feasible alternative to other forms of exercise to

promote initial weight loss. Weight loss and fat loss may be less with resistance training

compared to aerobic exercise unless energy expenditure is equated.27

There is some

evidence that resistance training combined with higher protein intake promotes weight and

fat loss while maintaining muscle mass, which is important in obese individuals who are

losing large amounts of weight and in older individuals with sarcopenic obesity, in whom

further muscle loss presents a significant health risk.40,41,43,44

Page 117: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

116

Clinicians can play a vital role in promoting physical activity along with dietary

change for weight management, which includes understanding of the published guidelines

for physical activity goals for prevention of weight gain (~150 minutes of MVPA per

week), active weight loss (150-420 minutes of MVPA per week), and weight maintenance

after loss (200-400 minutes of MVPA per week).40

Registered dietitians and other health

care providers should regularly promote the health benefits of regular physical activity,

although the final exercise program should be developed in collaboration with the patient

or client’s physician and a certified exercise specialist.

Behavioral Therapy

Behavioral Management

Real or perceived obstacles to weight loss may limit short term adherence and

long-term maintenance of weight loss. Behavioral therapy is a relatively inexpensive

strategy for weight control and is less invasive and more acceptable than surgical and/or

pharmacological approaches.21

Overall, behavioral therapy has been shown to be effective

in decreasing body weight (on average 7-10% over 6 months of intensive therapy), but

maintenance of weight loss has proven to be more challenging.10,24,45,46

One of the most common issues in any weight loss or weight maintenance program

is adherence.47

Length of time participating in an intervention is inversely associated with

adherence. 47,48

As participation time increases, adherence decreases, and weight loss is

not as pronounced as in the initial phase of loss.10,47,48

Self-monitoring of dietary intake

and physical activity is considered a central behavioral strategy to promote adherence and

Page 118: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

117

indirectly promote weight loss.47

Participants who attend more group counseling sessions

and complete self-monitoring assignments are more likely to lose weight compared to

those who do not self-monitor.45,48

While self-monitoring is time consuming, frequency

of self-monitoring appears to predict adherence to weight loss regimes.47

Additionally,

perceptions of the complexity of weight loss program rules have been inversely associated

with adherence and amount of weight loss achieved.49

Standard behavioral therapy that focuses on changing lifestyle behaviors has been

used as a weight loss strategy for decades,46,47

yet its efficacy in producing long-term

weight loss has not been clearly demonstrated. Data suggest that rewards, behavioral cues,

and reminders all key components of standard behavioral therapy, become less effective in

eliciting lifestyle behavior changes over time.45

Further, the psychosocial literature shows

that boredom related to the intervention, loss of motivation, and emotional eating are

common barriers to weight control.11,45,50

The efficacy of cognitive behavioral therapy in conjunction with standard

behavioral therapy was recently evaluated for weight control. 51

Cognitive behavioral

therapy is based on the concept that thoughts control feelings and behaviors, thus

managing thoughts regarding self-worth, body image and others can direct weight loss

behaviors and prevent post-intervention weight regain.51

A summary of the results of

cognitive behavioral therapy interventions is given in Table 2 and generally indicate

significant weight loss during the intervention, with concomitant weight regain during

follow-up.

Page 119: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

118

Maintenance Tailored Therapy is a newer behavioral approach that is focused not

only on the promotion of weight loss but also weight maintenance after weight loss. This

approach administers various behavioral strategies at different times, for different

durations and even at variable doses to keep participants actively engaged long term in the

overall behavior-based intervention. Jeffery et al 45

tested maintenance tailored therapy

using a 6-phase program, each lasting eight weeks, with a four-week break between each

phase. Each phase had a specific concentration: i. calorie counting and goal setting, ii.

walking 10,000 steps per day, iii. meal replacement, iv. 3000 kcal exercise/week, v. the

stop light diet, and vi. deposit contracts.45

Although statistically equivalent weight loss

was seen in intervention and control groups, the maintenance tailored therapy group

demonstrated weight loss during all phases as well as break periods; most important, no

relapse in weight loss had occurred after 18 months in the intervention (Table 2).45

While

preliminary results are encouraging, more research is needed regarding this type of

behavioral intervention.

Motivational interviewing is a common weight loss technique used by clinicians.

One study assessed physicians’ counseling strategies using the 5A’s model of weight loss

-i. Assess risk, behavior, and readiness to change, ii. Advise change of behaviors, iii.

Agree and set goals, iv. Assist in addressing barriers and securing support, v. Arrange for

follow-up, and showed that physicians use an average of 5 of the possible 18 counseling

techniques. 52

Patients whose physicians used more counseling techniques were more

motivated to lose weight.52

A separate study showed that patients whose physicians

employed motivational interviewing techniques lost 1.6 kg more weight after three months

Page 120: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

119

than those patients whose physicians did not use motivational interviewing.53

In an 8-

week weight loss study where participants were randomized to guided self help (GSH) or

GSH with motivaltional interviewing (MI), the GSH/MI group lost slightly more weight

and had less attrition than those receiving GSH alone.54

Based on present data and the

benefits of motivational interviewing, clinicians should be trained using this technique to

enhance weight loss in obese patients.

Pharmacotherapy

Dietary supplements that claim to treat obesity are widely distributed and readily

obtained in health food stores, grocery stores, pharmacies, on the Internet, and by mail.

According to the Natural Medicines Comprehensive Database, there are over 50

individual dietary supplements and 125 combination products marketed for weight loss.55

Population based surveys suggest that approximately that 15.2% of American adults

(20.6% women and 9.7% men) use dietary supplements for weight loss,56,57

most

commonly supplements with a stimulant as the active ingredient57

are used. Easy access,

low cost, no need for a prescription, and no need for behavioral modifications are the

primary reasons why individuals seek out dietary supplements as weight loss aids. Lack of

scientific evidence regarding efficacy and safety precludes informed recommendations for

use of weight-loss supplements, even if used only as adjunct to other weight management

strategies.

Common characteristics of individuals who used weight reduction supplements

included repeated, unsuccessful attempts at weight loss, and the use of multiple weight

Page 121: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

120

loss methods (>4) to achieve a target weight. 58

59

Higher usage of dietary supplements

was also observed in women (44.9%) then men (19.8%), and in obese minorities, aged 25-

34 years. Individuals of low socioeconomic status or who were less educated (high school

degree or less), were also more likely to use dietary supplements.59

Appetite suppressants are the most common supplement used for weight reduction.

These supplements are purported to promote weight loss by several mechanisms,

including increasing energy expenditure, increasing satiety, increasing fat oxidation,

inhibiting dietary fat absorption, modulating carbohydrate metabolism, increasing fat

excretion, increasing water elimination, and enhancing mood.56

Since multiple metabolic

pathways are targeted, these weight loss supplements are known to increase the risk of

adverse events. Meta-analysis of studies of Ephedra, for example, which contained

caffeine and resulted in only slightly higher weight loss compared to placebo, showed a

two to three-fold increased risk of developing severe neurological, psychological,

gastrointestinal and cardiovascular complications as well as death, and therefore was

deemed unsafe and removed from the market in 2004.56

The evidence in support of dietary supplements for weight reduction is not

convincing and there is a need for more research regarding long-term efficacy and

safety.60

The efficacy of the majority of supplements for altering weight and body fat

remains inconclusive. Side effects may be brand specific or vary within the same brand,

and there remains limited information regarding the dietary and drug interactions that

supplements may induce. The benefits and risks of dietary supplements for weight loss

have not been demonstrated in well-designed trials.

Page 122: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

121

Anti-Obesity Drugs

According to the National Institutes of Health, pharmacotherapy agents are recommended

for individuals who have been unable to achieve weight loss goals with diet and physical

activity alone, have a body mass index (BMI) greater than 27kg/m2, and exhibit serious

obesity comorbidities such as diabetes or high blood pressure.61,62 Currently, two

avenues involving merging of pharmacology and nutrition are being explored: 1)

inhibition of energy intake through targeting orexigenic signaling into the hypothalamic

feeding center and 2) increasing of energy expenditure through anorexic signals to

promote a caloric deficit (Table 3).63 Despite these efforts in drug development, it

remains unclear which targeted pathways will translate into safe, significant, and long

term weight loss.63

Although weight reduction with pharmaceutical treatment is typically modest (5-10%

reduction of total body weight) and similar to outcomes with behavioral interventions,

individuals on pharmacotherapy report an easier time maintaining weight post therapy

treatment.62 Additional benefits of pharmacotherapy include decreased blood pressure,

lipid levels, and blood glucose levels, respectively, along with an increase in insulin

sensitivity and reduced risk of cardiovascular disease and related co-morbidities.64

The majority of FDA-approved weight loss drugs are for short-term use (a few weeks or

months) (Table 4).65 Currently, there are two primary types of drugs for obesity

treatment: fat absorption inhibitors and anorexins.62 Both types are more effective when

Page 123: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

122

combined with additional weight-loss practices such as calorie-restriction, exercise, and

behavioral modification.

Fat-absorption inhibitors work by reducing dietary fat absorption. The most

recently FDA approved fat absorption inhibitor is Xenical (Hoffman-LaRoche Inc, South

San Francisco, CA), which works by blocking approximately 30% of dietary fat.62

To

date, Xenical is the only weight loss drug that has been approved for long-term use in

severely obese individuals in the United States although its safety and efficacy has only

been established up to 2 years (Table 4). The over-the-counter version, Alli ®, is also

available, but at a lower dose than Xenical®.

Anorexins (appetite suppressants) work by promoting satiation (reduced desire to

eat) by slowing down the gastric emptying process. Mechanistically, anorexins interfere

with neurological pathways operating through the hypothalamus by inhibiting orexigenic

pathways while simultaneously enhancing anorexigenic pathways (Table 3). Anorexins

also decrease appetite by increasing serotonin or catecholamine, the two primary

neurotransmitters responsible for controlling mood and appetite.66

The net result is

enhanced satiation (reduced desire to eat), satiety (enhanced level of fullness on eating), or

both. These drugs generally come in the form of tablets or extended-release capsules, and

can be obtained via prescription or over the counter.66

The risks associated with weight-loss medications include development of

tolerance or addiction, thus a change in medication therapy may be necessary for

continued weight loss and in the case of addiction, various restrictions may apply;

continuous monitoring is recommended (Table 4). The majority of these drugs are

Page 124: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

123

designed for short-term use to limit the risks. Limited success has been achieved at

reducing body weight in obese populations by modulating fat absorption or altering

signaling input to the brain's feeding center. Further, the probability of achieving weight

loss greater than 10% remains low. Typically, individual weight loss plateaus after 6

months.66

It is unclear whether this plateau is due to a tolerance or if the effectiveness of

the medication has been maximized. Although most side effects are mild in nature, it is

uncertain whether these effects will progress or subside as the body adjusts to the

medications.

Bariatric Surgery

For obese individuals unable to achieve normal weight for height through

behavioral or other approaches, bariatric surgery offers an effective method for major

weight loss. The most efficacious surgical procedures reduce body weight by ~35-40%,

and most of the weight loss is maintained for at least 15 years.67-70

Intestinal

malabsorption and gastric restriction are the two most obvious mechanisms to explain

weight loss after bariatric surgery. Malabsorptive procedures, such as the jejunoileal

bypass (JIB) and biliopancreatic diversion (BPD) have been associated with serious

complications which have led to the abandonment of JIB from clinical practice and

restricted use of BPD to the superobese (BMI > 50 kg·m-2

).69

Purely restrictive bariatric operations cause weight loss by limiting the capacity of

the stomach to accommodate food and constricting the flow of injected nutrients.

Gastroplasty, often called “stomach stapling”, involves placing a horizontal staple line to

Page 125: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

124

partition the stomach into a small, proximal pouch and a large distal remnant, connected

via a narrow stoma.71

High failure rate led to the vertical banded gastroplasty (VBG) in

which the partitioning line extends upward from the angle of his and a polypropylene

mesh reinforces the stoma.72

Although VBG effectively limits food consumption and

results in 30-50% reduction of body weight over 1-2 years, long-term results have been

disappointing.69

Recipients often compensate by eating frequent, small meals and calorie

dense foods. A nearly 80% failure rate has been reported after 10 years.73

Adjustable

gastric banding (AGB), a purely restrictive procedure, has been popular worldwide. An

adjustable variant of VBG, this approach involves placement of a prosthetic band around

the upper stomach to partition it into a small proximal pouch and a large, distal remnant,

connected through a narrow constriction.74,75

Free of anastomoses, this approach

eliminates staple-line dehiscence, is readily accomplished laparoscopically, and the

aperture can be modified noninvasively as needed. Risks include band slippage or erosion

into the stomach and reflux esophagitis. Weight loss after gastric banding is usually less

than bypass surgeries, but short and long-term complications are also less frequent.67

The Roux-en-y-gastric bypass (RYGB) appears to offer the best balance of

effectiveness versus risk and is the most widely used surgery in the U.S. for morbidly

obese people. Massive weight loss after this procedure reduces obesity-related

comorbidities and mortality. The operation may also improve glucose homeostasis

through physiological mechanisms not explained by weight loss alone,76

increasing

interest in its application in NIDDM patients.

Page 126: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

125

The modern RYGB divides the stomach into a small, proximal pouch and a

separate large, distal, remnant. The upper pouch is joined to the proximal jejunum through

a narrow gastrojejunal anastomosis (Roux-en-y configuration).76

Thus, the storage

capacity of the stomach is reduced to ~5% of its normal value, and injected food bypasses

approximately 95% of the stomach, the entire duodenum, and a small portion of the

proximal jejunum. Patients typically lose 35-40% of body weight, and long-term (> 15

years) maintenance of weight loss is good.68,70

Peri-operative mortality, largely from

pulmonary embolism or sepsis, is ~1%, but may be more in the hands of less experienced

surgeons. Post-operative complications occur in 10% of cases, and include deep vein

thrombosis, anastomotic leaks, internal hernias, gastrointestinal bleeding, ulcers in the

bypassed segments, torsion or volvulus of the roux limb, closed loop obstruction, stomal

stenosis, wound complications, staple-line disruption and gallstone formation with rapid

weight loss.67,69,70

Bypassing of the stomach and duodenum impairs absorption of iron,

calcium, thiamine and vitamin B12, thus supplementation of these micronutrients and

periodic monitoring for deficiencies are required.

In randomized, prospective trials, RYGB has caused 50-80% loss of excess body

weight as opposed to 30-50% after the equally restrictive VBG,77-80

and weight loss after

RYGB is more durable than after VBG. The two mechanisms cited most often to explain

the greater efficacy of RYGB over VBG are malabsorption and dumping syndrome.

However, clinically significant malabsorption, measured by indices such as albumin,

prealbumin, and fecal fat, is not observed after the standard proximal RYGB81-83

and the

severity of dumping, although it sometimes occurs, correlates poorly with the efficacy of

Page 127: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

126

RYGB.76

Cummings et al (2004) suggest RYGB is more effective than VBG primarily

because loss of appetite from RYGB is greater and more consistent than VBG. The effect

is not explained by early satiety from restriction since it extends beyond the early

postprandial period. Also, levels of leptin and insulin, adiposity hormones, decrease as

expected with weight loss and thus do not account for weight loss84,85

and its regulation at

a lower level. Similarly, alterations in gut hormones do not seem to mediate the effects

since cholecystokin, serotonin and vasoactive intestinal peptide are unaffected by gastric

bypass.86,87

In contrast, impairment in ghrelin, a circulating orexigen, may explain at least

some of the loss of appetite following RYGB. Ghrelin is produced by the stomach and, to

a lesser extent, the duodenum, the areas affected by RYGB. Because ingested nutrients are

dominant regulators of its production, and because most of the ghrelin-producing tissue is

permanently excluded from contact with ingested nutrients after RYGB, disruption of

ghrelin regulation may explain the appetite suppression following RYGB. Indeed, several

prospective trials have reported decreased or very low ghrelin,88-92

even after massive

weight loss, although one group reported an increase as expected with other modes of

weight loss.93

Ghrelin also exerts several diabetogenic effects, and suppression of ghrelin

after RYGB may enhance glucose disposal and contribute to the rapid reversal of Type 2

diabetes mellitus that follows gastric bypass.76

Genetics in Weight Loss Response

Regardless of the approach (e.g., diet, exercise, surgery, and pharmacological), and even

with good compliance, weight loss and maintenance results vary significantly among

Page 128: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

127

patients. This large variability amongst individuals on similar programs suggests genetics

play an important role in modifying the response to weight loss and weight maintenance

programs.94

Indeed, studies done with monozygotic twins show that weight loss on the

same intervention is quite similar within twin pairs, with significant differences between

pairs, demonstrating the strong contribution of genes to weight loss.95

A comprehensive discussion of the role of genetics in weight loss and maintenance

is beyond the scope of this review. For more information the reader is referred to Hainer 95

and Hetherington. 96

It is important for clinicians to understand that despite good

adherence, results for some individuals may be less than what was targeted. The Human

Obesity Gene Map, released in 2005, reports over 600 loci of single nucleotide

polymorphisms that may be associated with obesity.97

Most cases of obesity are thought

to be due to an interaction of multiple candidate genes, known as polygenic obesity,95

and

very few (perhaps 4-5% of severely obese individuals) are due to a single gene, referred to

as monogenic obesity. The complexity is evident in appetite dysregulation, for example,

which may be a consequence of multiple genetic polymorphisms in obese individuals, and

is a compelling example of polygenic obesity. Variations in the genes for cholecystokinin,

leptin and the leptin receptor, as well as those involved in lipid metabolism, have all been

implicated in increased meal size, poor postprandial compensation response, and

excessive snacking behaviors.95

Taken together, these factors reveal the complex

interaction between genetics and environment in obesity. While dramatic advances in

technology have allowed researchers to study the role of genes in weight loss, we are far

from providing individualized weight loss programs based on genetics.94

Page 129: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

128

Summary and Conclusion

Obesity remains a significant and challenging clinical condition. Effective

strategies for long term weight loss elude us. The following recommendations can be

proposed based on current knowledge:

Prevention is the most effective approach to reducing the substantial co-morbidity

associated obesity.

Clinicians have a primary responsibility to address weight control with patients

who are overweight/obese or demonstrating an upward trend in body weight.

Weight, waist circumference and when possible, body composition, should be

evaluated regularly to detect undesirable trends so that intervention can be initiated

early.

Targeted, behavior-based interventions that employ standard behavioral therapy,

motivational interviewing, or cognitive behavioral therapy can be effective in

promoting adherence and weight loss, although ongoing support is desirable to

counter recidivism.

Physical activity should be combined with energy restriction to promote caloric

deficit and weight loss and maintain lean mass and resting energy expenditure.

There are no effective dietary supplements for weight control; ephedra and

products containing ephredra-related compounds should be considered dangerous

and not be prescribed for weight control.

Page 130: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

129

Evidence suggests FDA-approved pharmacological agents, while limited in terms

of available products, can promote weight loss of 5 to 10%.

Commercial weight loss programs have been sparsely evaluated and rarely

compared in controlled studies; nevertheless, current evidence suggests these

programs can be effective for weight loss.

For very obese persons who have failed with other approaches, a surgical

procedure such as the RYGB or AGB can promote significant weight loss and

improve obesity comorbidities, although the risks of these procedures must be

carefully considered.

For many obese persons, even modest weight loss of 5-10% has significant health

benefits, although even this level of loss is difficult to maintain. Ongoing support for

behavior change is essential and can best be delivered by a multi-disciplinary team

including the patient’s physician and experts in nutrition, exercise and behavioral

therapy.

Page 131: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

130

Table 1. Efficacy of various diets and programs on weight loss and weight maintenance.

Diet/Program Tested Against Population Duration of

Intervention

Duration of

Follow Up

Findings Reference

Low Carbohydrate Diet

(Energy not restricted. 20g

CHO/day during induction

phase, increasing to 120g/day)

Med Diet / LF

n=322; ob

Israeli M&F;

medical

clinic

employees

6 months 18 months

>6 kg weight loss at 6 months;

4.7 kg total weight loss at 24

months

16

Med Diet (1500 kcals for

women; 1800 kcals for men.

35% kcals from fat, focusing

on olive oil and nuts)

LC / LF

n=322; ob

Israeli M&F;

medical

clinic

employees

6 months 18 months

~4.5 kg weight loss at 6

months; 4.4 kg total weight loss

at 24 months, virtually no

rebound

16

Low Fat Diet (1500 kcals for

females; 11800 kcals for males.

30% energy ffrom fat, 10%

from saturated fat, 300mg

cholesterol)

LC / Med Diet

n=322; ob

Israeli M&F;

medical

clinic

employees

6 months 18 months

~4.5 kg weight loss at 6

months; 2.9 kg total weight loss

at 24 months

16

Commercial Weight Loss

Program - Center Based

Commercial

Weight Loss

Program -

Phone Based /

Usual Care

n=442;

ovwt/ob F 2 years N/A

Energy restriction of 500-1000

kcal/day; prepackaged foods

comprised 42%-68% of energy

needs; weekly meetings with

consultant; goal of increased

physical activity.

7.4 kg, 6.2 kg, 2.0 kg weight

loss, respectively, after 2 years;

94% retention; meals provided

free of charge

17

Page 132: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

131

BBC (British Broadcasting

Corporation) Diet Trials

Comparison of

LC / Point

System / Meal

Replacement /

LF

n=293;

obwt/ob

British M&F

6 months 6 months

6 months: Weight loss of 6.0,

6.6, 4.8, 6.3 kg, respectively; 12

months: total weight loss (54%

of original sample) of 8.5, 8.03,

6.49, 8.76 kg, respectively

20

Internet Behavior Therapy

(Received weekly lessons

regarding PA and nutrition;

submitted self monitoring

diaries online; received

feedback from therapist

regarding diaries)

Internet

Education

n=91;

ovwt/ob

M&F;

hospital

employees

6 months N/A

Both groups were given goals

to restrict energy intake to

1200-1500 kcals/day and to

increase energy expenditure to

at least 1000 kcals/week.

4.1 kg weight loss compared to

1.6 kg weight loss

22

Commercial Internet Weight

Loss

LEARN

Weight Loss

Manual

n=47;

ovwt/ob F 4 months 8 months

4 months: 0.9% compared to

3.6% weight loss. 12 months:

1.1% compared to 4.0% weight

loss.

23

Page 133: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

132

FU= Follow Up; SBT=Standard Behavior Therapy; GSH=Guided Self Help; MI=Motivational Interviewing;

M=Male; F=Female

Table 2. Efficacy of various theories and programs on weight loss and weight maintenance.

Theory/Pr

ogram

Tested

Against

Populatio

n

Duration of

Intervention FU Findings Reference

Cognitive

Theory

SBT &

GSH

n=150; ob

F 10 months 3 years

10 months: 8.17 kg, 11.6,

5.24 kg weight loss,

respectfully. 3 years post-

intervention: Total weight

loss of 0.84, 3.21, .04 kg,

respectfully

51

Cognitive

Theory

Exercise

Dietetic

Treatment

n=204;

ovwt/ob

M&F

10 weeks 1 year

Decrease 1.36 and 1.44

points on BMI scale after

10 weeks. Less rebound

compared to EDT after 1

year.

98

Cognitive

Theory +

Mediterra

nean Diet

N/A

n=1406

ovwt/ob

Spanish

M&F

8.5 months N/A

Non-randomized; 7.8 kg

weight loss; very low

attrition rate

50

Mainte

nance

Tailored

Therapy

SBT n=213;

ob M&F

18

months

12

months

18 months: 8.2 kg and

9.3 kg weight loss

respectively. Higher

adherence than SBT; no

rebound over 18 months

versus significant rebound

in SBT. Follow up: 31%

less weight regain than

SBT

45

Guided

Self Help

+

Motivatio

nal

Interview

ing

Guided

Self Help

n=39;

ovwt

M&F

11 weeks N/A

Decrease 1.48 on BMI

scale, compared to -.7

points in GSH group;

higher retention rate in

GSH/MI

54

Page 134: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

133

Table 3. Signaling pathways targeted by current

anti-obesity medications and their action

Inhibit EI Enhance EE

Orexigenic Signals Anorexic Signals

Neuropeptide Y (NPY) Pro-opiomelanocortin

(POMC)

Agouti-related protein

(ARGP)

Melanocortin system (a-

MSH)

Melanin-containing hormone:

(MCH)

Cocaine and

anphetamine-regulated

transcript (CART)

Endocanabinoids Glucagon like Peptide-1

(GLP-1)

Hypocretins/Orexins Amine neurotransmittrs

Ghrelin Peptide YY

EI = energy intake, EE = energy expenditure

Page 135: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

134

Table 4. Weight Loss Medications

Conventional

Medicines Trade Name Mechanism of action Efficacy Side Effects

Approved

Use Reference

Orlistat Alli (OTC )

Blocks the digestion

and absorption of fat in

stomach and intestines.

Reduces 25% of

dietary fat; weight-

loss less for OTC vs.

prescription

Loose or more frequent

stools; intestinal cramps,

gas, oily spotting; less

GI events than

prescription

Long-term 99,62

**Orlistat Xenical

(prescription)

Gastric and pancreatic

lipase inhibitor; reduces

absortion of 30%

dietary fat consumed

Reduces ≈30%

dietary fat

absorption

Intestinal cramps, gas,

oily spotting Long-term

100

Benzphetamin

e Didrex

CNS stimulant;

mechanism uncertain;

decreases appetite,

increases energy

expenditure

Small increase in

weight loss of drug-

treated patients

only a fraction of a

pound a week

Palpitation, tachycardia,

increased blood pressure,

restlessness, dizziness,

insomnia, nausea,

diarrhea, other

gastrointestinal

disturbances

Short-term 101,62

Diethylpropion Tenuate

CNS stimulant;

mechanism uncertain;

decreases appetite,

increases energy

expenditure

Weight loss is 0.5

kg per week;

tolerance to

medications in this

class usually

develops a few

weeks

Increased blood

pressure; tachycardia,

insomnia, dizziness;

increased risk for

valvulopathy

Short-term 62

Page 136: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

135

Phendimetrazi

ne

Bontril;

Prelu-2

Stimulates satiety in

hypothalamus and

limbic regions

Efficacy with other

anorectic agents has

not been studied;

combined use has

the potential for

serious cardiac

problems.

Arteriosclerosis,

symptomatic

cardiovascular disease,

moderate and severe

hypertension,

hyperthyroidism, and

glaucoma.

Short-term 102

Phentermine Adipex;

Ionamine

CNS stimulant;

mechanism uncertain;

decreases appetite,

increases energy

expenditure

Not established

Dizziness, dry mouth,

difficulty sleeping,

irritability, nausea,

vomiting, diarrhea, or

constipation may occur.

Short-term 62

Phenylpropran

olamine Dexatrim

May act through the

paraventricular

hypothalamus to

suppress appetite; may

be mediated by the

alpha-1 adrenergic

satiety receptors.

associated with an

increased risk of

hemorrhagic stroke

in women; FDA

does not recommend

using this product

closing of your throat;

swelling of lips, tongue,

or face; or hives;

short-term;

removed

from market

due to

increased risk

of

hemorrhagic

stoke

103

Rimonabant Accomplia

blocks endocannabinoid

receptors in brain,

adipose tissue, digestive

system, liver and

muscles; anorexic effect

on CNS, also acts on

gut to increase

sensation of satiety.

Average 4-5kg

weight reduction;

decreased waist

circumference;

increased in HDL-C,

decreased in LDL-C

particles, decreased

insulin resistance

naseau, diarrhea,

vomiting, anxiety,

depression, dizziness

Long term 100

Page 137: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

136

***Sibutramin

e

Meridia;

Reducti

monoamine-reuptake

inhibitor, to reduce food

intake

Average weight

losses of 4–5 kg;

improvements in

HDL-C,

triglycerides and

HBA1c; supported

by 2 year efficacy

and safety trial

Increases in blood

pressure and heart rate

Long-term;

removed

from market

in 2010 due

to increased

risk of serious

heart events,

including

non-fatal

heart attack

and stroke.

100,104

*DEA schedule= abuse potential

**Orlistat is also available in a reduced-strength form without a prescription (Alli)

***Sibutramine recently was removed from the market in 2010.

Page 138: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

137

References (for Appendix C)

1. Flegal KM, Carroll MD, Ogden CL, Curtin LR. Prevalence and trends in obesity among

US adults, 1999-2008. JAMA. 2010;303:235-41.

2. Ogden CL, Carroll MD, Curtin LR, Lamb MM, Flegal KM. Prevalence of high body

mass index in US children and adolescents, 2007-2008. JAMA. 2010;303:242-9.

3. Guh DP, Zhang W, Bansback N, Amarsi Z, Birmingham CL, Anis AH. The incidence of

co-morbidities related to obesity and overweight: a systematic review and meta-analysis.

BMC Public Health. 2009;9:88.

4. http://www.businessweek.com/lifestyle/content/healthday/648708.html. Accessed

January 15, 2011.

5. Cummings S, Parham ES, Strain GW. Position of the American Dietetic Association:

Weight management. J Am Diet Assoc. 2009;109:330-46.

6. NHLBI. Obesity Education Initiative Expert Panel on the Identification, Evaluation, and

Treatment of Overweight and Obesity in adults. Clinical guidelines on the identification,

evaluation, and treatment of overweight and obesity in adults: The Evidence Report. NIH

Publication No. 98-4083, Sept 1998. Washington, DC: NIH - National Heart, Lung, and

Blood Institute.

7. Harris JA, Benedict FG. A biometric study of basal metabolism in man (publication no.

279). Washington, DC. Carnegie Institute of Washington. 1919.

8. Mifflin MD, St Jeor ST, Hill LA, Scott BJ, Daugherty SA, Koh YO. A new predictive

equation for resting energy expenditure in healthy individuals. Am J Clin Nutr.

1990;51:241-7.

9. Cannon CP, Kumar A. Treatment of overweight and obesity: lifestyle, pharmacologic,

and surgical options. Clin Cornerstone. 2009;9:55-68; discussion 9-71.

10. Turk MW, Yang K, Hravnak M, Sereika SM, Ewing LJ, Burke LE. Randomized clinical

trials of weight loss maintenance: a review. J Cardiovasc Nurs. 2009;24:58-80.

11. Hainer V, Toplak H, Mitrakou A. Treatment modalities of obesity: what fits whom?

Diabetes Care. 2008;31 Suppl 2:S269-77.

12. Hession M, Rolland C, Kulkarni U, Wise A, Broom J. Systematic review of randomized

controlled trials of low-carbohydrate vs. low-fat/low-calorie diets in the management of

obesity and its comorbidities. Obes Rev. 2009;10:36-50.

13. Acheson KJ. Carbohydrate for weight and metabolic control: where do we stand?

Nutrition. 2010;26:141-5.

14. Brehm BJ, D'Alessio DA. Weight loss and metabolic benefits with diets of varying fat

and carbohydrate content: separating the wheat from the chaff. Nat Clin Pract Endocrinol

Metab. 2008;4:140-6.

15. Jimenez-Cruz A, Jimenez AB, Pichardo-Osuna A, Chaudry T, Bacardi-Gascon M. Long

term effect of Mediterranean diet on weight loss. Nutr Hosp. 2009;24:753-4.

16. Shai I, Schwarzfuchs D, Henkin Y, et al. Weight loss with a low-carbohydrate,

Mediterranean, or low-fat diet. N Engl J Med. 2008;359:229-41.

17. Rock CL, Flatt SW, Sherwood NE, Karanja N, Pakiz B, Thomson CA. Effect of a free

prepared meal and incentivized weight loss program on weight loss and weight loss

Page 139: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

138

maintenance in obese and overweight women: a randomized controlled trial. JAMA.

2010;304:1803-10.

18. Martin CK, Talamini L, Johnson A, Hymel AM, Khavjou O. Weight loss and retention in

a commercial weight-loss program and the effect of corporate partnership. Int J Obes

(Lond). 2010;34:742-50.

19. Wing RR. Treatment options for obesity: do commercial weight loss programs have a

role? JAMA. 2010;304:1837-8.

20. Truby H, Baic S, deLooy A, et al. Randomised controlled trial of four commercial weight

loss programmes in the UK: initial findings from the BBC "diet trials". BMJ.

2006;332:1309-14.

21. Arem H, Irwin M. A review of web-based weight loss interventions in adults. Obes Rev.

2010.

22. Tate DF, Wing RR, Winett RA. Using internet technology to deliver a behavioral weight

loss program. JAMA. 2001;295:1172-7.

23. Womble LG, Wadden TA, McGuckin BG, Sargent SL, Rothman RA, Krauthamer-Ewing

ES. A randomized controlled trial of a commercial internet weight loss program. Obes

Res. 2004;12:1011-8.

24. Wing RR, Crane MM, Thomas JG, Kumar R, Weinberg B. Improving weight loss

outcomes of community interventions by incorporating behavioral strategies. Am J Public

Health. 2010;100:2513-9.

25. Sherwood NE, Jeffery RW, Welsh EM, Vanwormer J, Hotop AM. The drop it at last

study: six-month results of a phone-based weight loss trial. Am J Health Promot.

2010;24:378-83.

26. Ross R, Dagnone D, Jones PJ, et al. Reduction in obesity and related comorbid conditions

after diet-induced weight loss or exercise-induced weight loss in men. A randomized,

controlled trial. Ann Intern Med. 2000;133:92-103.

27. Williams D, Teixeira PJ, Going SB. Exercise. In: Human Body Composition, Second

Edition. In: Heymsfield S. W, Z., Lohman T., and Going, S. (eds.), ed. Champaign, IL:

Human Kinetics; 2004:313-30.

28. Ross R, Pedwell H, Rissanen J. Effects of energy restriction and exercise on skeletal

muscle and adipose tissue in women as measured by magnetic resonance imaging. Am J

Clin Nutr. 1995;61:1179-85.

29. Janssen I, Fortier A, Hudson R, Ross R. Effects of an energy-restrictive diet with or

without exercise on abdominal fat, intermuscular fat, and metabolic risk factors in obese

women. Diabetes Care. 2002;25:431-8.

30. Haskell WL, Lee IM, Pate RR, et al. Physical activity and public health: updated

recommendation for adults from the American College of Sports Medicine and the

American Heart Association. Med Sci Sports Exerc. 2007;39:1423-34.

31. Physical Activity Guidelines for Americans. U.S. Dept. of Health and Human Services.

http://health.gov/paguidelines/guidelines/chapter1.aspx. 2008. (Accessed at

32. Schoeller DA, Shay K, Kushner RF. How much physical activity is needed to minimize

weight gain in previously obese women? Am J Clin Nutr. 1997;66:551.

Page 140: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

139

33. Jakicic JM, Marcus BH, Gallagher KI, Napolitano M, Lang W. Effect of exercise

duration and intensity on weight loss in overweight, sedentary women: a randomized

trial. JAMA. 2003;290:1323-30.

34. Jakicic JM, Marcus BH, Lang W, Janney C. Effect of exercise on 24-month weight loss

maintenance in overweight women. Arch Intern Med. 2008;168:1550-9; discussion 9-60.

35. Jakicic JM, Clark K, Coleman E, et al. American College of Sports Medicine position

stand. Appropriate intervention strategies for weight loss and prevention of weight regain

for adults. Med Sci Sports Exerc. 2001;33:2145-56.

36. Jeffery RW, Wing RR, Sherwood NE, Tate DF. Physical activity and weight loss: does

prescribing higher physical activity goals improve outcome? Am J Clin Nutr.

2003;78:684-9.

37. Saris W, Blair SN, Van Baak MA, et al. How much physical activity is enough to prevent

unhealthy weight gain? Outcome of the IASO 1st Stock Conference and consensus

statement. Obes Reviews. 2003;4:101-14.

38. Wing RR, Phelan S. Long-term weight loss maintenance. Am J Clin Nutr. 2005;82:222S-

5S.

39. Tate DF, Jeffery RW, Sherwood NE, Wing RR. Long-term weight losses associated with

prescription of higher physical activity goals. Are higher levels of physical activity

protective against weight regain? Am J Clin Nutr. 2007;85:954-9.

40. Donnelly JE, Blair SN, Jakicic JM, Manore MM, Rankin JW, Smith BK. American

College of Sports Medicine Position Stand. Appropriate physical activity intervention

strategies for weight loss and prevention of weight regain for adults. Med Sci Sports

Exerc. 2009;41:459-71.

41. Schmitz KH, Hannan PJ, Stovitz SD, Bryan CJ, Warren M, Jensen MD. Strength training

and adiposity in premenopausal women: strong, healthy, and empowered study. Am J

Clin Nutr. 2007;86:566-72.

42. Schmitz KH, Jensen MD, Kugler KC, Jeffery RW, Leon AS. Strength training for obesity

prevention in midlife women. Int J Obes Relat Metab Disord. 2003;27:326-33.

43. Layman DK, Evans E, Baum JI, Seyler J, Erickson DJ, Boileau RA. Dietary protein and

exercise have additive effects on body composition during weight loss in adult women. J

Nutr. 2005;135:1903-10.

44. Doi T, Matsuo T, Sugawara M, et al. New approach for weight reduction by a

combination of diet, light resistance exercise and the timing of ingesting a protein

supplement. Asia Pac J Clin Nutr. 2001;10:226-32.

45. Jeffery RW, Levy RL, Langer SL, et al. A comparison of maintenance-tailored therapy

(MTT) and standard behavior therapy (SBT) for the treatment of obesity. Prev Med.

2009;49:384-9.

46. Sarwer DB, von Sydow Green A, Vetter ML, Wadden TA. Behavior therapy for obesity:

where are we now? Curr Opin Endocrinol Diabetes Obes. 2009;16:347-52.

47. Acharya SD, Elci OU, Sereika SM, et al. Adherence to a behavioral weight loss treatment

program enhances weight loss and improvements in biomarkers. Patient Prefer

Adherence. 2009;3:151-60.

Page 141: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

140

48. Greenberg I, Stampfer MJ, Schwarzfuchs D, Shai I. Adherence and success in long-term

weight loss diets: the dietary intervention randomized controlled trial (DIRECT). J Am

Coll Nutr. 2009;28:159-68.

49. Mata J, Todd PM, Lippke S. When weight management lasts. Lower perceived rule

complexity increases adherence. Appetite. 2010;54:37-43.

50. Corbalan MD, Morales EM, Canteras M, Espallardo A, Hernandez T, Garaulet M.

Effectiveness of cognitive-behavioral therapy based on the Mediterranean diet for the

treatment of obesity. Nutrition. 2009;25:861-9.

51. Cooper Z, Doll HA, Hawker DM, et al. Testing a new cognitive behavioural treatment for

obesity: A randomized controlled trial with three-year follow-up. Behav Res Ther.

2011;48:706-13.

52. Jay M, Gillespie C, Schlair S, Sherman S, Kalet A. Physicians' use of the 5As in

counseling obese patients: is the quality of counseling associated with patients'

motivation and intention to lose weight? BMC Health Serv Res. 2010;10:159.

53. Pollak KI, Alexander SC, Coffman CJ, et al. Physician communication techniques and

weight loss in adults: Project CHAT. Am J Prev Med. 2010;39:321-8.

54. DiMarco ID, Klein DA, Clark VL, Wilson GT. The use of motivational interviewing

techniques to enhance the efficacy of guided self-help behavioral weight loss treatment.

Eat Behav. 2009;10:134-6.

55. Saper RB, Eisenberg DM, Phillips RS. Common dietary supplements for weight loss. Am

Fam Physician. 2004;70:1731-8.

56. Alraei RG. Herbal and dietary supplements for weight loss. Topics in Clinical Nutrition.

2010;25:136-50.

57. Blanck HM, Serdula MK, Gillespie C, et al. Use of nonprescription dietary supplements

for weight loss is common among Americans. J Am Diet Assoc. 2007;107:441-7.

58. Pillitteri JL, Shiffman S, Rohay JM, Harkins AM, Burton SL, Wadden TA. Use of dietary

supplements for weight loss in the United States: results of a national survey. Obesity

(Silver Spring, Md. 2008;16:790-6.

59. Herber D. "Herbal preparations for obesity: are they useful?". Primary Care

2003;30:441-63.

60. Lenz TL, Hamilton WR. Supplemental products used for weight loss. J Am Pharm Assoc

(2003). 2004;44:59-67; quiz -8.

61. "Clinical Guidelines On the Identification, Evaluation, and Treatment of Overweight and

Obesity in Adults." The Evidence Report. National Institutes of Health, Sept. 1998. Web.

<http://www.nhlbi.nih.gov/guidelines/obesity/ob_gdlns.pdf>.

62. National Institute of Diabetes and Digestive and Kidney Diseases (U.S.), and Weight-

control Information Network (U.S.). 2010. Prescription medications for the treatment of

obesity. [Bethesda, MD]: National Institute of Diabetes and Digestive and Kidney

Diseases, National Institutes of Health.

http://win.niddk.nih.gov/publications/prescription.htm#meds.

63. Bays HE. Current and investigational antiobesity agents and obesity therapeutic treatment

targets. Obes Res. 2004;12:1197-211.

64. Neff LM, Aronne LJ. Pharmacotherapy for obesity. Curr Atheroscler Rep. 2007;9:454-

62.

Page 142: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

141

65. Hofbauer KG, Nicholson JR, Boss O. The obesity epidemic: current and future

pharmacological treatments. Annu Rev Pharmacol Toxicol. 2007;47:565-92.

66. Sonnenberg GE, Matfin G, Reinhardt RR. Drug treatments for obesity: where are we

heading and how do we get there? Current Obesity Treatment Options. British Journal of

Diabetes and Vascular Disease. 2007;7:111-8.

67. Brolin RE. Bariatric surgery and long-term control of morbid obesity. JAMA.

2002;288:2793-6.

68. Jones KB, Jr. Experience with the Roux-en-Y gastric bypass, and commentary on current

trends. Obes Surg. 2000;10:183-5.

69. Mun EC, Blackburn GL, Matthews JB. Current status of medical and surgical therapy for

obesity. Gastroenterology. 2001;120:669-81.

70. Pories WJ, Swanson MS, MacDonald KG, et al. Who would have thought it? An

operation proves to be the most effective therapy for adult-onset diabetes mellitus. Ann

Surg. 1995;222:339-50; discussion 50-2.

71. Pace WG, Martin EW, Jr., Tetirick T, Fabri PJ, Carey LC. Gastric partitioning for morbid

obesity. Ann Surg. 1979;190:392-400.

72. Mason EE. Vertical banded gastroplasty for obesity. Arch Surg. 1982;117:701-6.

73. Balsiger BM, Poggio JL, Mai J, Kelly KA, Sarr MG. Ten and more years after vertical

banded gastroplasty as primary operation for morbid obesity. J Gastrointest Surg.

2000;4:598-605.

74. Bo O, Modalsli O. Gastric banding, a surgical method of treating morbid obesity:

preliminary report. Int J Obes. 1983;7:493-9.

75. Kuzmak L. Stoma adjustable silicone gastric banding. Prob Gen Surg. 1992;9:298-317.

76. Cummings DE, Overduin J, Foster-Schubert KE. Gastric bypass for obesity: mechanisms

of weight loss and diabetes resolution. J Clin Endocrinol Metab. 2004;89:2608-15.

77. Naslund I, Wickbom G, Christoffersson E, Agren G. A prospective randomized

comparison of gastric bypass and gastroplasty. Complications and early results. Acta Chir

Scand. 1986;152:681-9.

78. Sugerman HJ, Starkey JV, Birkenhauer R. A randomized prospective trial of gastric

bypass versus vertical banded gastroplasty for morbid obesity and their effects on sweets

versus non-sweets eaters. Ann Surg. 1987;205:613-24.

79. Howard L, Malone M, Michalek A, Carter J, Alger S, Van Woert J. Gastric Bypass and

Vertical Banded Gastroplasty- a Prospective Randomized Comparison and 5-Year

Follow-up. Obes Surg. 1995;5:55-60.

80. Nightengale ML, Sarr MG, Kelly KA, Jensen MD, Zinsmeister AR, Palumbo PJ.

Prospective evaluation of vertical banded gastroplasty as the primary operation for

morbid obesity. Mayo Clin Proc. 1991;66:773-82.

81. Faraj M, Jones P, Sniderman AD, Cianflone K. Enhanced dietary fat clearance in

postobese women. J Lipid Res. 2001;42:571-80.

82. MacLean LD, Rhode BM, Nohr CW. Long- or short-limb gastric bypass? J Gastrointest

Surg. 2001;5:525-30.

83. Naslund I. Gastric bypass versus gastroplasty. A prospective study of differences in two

surgical procedures for morbid obesity. Acta Chir Scand Suppl. 1987;536:1-60.

Page 143: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

142

84. Hickey MS, Pories WJ, MacDonald KG, Jr., et al. A new paradigm for type 2 diabetes

mellitus: could it be a disease of the foregut? Ann Surg. 1998;227:637-43; discussion 43-

4.

85. Stoeckli R, Chanda R, Langer I, Keller U. Changes of body weight and plasma ghrelin

levels after gastric banding and gastric bypass. Obes Res. 2004;12:346-50.

86. Kellum JM, Kuemmerle JF, O'Dorisio TM, et al. Gastrointestinal hormone responses to

meals before and after gastric bypass and vertical banded gastroplasty. Ann Surg.

1990;211:763-70; discussion 70-1.

87. Pappas TN. Physiological satiety implications of gastrointestinal antiobesity surgery. Am

J Clin Nutr. 1992;55:571S-2S.

88. Cummings DE, Weigle DS, Frayo RS, et al. Plasma ghrelin levels after diet-induced

weight loss or gastric bypass surgery. N Engl J Med. 2002;346:1623-30.

89. Geloneze B, Tambascia MA, Pilla VF, Geloneze SR, Repetto EM, Pareja JC. Ghrelin: a

gut-brain hormone: effect of gastric bypass surgery. Obes Surg. 2003;13:17-22.

90. Tritos NA, Mun E, Bertkau A, Grayson R, Maratos-Flier E, Goldfine A. Serum ghrelin

levels in response to glucose load in obese subjects post-gastric bypass surgery. Obes

Res. 2003;11:919-24.

91. Leonetti F, Silecchia G, Iacobellis G, et al. Different plasma ghrelin levels after

laparoscopic gastric bypass and adjustable gastric banding in morbid obese subjects. J

Clin Endocrinol Metab. 2003;88:4227-31.

92. Couce M, Cottam D, Esplen J, Teijeiro R, Schauer PR, Burguera B. Central vs.

peripheral ghrelin: impact on human obesity. NAASO annual meeting. Obes Res.

2003;11(Suppl):A35.

93. Holdstock C, Engstrom BE, Ohrvall M, Lind L, Sundbom M, Karlsson FA. Ghrelin and

adipose tissue regulatory peptides: effect of gastric bypass surgery in obese humans. J

Clin Endocrinol Metab. 2003;88:3177-83.

94. Deram S, Villares SM. Genetic variants influencing effectiveness of weight loss

strategies. Arq Bras Endocrinol Metabol. 2009;53:129-38.

95. Hainer V, Zamrazilova H, Spalova J, et al. Role of hereditary factors in weight loss and

its maintenance. Physiol Res. 2008;57 Suppl 1:S1-15.

96. Hetherington MM, Cecil JE. Gene-environment interactions in obesity. Forum Nutr.

2010;63:195-203.

97. Rankinen T, Zuberi A, Chagnon YC, et al. The human obesity gene map: the 2005

update. Obesity (Silver Spring, Md. 2006;14:529-644.

98. Werrij MQ, Jansen A, Mulkens S, Elgersma HJ, Ament AJ, Hospers HJ. Adding

cognitive therapy to dietetic treatment is associated with less relapse in obesity. J

Psychosom Res. 2009;67:315-24.

99. Orlistat (Alli). GlaxoSmithKline; Middlesex, UK; March 2011.

100. Padwal RS, Majumdar SR. Drug treatments for obesity: orlistat, sibutramine, and

rimonabant. Lancet. 2007;369:71-7.

101. Manati PR. Pfizer USPI, 00674, August 2010.

www.pfizer.com/files/products/uspi_didrex.pdf.

102. Valeant Pharmaceutical International. Phendimetrazine tartrate [package insert]. Valeant

Pharmaceuticals North America; One Enterprise; Aliso Viejo, CA 92656 USA.

Page 144: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

143

103. Alger S, Larson K, Boyce VL, et al. Effect of phenylpropanolamine on energy

expenditure and weight loss in overweight women. Am J Clin Nutr. 1993;57:120-6.

104. James WP, Astrup A, Finer N, et al. Effect of sibutramine on weight maintenance after

weight loss: a randomised trial. STORM Study Group. Sibutramine Trial of Obesity

Reduction and Maintenance. Lancet. 2000;356:2119-25.

Page 145: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

144

APPENDIX D

ADDITIONAL PUBLICATION #2:

ASSOCIATIONS BETWEEN MOTHER’S BMI, FRUIT AND VEGETABLE INTAKE

AND AVAILABILITY, AND CHILD’S BODY SHAPE AS REPORTED BY WOMEN

RESPONDING TO AN ANNUAL SURVEY

Authors: Caitlin A. Dow, Betsy C. Wertheim, Elizabeth Pivonka, Cynthia A. Thomson

as published in Food and Nutrition Sciences. 2012, 3, 1636-1643.

Abstract

Previous evidence indicates that a child’s body mass index (BMI) and eating behaviors

are often related to the BMI and eating behaviors of his/her parents. Additionally, there

is evidence suggesting that fruit and vegetable intake may impart weight control benefits.

The purpose of this study was to examine the relationship between mother’s BMI and the

intake/availability of fruits and vegetables in the home, as well as mother’s perceived

body shape of her child. This is a cross sectional, descriptive analysis of results from a

large internet-based survey of Generation X and Y mothers evaluating the role of fruit

and vegetable consumption and health behaviors in U.S. families. Mothers (n=1,469)

with children under the age of 18 living in the home reported her BMI, her fruit and

vegetable intake, and fruit and vegetable availability in the home. Additionally, mothers

with children between the ages of 2 and 12 (n=1,177) reported her child’s body shape

(using graduated images of children ranging from the 3rd

-97th

percentiles of BMI).

Mother’s BMI was not related to fruit or vegetable intake, though it was inversely related

Page 146: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

145

to fruit, but not vegetable, availability in the home. Mother’s BMI was also

positively related to child’s body shape, and mother’s fruit, but not vegetable, intake was

inversely related to child’s body shape. Our findings support a potential role for fruit

availability promoting healthy BMI in mothers and/or healthier body shape in their

children.

Introduction

Obesity is a health concern in adults and children [1-3], ultimately resulting in an

increased risk for chronic diseases including Type 2 Diabetes, cardiovascular disease, and

cancer [4, 5]. Evidence suggests that children’s body mass index (BMI) and eating

behaviors are associated with the BMI and eating behaviors of their parents [2, 6, 7].

Increased fruit and vegetable (FV) consumption as part of a healthy, balanced diet is an

established health goal in the U.S. [8] and is often recommended by practitioners as a

targeted behavioral strategy to reduce body weight [9-11].

The social ecological theory suggests that the environment plays an important role in

behavior development, and the family environment is especially important in the

development of eating behaviors in children [12]. Evidence correlating FV intake in

children and the role of parental FV consumption and attitudes has been evaluated in both

systematic reviews and nationally representative surveys [12, 13], though a recent

systematic review and meta-analysis found only a weak association [14]. A review

evaluating the role of the home environment on FV intake in children and adolescents

showed that availability, parental modeling, and parental intake were all positively

Page 147: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

146

associated with children’s FV consumption [12]. The role of the mother in children’s

eating behaviors may be of particular importance, as mothers characteristically spend

more time than fathers with children and have more influence over feeding activities [15].

There is a large body of evidence suggesting that parental intake of FV, specifically, is

highly associated with children’s intake [7, 13, 16].

Despite the established role of parental choices and modeling on children’s FV intake,

there is a lack of data regarding the complex relationship between a mother’s FV intake

and her BMI and the relationship of these parameters to her child’s FV intake and body

shape (i.e. ectomorph, mesomorph, or endomorph).

A large internet survey of mothers with children residing in the home conducted annually

on behalf of the Produce for Better Health Foundation affords a unique opportunity to

further explore these relationships. Specifically, this report sought to evaluate the

relationships between FV intake/availability and BMI of mothers, maternal BMI and her

child’s body shape, and maternal FV intake/availability and mother’s perception of her

child’s body shape. We anticipated that FV intake/availability would have an inverse

relationship with BMI, maternal BMI would be associated with larger reported body

shape of the child, and maternal FV intake/availability would be associated with smaller

perceived child’s body shape.

METHODS

Survey Development

Page 148: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

147

An annual online consumer survey was originally developed by the Produce for

Better Health Foundation Research Board in collaboration with Ogilvy Public Relations

Worldwide, the public relations agency for Fruits & Veggies—More Matters®

and

OnResearch, Inc. (Ontario, Canada). A pilot study was conducted in 2006 in Generation

X (GenX; individuals born between 1965-1979) mothers (n=73), further reviewed and

refined, and sent to outside reviewers with behavioral expertise for additional review. The

baseline survey was fielded in February 2007 in 1,000 mothers and has been completed

annually since 2007 in US mothers.

Questions were developed to obtain information about knowledge, attitudes, and beliefs

of GenX mothers and to gain insight into barriers to providing fruits and vegetables to

their families. The sample of potential participants was drawn from membership lists of

survey panels for online companies who partner with OnResearch, Inc. Potential

respondents who met inclusion criteria were invited via email to participate in the survey.

Respondents included mothers born between 1965 and 1990 with at least one child under

the age of 18. Those who completed the survey were given a small cash reward for

participating. Each year, the survey is completed in women who have not participated in

previous years.

Most questions were answered using a 5-point Likert scale, though several questions

required open-ended responses. Respondents were asked to provide information

regarding their personal FV intake and FV availability in their home (including fresh,

canned, frozen, or dried). The survey stated that a serving of fruit/vegetable was

Page 149: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

148

approximately the size of a tennis ball, and women provided a numerical response for

the number of servings they ate each day. Women reported their average daily intake of

fruits and vegetables separately.

Additional Survey Sections Added in 2011 Survey

In 2011, additional survey questions were developed that had not been used in previous

surveys. All data for the current analysis were collected January 14-31, 2011. Mother’s

BMI was calculated using information from those respondents who elected to provide

data on their height and weight (92.9%, n=1,486). Those who reported a BMI <15 kg/m2

(n=9) or >60 kg/m2 (n=8) were excluded from the analytical dataset. Thus, data from

1,469 mothers were included in our analyses.

Additionally in 2011, respondents were asked to report on their child’s body shape only if

they had children between the ages of 2 and 12 years living in the home. If a woman

reported having more than one child between 2 and 12 years of age, they were asked to

report on the body shape of only one child chosen at random by the survey software

(n=1,177). The images used to inform on child’s body shape were produced by Truby and

Paxton in 2002 [17]. The children depicted in these images ranged in size based on the

1979 National Center for Health Statistics (NCHS) BMI percentiles (A=3rd

, B=10th

,

C=25th

, D=50th

, E=75th

, F=90th

, and G=97th

) (Figure 1). In order to assess outcomes

associated with children’s body size, the eight categories were collapsed into clinically

relevant BMI groups: ≤25th

percentile (groups A, B, and C combined), 50th

percentile

(group D), 75th

percentile (group E), and ≥90th

percentile (groups F and G combined).

Page 150: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

149

Statistical Analysis

Associations between mother’s BMI and fruit or vegetable intake were tested using linear

regression. Wilcoxon’s rank sum test was used to test the difference in FV intake

between women who opted to provide height and weight compared to those who did not.

The relationship between FV intake and FV availability in the home was tested using

multinomial logistic regression and assessed for confounding by mother’s BMI, though

no confounding was observed. Linear regression models were also used to evaluate the

differences in BMI between women who selected specific categories of FV availability

among the variety of responses available. Associations were assessed for potential

confounding by education, employment, marital status, age, number of children, daily

physical activity, fruit or vegetable intake, and ethnicity. Covariates that resulted in a beta

coefficient change of more than 10% were included in the final adjusted model. The

covariates included in the model for fruit availability and BMI were physical activity and

education, whereas the model for vegetable availability and BMI included physical

activity, education, marital status, and ethnicity.

Differences in BMI between mothers who selected various body shapes for their children

were tested using ordinal logistic regression. The association between child’s body shape

and FV availability in the home was tested using chi-square tests. Lastly, the relationship

between mother’s FV intake and child’s body shape was tested using ordinal logistic

regression. Trends were tested by including child’s body shape category as an ordinal

variable in the model, wherein categories A/B/C=1, D=2, E=3, and F/G=4. This trend

Page 151: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

150

was assessed for potential confounding by mother’s BMI; no confounding was

observed. All tests were considered statistically significant at p<0.05. Statistical

analyses were performed using Stata 11.1 (StataCorp, College Station, TX).

RESULTS

Survey respondents had a mean ± SD age of 36.4 ± 6.1 years and 2.0 ± 1.0 children under

age 18 living in the home. The mean BMI (calculated from self-reported height and

weight) of mothers was 27.5 ± 7.3 kg/m2, and 54.2% of women were considered

overweight or obese (BMI ≥ 25 kg/m2). Mothers reported a mean number of daily

servings of fruits of 2.6 ± 3.0 and a median of 2 servings. The mean number of daily

servings of vegetables was 2.5 ± 2.0 servings, and the median intake was 2 servings

(Table 1). There was no difference in fruit or vegetable intake between women who

elected to provide height/weight compared to those who did not (P=0.599 and P=0.680,

respectively).

Associations between Mothers’ BMI, FV Intake, and FV Availability

No significant association was found between mother’s BMI and self-reported daily

servings of fruit or vegetable intake (P=0.125 and P=0.099, respectively). However,

women who reported greater availability of FV in their home did report significantly

greater daily intake than those who reported lower availability (Table 2).

BMI of respondents was compared across FV availability categories. Women who

reported that fruit was “always available” had a significantly lower BMI than those who

Page 152: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

151

reported that fruit was “almost always/ usually available” (26.6 ± 6.6 versus 28.1 ±

7.9 kg/m2, respectively; P<0.001) or “occasionally/ rarely/ never available” (26.6 ± 6.6

versus 29.4 ± 7.1 kg/m2; P<0.001) in the home (Table 3). This relationship held when

adjusted for education and physical activity. Furthermore, the BMI of women who

selected that vegetables were “always available” was significantly lower than those who

selected that vegetables were “almost always/usually available” (26.9 ± 6.8 versus 28.1 ±

7.8, respectively; P=0.001); women who selected that vegetables were

“occasionally/rarely/never available” also demonstrated a higher BMI compared to the

“always available group,” but this difference was not significantly different, likely due to

the small number of women reporting low availability (n=74). After adjusting for

education, physical activity, marital status, and ethnicity, the relationship between BMI

and vegetables “almost always/usually available” was attenuated and no longer

statistically significant (P=0.100) (Table 3).

Associations between Child’s Body Shape, FV Availability, and Mother’s FV Intake

Child’s body shape was assessed using the images in Figure 1. Most women (n=915,

77.7%) selected that their child resembled images A, B, or C (relating to ≤25th

BMI

percentile) while very few women (n=70, 5.9%) selected images F/G (relating to ≥90th

BMI percentile). There was a significant association between mother’s BMI and child’s

body shape (P=0.001) (Figure 2). Additionally, those women who selected that her child

resembled endomorphic images (E or F/G) had significantly higher BMIs than those who

selected that their child resembled more ectomorphic or mesomorphic images (A/B/C).

Page 153: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

152

No significant differences existed amongst categories of child’s body shape when

compared to FV availability categories (data not shown). However, mothers who

reported greater fruit intake were more likely to report their child’s body shape in the

lower BMI categories (P=0.007), though this inverse association did not hold true in

relation to mother’s vegetable intake and child’s body shape (P=0.809) (Table 4).

DISCUSSION

This study provides hypothesis-generating results regarding the role of mothers’ practices

specific to FV intake/availability and both her and her child’s body size. There was a

significant inverse relationship between mother’s BMI and fruit and, to a lesser extent,

vegetable availability, though we did not find a significant association between mother’s

BMI and reported FV intake. We also found an association between mother’s fruit, but

not vegetable, intake and the reported body shape of her child; mothers who reported

children in the higher percentiles of BMI reported significantly lower fruit intake than

mothers who reported their child’s body size in the lower or normal BMI percentiles.

Short-term interventions indicate that increasing FV intake may be a good strategy for

weight control as part of a balanced diet, as FV’s increase feelings of satiety and may

reduce intake of high-energy foods [9, 18]. However, observational data evaluating the

relationship between FV intake and BMI provide inconclusive results [9, 18, 19]. A

systematic review of the literature suggests that there may be a relationship between low

FV intake and higher BMI [19]; however, most studies that assess this relationship are

designed with endpoints of FV intake and chronic disease outcomes [20], making it

difficult to accurately extrapolate their findings to obesity, specifically. Our study did

Page 154: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

153

show a statistically significant relationship between fruit availability and BMI in

mothers. These findings are consistent with much of the research regarding home FV

availability and weight status, most of which has focused on children and adolescents and

not adults [21, 22]. Interestingly, this effect was not seen with vegetable availability and

BMI, as mothers who selected that vegetables were “occasionally/rarely/never available”

in their home had lower BMIs than those who selected that vegetables were “almost

always/usually available.” Supporting our data, an analysis from the USDA’s 1994-1996

Continuing Survey of Food Intakes by Individuals (CSFII) found that lower BMI was

associated with greater fruit, but not vegetable, intake in adults and children [23].

Although we did not see a significant difference in BMI between women selecting the

highest and lowest vegetable availability categories, it should be noted that women who

indicated that vegetables were always available in their home had a lower BMI than those

who did not choose this option. In addition, the BMI of this group was similar to the

BMI of the group who indicated that fruits were always available in the home. One

important issue that may potentially explain these results is the very low number of

women who selected the lowest categories of vegetable availability (n=74) compared

with those who indicated that vegetables were always available (n=753). Furthermore,

we did not obtain information regarding fruit and vegetable preparation methods.

Vegetables consumed may have been fried or eaten with high calorie sauces or butter.

This effect may explain the lack of an association between vegetable availability and

BMI.

Page 155: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

154

According to the social ecological theory, there are many factors that influence

children’s weight status, including parental behaviors and beliefs [12, 15]. Our study

explored the relationship between mother’s BMI and child’s (aged 2-12 years) body

shape. Child’s body shape was used as a proxy for child’s BMI, as validated by Saxton

et al. [24]. We found a significant association between mother’s BMI and child’s body

shape. Women who selected that their child resembled an image of a larger body size

had higher BMIs than those who selected a smaller body size. These findings agree with

previous reports regarding parental BMI and child’s weight status [25, 26].

In addition to the relationship between mother’s BMI and child’s body shape, we

explored additional variables that may influence child’s weight status. Previous work

suggests that FV availability in the home is related to children’s FV intake [22].

Children’s FV intake may then confer benefits such as healthier weight status and

reduced adiposity [18, 22]. Despite this rationale, we failed to observe a relationship

between FV availability and child’s body shape. However, actual intake of FV in children

was not assessed.

There is a large body of evidence exploring the relationship between mother-child FV

intake resemblance [13, 14, 27] as well as FV intake and BMI in both adults and children

[18, 19]. To our knowledge, this is the first study examining the relationship between

maternal FV intake and child’s body shape. We observed a significant inverse association

between mother’s fruit intake and child’s body shape, suggesting that lower maternal

fruit intake may be associated with her child’s larger body size. This relationship did not

exist between vegetable intake and child’s shape, perhaps for the same reasons that it did

Page 156: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

155

not exist between mother’s BMI and home vegetable availability. These findings

provide further evidence suggesting that interventions to improve FV intake in children

should also focus on promoting FV intake in mothers, as the family environment is a

strong determinant of behavior development [12].

This is the first internet-based study to target a specific generation of mothers and for

which we have novel information regarding mother’s perception of child’s body size to

evaluate the role of FV in family health. The study was large with responses from over

1,400 mothers. However, the study does have limitations. This study was conducted

using cross-sectional data, limiting the ability to determine causation. Additionally, this

study was conducted online and is therefore biased towards women with internet access.

This study was also based on self-reported height and weight, FV intake, and FV

availability; thus, respondents may have provided inaccurate information. Without a

marker of validation (height and weight measured by a trained professional, a biomarker

of intake such as serum carotenoids, or in-home appraisal of FV availability), we cannot

ascertain the validity of self-reported data in the current study. We also did not have any

information regarding other aspects of the diet, making it difficult to control for other

potential confounders. Furthermore, social desirability may have resulted in answers that

were not truly representative of actual behaviors and beliefs.

Additionally, our survey collected information on mother’s perceptions of her child’s

body shape. Although these images were designed using BMI percentiles developed over

30 years ago, the scale itself was not developed until 2002 [17] and currently represents

the most up-to-date scale of children’s body images. Furthermore, this scale is unique as

Page 157: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

156

each image is an actual photograph of a child that correlates to a specific BMI,

whereas previously developed scales simply depict silhouettes that do not necessarily

correspond to actual BMI values [17]. A validation study by Saxton et al. in 2009

indicated that children were able to identify their body shape using these images with

reasonably high accuracy [24]. However, there is literature that indicates that mothers

tend to underestimate their child’s body size [28, 29]. Currently, 16.9% of American

children are classified as obese [30], while only 5.9% of women in this survey chose the

body images that most closely correlate with obesity (F=90th

percentile; G=97th

percentile). This suggests that some women may have misclassified her child or that the

sample of women surveyed is not largely representative of the American population.

Conclusions

The relationships between mother’s BMI, FV intake, FV availability, and child’s weight

status are extremely complex and not well understood. Using a national sampling of

mothers, we were able to evaluate FV availability in the home in relation to the BMI of

the mother and also the body size of the child. The current study supports a role of fruit

and, to a lesser degree, vegetable availability in BMI of mothers. Our data suggest a

relationship between mother’s fruit, but not vegetable, intake and child’s body shape.

The evidence generated from this study, particularly that regarding women’s BMI and

home FV availability, and mother’s FV intake and child’s body shape, should be further

explored in longitudinal studies in order to inform on future interventions targeting FV

consumption and healthy weight in the family setting.

Page 158: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

157

FIGURES

Figure 1. Child’s body shape images shown to mothers responding to an annual survey

regarding fruits and vegetables. Images produced by Truby and Paxton et al.

Page 159: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

158

10

20

30

40

50

60

Mo

ther's b

ody m

ass ind

ex (

kg/m

2)

A/B/C D E F/G

Child’s Body Shape

n=91

5 BMI=26.8±6.5 n=12

3 BMI=26.5±6.9 n=6

9 BMI=29.1±8.0* n=70

BMI=30.9±9.6*

M

oth

er’

s

Bo

dy

Ma

ss

Ind

ex

(kg

/m2)

BMI values represent mean ± SD. *Significantly different from the BMI of mothers who selected images A/B/C. P trend=0.001

Figure 2. Comparison of BMI of mother’s who selected images that most

closely resembled their child’s body shape.

Page 160: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

159

TABLES

Table 1. Characteristics of mothers responding to an

annual survey regarding fruits and vegetables in

2011 (n=1469)

n (%) or

mean ± SD

Age (years) 36.4 ± 6.1

Number of children per mother 2.0 ± 1.0

Number of girls per mother 1.0 ± 0.9

Number of boys per mother 1.0 ± 0.9

Age of children (years) 7.6 ± 4.6

Ethnicity

White/Caucasian 1171 (79.7)

Black/African American 107 (7.3)

Hispanic/Latino 97 (6.6)

Asian/Pacific Islander 75 (5.1)

Other 19 (1.3)

Annual household income

<$25,000 274 (18.7)

$25,000-$49,999 400 (27.2)

$50,000-$74,999 348 (23.7)

$75,000-$99,999 235 (16.0)

$100,000-$149,999 161 (11.0)

>$150,000 51 (3.5)

Marital status

Married/living with someone 1178 (80.2)

Single 150 (10.2)

Separated/divorced 133 (9.1)

Widowed 8 (0.5)

Aerobic physical activity (min/day)

<30 814 (55.4)

30-60 590 (40.2)

>60 65 (4.4)

Body mass index (kg/m2) 27.5 ± 7.3

Underweight (<18.49) 49 (3.3)

Normal weight (18.5-24.9) 625 (42.6)

Overweight (25.0-29.9) 368 (25.1)

Obese (>30.0) 427 (29.1)

Fruit intake (servings/day)

2.6 ± 3.0

[2.0]a

Vegetable intake (servings/day)

2.5 ± 2.0

[2.0]a

aMedian intake of fruits or vegetables.

Page 161: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

160

Table 2. Reported home fruit/vegetable availability

versus fruit/vegetable intake in mothers responding to

an annual survey (n=1,463)

Intake

(servings/day)

Fruit Availability

Always 2.8 ± 1.5

Almost always/usually 2.2 ± 1.5a

Occasionally/rarely/never 1.4 ± 1.4a,b

Vegetable Availability

Always 2.7 ± 1.5

Almost always/usually 2.1 ± 1.5a

Occasionally/rarely/never 1.4 ± 1.2a,b

aSignificantly different from "always” available

(P<0.001). bSignificantly different from "almost always/usually”

available (P<0.001).

Page 162: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

161

Table 3. Differences in BMI based on fruit/vegetable availability in mothers responding to an annual survey regarding fruits/vegetable in

health (n=1,463)

n (%)

BMI

(kg/m2)

Unadjusted

Adjusted

β Coefficient (95%

CI) P value β Coefficient (95% CI) P value

Fruit Availability

Always 734 (50.0) 26.6 ± 6.6 Reference Reference

Almost always/usually 629 (42.8) 28.1 ± 7.9 1.56 (0.80-2.33) <0.001 1.053 (0.28-1.83)a 0.008

a

Occasionally/rarely/never 106 (7.2) 29.4 ± 7.1 2.77 (1.30-4.24) <0.001 1.912 (0.44-3.38)a 0.011

a

Vegetable Availability

Always 753 (51.3) 26.9 ± 6.8 Reference Reference

Almost always/usually 642 (43.7) 28.1 ± 7.8 1.25 (0.49-2.01) 0.001 0.628 (-0.12-1.38)b 0.100

b

Occasionally/rarely/never 74 (5.0) 27.9 ± 7.1 1.06 (-0.66-2.79) 0.229 0.068 (-1.62-1.76)b 0.937

b

aAdjusted for education and physical activity.

bAdjusted for education, physical activity, marital status, and ethnicity.

Page 163: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

162

Table 4. Mother's fruit/vegetable intake versus mother’s report of

child's body shape in mothers responding to an annual survey

regarding the role of fruits/vegetables in health

Child’s body

shape category

Fruit or

Vegetable n (%)

Mother’s Intake

(mean ± SD)

A/B/C Fruit 910

(77.8) 2.5 ± 1.6

D Fruit 123

(10.5) 2.5 ± 1.4

E Fruit 68 (5.8) 2.3 ± 1.3

F/G Fruit 68 (5.8) 2.0 ± 1.6

Total Fruit

1169

(100) a0.007

A/B/C Vegetable 912

(77.7) 2.4 ± 1.6

D Vegetable 123

(10.5) 2.5 ± 1.5

E Vegetable 69 (5.9) 2.6 ± 1.7

F/G Vegetable 69 (5.9) 2.1 ± 1.3

Total Vegetable 1173

(100) a0.809

aP for trend across body shape categories.

Page 164: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

163

References (for Appendix D)

[1] D. S. Freedman, Obesity - United States, 1988-2008, MMWR Surveill Summ, Vol. 60

Suppl, No. 2011, pp. 73-7. doi: su6001a15 [pii]

[2] D. S. Ward, A. E. Vaughn, K. I. Bangdiwala, M. Campbell, D. J. Jones, A. T. Panter,

et al., Integrating a Family-Focused Approach into Child Obesity Prevention: Rationale

and Design for the My Parenting Sos Study Randomized Control Trial, BMC Public

Health, Vol. 11, No. 2011, pp. 431. doi: 1471-2458-11-431 [pii]

10.1186/1471-2458-11-431

[3] P. M. Anderson, and K. E. Butcher, Childhood Obesity: Trends and Potential Causes,

Future Child, Vol. 16, No. 1, 2006, pp. 19-45. doi:

[4] A. H. Mokdad, B. A. Bowman, E. S. Ford, F. Vinicor, J. S. Marks, and J. P. Koplan,

The Continuing Epidemics of Obesity and Diabetes in the United States, JAMA, Vol.

286, No. 10, 2001, pp. 1195-200. doi: joc10856 [pii]

[5] F. Bianchini, R. Kaaks, and H. Vainio, Overweight, Obesity, and Cancer Risk,

Lancet Oncol, Vol. 3, No. 9, 2002, pp. 565-74. doi: S1470204502008495 [pii]

[6] M. Bryant, J. Stevens, L. Wang, R. Tabak, J. Borja, and M. E. Bentley, Relationship

between Home Fruit and Vegetable Availability and Infant and Maternal Dietary Intake

in African-American Families: Evidence from the Exhaustive Home Food Inventory, J

Am Diet Assoc, Vol. 111, No. 10, 2011, pp. 1491-7. doi: S0002-8223(11)01214-4 [pii]

10.1016/j.jada.2011.07.007

[7] L. Hall, C. E. Collins, P. J. Morgan, T. L. Burrows, D. R. Lubans, and R. Callister,

Children's Intake of Fruit and Selected Energy-Dense Nutrient-Poor Foods Is Associated

with Fathers' Intake, J Am Diet Assoc, Vol. 111, No. 7, 2011, pp. 1039-44. doi: S0002-

8223(11)00421-4 [pii]

10.1016/j.jada.2011.04.008

[8] HealthyPeople2020, Healthy People 2020: Nutrition and Weight Status, Vol. No.

2010. doi:

[9] B. J. Rolls, J. A. Ello-Martin, and B. C. Tohill, What Can Intervention Studies Tell

Us About the Relationship between Fruit and Vegetable Consumption and Weight

Management?, Nutr Rev, Vol. 62, No. 1, 2004, pp. 1-17. doi:

[10] K. He, F. B. Hu, G. A. Colditz, J. E. Manson, W. C. Willett, and S. Liu, Changes in

Intake of Fruits and Vegetables in Relation to Risk of Obesity and Weight Gain among

Middle-Aged Women, Int J Obes Relat Metab Disord, Vol. 28, No. 12, 2004, pp. 1569-

74. doi: 0802795 [pii]

10.1038/sj.ijo.0802795

[11] S. Alinia, O. Hels, and I. Tetens, The Potential Association between Fruit Intake

and Body Weight--a Review, Obes Rev, Vol. 10, No. 6, 2009, pp. 639-47. doi: OBR582

[pii]

10.1111/j.1467-789X.2009.00582.x

[12] N. Pearson, S. J. Biddle, and T. Gorely, Family Correlates of Fruit and Vegetable

Consumption in Children and Adolescents: A Systematic Review, Public Health Nutr,

Vol. 12, No. 2, 2009, pp. 267-83. doi: S1368980008002589 [pii]

10.1017/S1368980008002589

Page 165: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

164

[13] M. A. Beydoun, and Y. Wang, Parent-Child Dietary Intake Resemblance in the

United States: Evidence from a Large Representative Survey, Soc Sci Med, Vol. 68, No.

12, 2009, pp. 2137-44. doi: S0277-9536(09)00205-6 [pii]

10.1016/j.socscimed.2009.03.029

[14] Y. Wang, M. A. Beydoun, J. Li, Y. Liu, and L. A. Moreno, Do Children and Their

Parents Eat a Similar Diet? Resemblance in Child and Parental Dietary Intake:

Systematic Review and Meta-Analysis, J Epidemiol Community Health, Vol. 65, No. 2,

2011, pp. 177-89. doi: jech.2009.095901 [pii]

10.1136/jech.2009.095901

[15] S. Scaglioni, M. Salvioni, and C. Galimberti, Influence of Parental Attitudes in the

Development of Children Eating Behaviour, Br J Nutr, Vol. 99 Suppl 1, No. 2008, pp.

S22-5. doi: S0007114508892471 [pii]

10.1017/S0007114508892471

[16] M. Rasmussen, R. Krolner, K. I. Klepp, L. Lytle, J. Brug, E. Bere, et al.,

Determinants of Fruit and Vegetable Consumption among Children and Adolescents: A

Review of the Literature. Part I: Quantitative Studies, Int J Behav Nutr Phys Act, Vol. 3,

No. 2006, pp. 22. doi: 1479-5868-3-22 [pii]

10.1186/1479-5868-3-22

[17] H. Truby, and S. J. Paxton, Development of the Children's Body Image Scale, Br J

Clin Psychol, Vol. 41, No. Pt 2, 2002, pp. 185-203. doi:

[18] B. C. Tohill, J. Seymour, M. Serdula, L. Kettel-Khan, and B. J. Rolls, What

Epidemiologic Studies Tell Us About the Relationship between Fruit and Vegetable

Consumption and Body Weight, Nutr Rev, Vol. 62, No. 10, 2004, pp. 365-74. doi:

[19] T. A. Ledoux, M. D. Hingle, and T. Baranowski, Relationship of Fruit and

Vegetable Intake with Adiposity: A Systematic Review, Obes Rev, Vol. 12, No. 5, 2011,

pp. e143-50. doi: 10.1111/j.1467-789X.2010.00786.x

OBR786 [pii]

[20] J. Pomerleau, K. Lock, C. Knai, and M. McKee, Interventions Designed to Increase

Adult Fruit and Vegetable Intake Can Be Effective: A Systematic Review of the

Literature, J Nutr, Vol. 135, No. 10, 2005, pp. 2486-95. doi: 135/10/2486 [pii]

[21] K. W. Bauer, D. Neumark-Sztainer, J. A. Fulkerson, P. J. Hannan, and M. Story,

Familial Correlates of Adolescent Girls' Physical Activity, Television Use, Dietary

Intake, Weight, and Body Composition, Int J Behav Nutr Phys Act, Vol. 8, No. 2011, pp.

25. doi: 1479-5868-8-25 [pii]

10.1186/1479-5868-8-25

[22] R. Jago, T. Baranowski, and J. C. Baranowski, Fruit and Vegetable Availability: A

Micro Environmental Mediating Variable?, Public Health Nutr, Vol. 10, No. 7, 2007,

pp. 681-9. doi: S1368980007441441 [pii]

10.1017/S1368980007441441

[23] M. R. Lin BH, Higher Fruit Consumption Linked with Lower Body Mass Index.,

Food Reviews, Vol. No. 25, 2002, pp. 28-32. doi:

[24] J. Saxton, C. Hill, P. Chadwick, and J. Wardle, Weight Status and Perceived Body

Size in Children, Arch Dis Child, Vol. 94, No. 12, 2009, pp. 944-9. doi:

adc.2009.162578 [pii]

Page 166: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

165

10.1136/adc.2009.162578

[25] C. Maffeis, G. Talamini, and L. Tato, Influence of Diet, Physical Activity and

Parents' Obesity on Children's Adiposity: A Four-Year Longitudinal Study, Int J Obes

Relat Metab Disord, Vol. 22, No. 8, 1998, pp. 758-64. doi:

[26] C. Power, T. Pouliou, L. Li, R. Cooper, and E. Hypponen, Parental and Offspring

Adiposity Associations: Insights from the 1958 British Birth Cohort, Ann Hum Biol, Vol.

38, No. 4, 2011, pp. 390-9. doi: 10.3109/03014460.2011.591827

[27] E. L. Gibson, J. Wardle, and C. J. Watts, Fruit and Vegetable Consumption,

Nutritional Knowledge and Beliefs in Mothers and Children, Appetite, Vol. 31, No. 2,

1998, pp. 205-28. doi: S0195-6663(98)90180-5 [pii]

10.1006/appe.1998.0180

[28] Y. Manios, K. Kondaki, G. Kourlaba, E. Vasilopoulou, and E. Grammatikaki,

Maternal Perceptions of Their Child's Weight Status: The Genesis Study, Public Health

Nutr, Vol. 12, No. 8, 2009, pp. 1099-105. doi: S1368980008004412 [pii]

10.1017/S1368980008004412

[29] M. L. de Hoog, K. Stronks, M. van Eijsden, R. J. Gemke, and T. G. Vrijkotte,

Ethnic Differences in Maternal Underestimation of Offspring's Weight: The Abcd Study,

Int J Obes (Lond), Vol. No. 2011. doi: 10.1038/ijo.2011.199

ijo2011199 [pii]

[30] C. L. Ogden, M. D. Carroll, B. K. Kit, and K. M. Flegal, Prevalence of Obesity and

Trends in Body Mass Index among Us Children and Adolescents, 1999-2010, JAMA,

Vol. 307, No. 5, 2012, pp. 483-90. doi: jama.2012.40 [pii]

10.1001/jama.2012.40

Page 167: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

166

APPENDIX E

ADDITIONAL PUBLICATION #3:

PREDICTORS OF CARDIOMETABOLIC RISK FACTOR IMPROVEMENTS WITH

WEIGHT LOSS IN WOMEN

Authors: Caitlin A. Dow, MS; Cynthia A. Thomson, PhD, RD; Shirley W. Flatt, MS;

Nancy E. Sherwood, PhD; Njeri Karanja, PhD; Bilge Pakiz, EdD; Cheryl L. Rock, PhD,

RD

Submitted to: Journal of the American Heart Association, February 2013

Abstract

Background: Weight loss is associated with improvements in cardiometabolic risk

factors, including serum glucose, insulin, C-reactive protein (CRP), and blood lipids. Few

studies have evaluated the long-term (>18 months) effect of weight loss on these risk

factors or sought to identify factors associated with sustained improvements in these

measures.

Methods and Results: In 442 overweight/obese women (mean [SD] age of 44 [10]

years) participating in a weight loss trial, we sought to identify predictors of weight loss-

associated cardiometabolic risk factors at 12 and 24 months of intervention. Total

cholesterol (TC), LDL-cholesterol, HDL-cholesterol, non-HDL cholesterol, triglycerides,

insulin, glucose, CRP, and cardiopulmonary fitness were measured at baseline, 12

months, and 24 months. At 24 months, significant reductions in body weight, waist

circumference, CRP, TC, LDL-C, HDL-C, and non-HDL cholesterol were observed

Page 168: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

167

(P<0.05). At 24 months, mean TC and non-HDL cholesterol were reduced regardless of

the amount of weight lost, whereas reductions in LDL-C, CRP, insulin, and triglycerides

were observed only in those who lost ≥10% body weight. Step test performance

improved only in those who lost ≥10% body weight after 24 months. Change in weight

demonstrated a positive predictive value for change in cholesterol, insulin, glucose, and

triglycerides. Baseline level of the biomarker showed the greatest predictive value for

follow-up measures for insulin, cholesterol, glucose, and triglycerides.

Conclusion: Our data extend the results from short-term weight loss trials and suggest

that the magnitude of weight loss and baseline risk factors are associated with

improvements in cardiometabolic risk factors even after 24 months.

INTRODUCTION

Obesity remains a significant risk factor for disease-associated morbidity and mortality in

the U.S., including that associated with diabetes, coronary heart disease, and stroke.1

Obese individuals often present with elevated fasting values of glucose,2 lipids,

1 and

inflammatory markers,2 as well as insulin resistance.

3 Even with modest weight loss,

individuals may demonstrate significant improvements in metabolic risk factors for

obesity-related disease, particularly cardiovascular disease and diabetes.1,4-6

It is

estimated that a 10% reduction in body weight in an obese individual can promote

significant reductions in metabolic parameters including circulating glucose, insulin and

inflammatory markers, even if the body mass index (BMI) remains within an at-risk

category.7 Yet, few studies have evaluated this relationship beyond 12 months.

Page 169: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

168

The ability to promote weight loss through dietary interventions is well-established, and

behavioral weight loss programs and counseling remain a primary clinical approach to

reduce body weight in overweight and obese adults.6 These programs vary in intensity,

delivery approach, behavioral constructs applied, dietary plan and duration. But

independent of the approach, energy deficit can be achieved and is associated with

modest weight loss.3,5,8

Further, the demonstrated weight loss associated with targeted

dietary interventions generally translates to improvements in metabolic indices associated

with obesity-associated disease. 4,6,9-11

In a recently conducted randomized controlled weight loss study, women were

randomized to receive a structured weight loss program delivered either in person or via

telephone or to receive minimal diet counseling, considered usual care, for a period of 24

months. Women assigned to the structured weight loss program arms demonstrated

greater reductions in body weight, BMI, and waist circumference over time than those in

usual care condition.12

Changes in lipids and CRP according to study arm assignment

have been previously reported.12

The purpose of this analysis was to assess changes in

cardiometabolic risk factors associated with weight loss. Further, this analysis sought to

determine what factors (baseline value of biomarker, baseline weight, % change in body

weight, age, ethnicity) predicted improvement in cardiometabolic risk factors at 12 and

24 months.

Page 170: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

169

METHODS

Study Subjects and Recruitment

The weight loss intervention trial recruited 446 overweight/obese women from a screened

sample of 564 women. Women were eligible to participate if they were >18 years of age,

had a BMI between 25 and 40 kg/m2 and were a minimum of 15 kg above ideal body

weight as defined by the 1983 Metropolitan Life tables.13

Participants were recruited via

list serves and flyers distributed through each of the four study sites (University of

California, San Diego; University of Arizona, Tucson; University of Minnesota,

Minneapolis; and Kaiser Permanente Center Northwest, Portland, OR). Any woman who

was pregnant, planning a pregnancy, lactating or reported a history of eating disorders,

other psychiatric disorders, co-morbid conditions, food allergies, or was unable to

perform a 3-minute step-test for cardiopulmonary fitness assessment was excluded from

enrollment. All study participants completed the consent process and provided written

informed consent prior to randomization. The Institutional Review Boards approved this

study for Human Subjects’ Protection at each recruitment site prior to initiation. Clinical

trials Identifier: NCT00640900.

Intervention

The details of the study trial participation have been previously described.12

Briefly,

eligible women were randomized in a 3:3:2 allocation to center-based, telephone-based,

or usual care that included weight loss counseling by a dietetics professional. Women in

the center-based intervention arm visited the weight loss center weekly for brief

Page 171: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

170

counseling sessions and weigh-ins with designated, trained staff; they also had access to

web-based resource materials. Women in the telephone-based arm similarly received

weekly counseling by telephone to reinforce weight loss behaviors and goals and also had

access to web-based resource materials. Prepackaged meals and snacks provided 42-68%

of total energy needs of the prescribed eating plan and were provided free-of-charge for

participants randomized to these two study arms. Generally after the initial 12-month

period, women were transitioned to fewer pre-packaged meals and more self-selected

food items. In addition to the prepackaged foods, women assigned to the weight loss

program received one-on-one counseling to promote adherence to a low fat (20-30% total

energy), reduced energy (1200-2000 kcal), high fruit and vegetable eating plan with

recommendations for increased physical activity to achieve current guidelines14

of 30

minutes of planned exercise 5 or more days/week.

Usual care counseling sessions took place once at baseline and again at 6 months. Usual

care study participants received written educational materials to support their dietary

goals (500-1000 kcal/day deficit); regular physical activity was also promoted.

Outcomes

The primary outcome of the study was change in body weight. Weight and height (from

which BMI was calculated) were measured at baseline and every 6 months throughout

the study. Waist and hip circumference were also measured and recorded at each time

point. A 3-minute step-test was administered at baseline and repeated at 12 and 24

Page 172: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

171

months. This test measures heart rate during the first 30 seconds of recovery from

stepping.

Subjects also provided a fasting blood sample at baseline, 12, and 24 months for

evaluation of cardiometabolic risk factors. Specifically, fasting plasma was sampled to

measure lipid profiles including total cholesterol, triglycerides, and high density

lipoprotein (HDL) using enzymatic methods (Kodak Ektachem Analyzer System,

Johnson & Johnson Clinical Diagnostics, New York, NY). Low density lipoprotein

(LDL) was calculated using the Friedewald equation.15

Non-HDL cholesterol (non-

HDL-C) was calculated by subtracting HDL from the total cholesterol value. Blood

glucose was determined using enzymatic methods (Kodak Ektachem DT60 Analyzer,

Johnson & Johnson, Rochester, NY). A double antibody radioimmunoassay, with <0.2%

cross-reactivity with human proinsulin, was used for the analysis of fasting serum insulin

(Linco Research Inc., St. Charles, MO). The homeostasis model assessment of insulin

resistance (HOMA-IR) was calculated by dividing the product of fasting glucose (mg/dL)

and fasting insulin (μU/mL) by 405.16

CRP was quantified using a high sensitivity

polystyrene-enhanced turbidimetric in vitro immunoassay kit (DiaSorin Inc., Stillwater,

MN).

Statistical Analysis

Risk categories for levels of CRP,17

total cholesterol,18

HDL-cholesterol,18

LDL-

cholesterol,19

non-HDL cholesterol,20

glucose,18

insulin,21

HOMA-IR,16

waist,22

Page 173: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

172

triglycerides,18

and cardiopulmonary fitness23

were identified and used as cut-points in

tabulation.

Differences in biochemical risk factors between baseline and 12 or 24 months (overall

and stratified by biomarker risk category) were analyzed using paired t-tests. Regression

models predicted change in laboratory values as a function of ethnicity, age, baseline

levels of laboratory analyte, waist circumference, and weight change. All regression

models were adjusted for randomization assignment, cardiorespiratory fitness, and

waist/height ratio. For each term in each model, we present parameter estimates (beta

coefficients). To further explore the relationship between change in body weight and

cardiometabolic risk factors, a stratified analysis was conducted in which women were

classified as either losing ≥10% baseline body weight or losing <10% of baseline body

weight. Analyses were conducted using SAS version 9.3 (SAS Institute, Inc., Cary,

NC).

RESULTS

Four hundred and forty six overweight/obese women were enrolled; four were removed

from analysis due to post-randomization exclusions, leaving 442 in the study sample.

Thirty-five women did not complete the 12-month clinic visit, and of the 407 remaining

women, inadequate blood sample was available to complete glucose (1 subject) or lipid

(2 subjects) analysis.

Page 174: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

173

Table 1 provides baseline demographic characteristics of the study population. Women

enrolled in the study were a mean (SD) age of 44 (10) years, a mean weight of 92.1

(10.8) kg with a BMI of 33.9 (3.4), and a waist circumference of 108.6 (9.5) cm. At

baseline, 159 (36%) women were postmenopausal. At follow-up, mean weight loss as a

percentage of baseline weight was 8.6% (0.4%) in 417 subjects with available measured

body weight at 12 months, and 6.6% (0.5%) in 407 subjects with weight data at 24

months. At study entry, 2 women were using hypoglycemic agents, and 1% (n=5) were

using them at study end. Twenty-six subjects took lipid-lowering drugs at study entry, 30

women took them at 24 months, and 17 women reported estrogen use throughout the

study (data not shown).

When categorized by cardiometabolic risk level (Table 2), the median level of CRP was

3.0 mg/L, and 45% of subjects exceeded this level. There was an overall decrease in

median (interquartile range) of CRP from 3.0 (1.6-5.6) to 1.9 (1.0-4.1) after 12 months,

with the greatest decrease in women who entered the trial with a CRP value ≥3.0 mg/L.

In a sensitivity analysis excluding subjects with CRP ≥10 mg/L (n=38), paired t-tests

among subjects with CRP ≥3 mg/L and <10 mg/L still showed significant decreases in

log-transformed CRP at 12 and 24 months (data not shown). Baseline mean glucose

values were within the normal range with a mean (SD) of 94 (11) mg/dL. At both 12 and

24 months, the follow-up metabolic assessments suggested that participation in the trial

was associated with a significant increase in fasting glucose among women who entered

the trial with a glucose value <100 mg/dL, whereas at 24 months there was a significant

Page 175: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

174

decrease in glucose in those with elevated baseline levels. Baseline insulin levels were

elevated above the 15 μU/mL cut-point at an average value of 17.8 (8.9) μU/mL and were

significantly reduced after 12 months. Insulin levels significantly increased at 24 months

in those with baseline values <15 μU/mL from 11.1 (2.7) μU/mL to 13.4 (5.5) μU/mL.

Overall and in the high-risk subjects, HOMA-IR showed a pattern similar to insulin, with

a decrease at 12 months that was no longer significant at 24 months. Total cholesterol

decreased in the entire cohort after 24 months, on average by 12 mg/dL from 196 (36)

mg/dL to 184 (37) mg/dL. LDL-cholesterol was significantly decreased at 12 and 24-

month follow-up in participants with a baseline level above 130 mg/dL. HDL values

were slightly below target at 57 (16) mg/dL at baseline, and increased after 12 months of

intervention only in women with depressed baseline values (<60 mg/dL), though HDL-

cholesterol returned to baseline values by 24 months in this group. Non-HDL cholesterol

values were significantly reduced after 24 months in women who started with elevated

levels (≥160 mg/dL). Triglycerides were reduced after 12 months only in women with

elevated baseline values. In a sensitivity analysis that excluded women (n=10) who

began a lipid-lowering regimen during the trial, results in lipid changes were essentially

unchanged (data not shown). Waist circumference was significantly reduced after 12 and

24 months. At baseline, all women had a waist-to-height ratio (WHtR) above the

recommended cutoff point of 0.5. WhtR was significantly reduced from 0.66 (0.06) at

baseline to 0.60 (0.06) and 0.61 (0.07) at 12 and 24 months, respectively. In sensitivity

analyses limited to postmenopausal women, outcomes were similar to those in the entire

cohort.

Page 176: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

175

As shown in Table 3, baseline level of each biomarker was strongly and inversely

associated with follow-up measures in the individual marker. Further, age was positively

associated with cholesterol levels. Baseline weight or waist circumference was not

associated with change in any of the markers with the exception of waist circumference

being positively associated with insulin level at 12 months (R2= 0.29).

Models were stratified by weight loss percentage at both 12 months (Table 4) and 24

months (Table 5). At 12 months, 167 women lost at least 10% of initial body weight. In

these women, CRP was reduced from 2.9 mg/L to 1.2 mg/L. WHtR, total cholesterol,

and LDL-C were reduced regardless of weight loss percentage (Table 4). However, in

those who lost ≥10% of initial weight, reduction in total cholesterol was more

pronounced, dropping from a mean (SD) of 194 (38) mg/dL to 185 (38) mg/dL versus

from 198 (34) mg/dL to 192 (34) mg/dL in women losing <10% of initial weight.

Likewise, at 24 months (Table 5), total cholesterol was reduced, regardless of degree of

weight loss, reducing to 180 (32) mg/dL in those who lost ≥10% of initial weight versus

186 (33) mg/dL in those who lost <10% of initial weight. LDL-C reduction was only

maintained at 24 months in those who lost at least 10% of their weight. Non-HDL-C was

reduced in both weight loss categories at 12 and 24 months. Reductions in triglycerides,

insulin, and HOMA-IR were only evident in women who lost ≥10% body weight at both

12 and 24 months.

Page 177: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

176

Cardiopulmonary fitness (based on the 3-minute step-test) also improved in study

participants. Interestingly, this effect was independent of the degree of weight loss only

at 12 months (Table 4). After 24 months, however, cardiopulmonary fitness significantly

improved in women whose weight loss was ≥10%, while it was significantly reduced in

those who did not lose 10% of initial weight, compared to baseline values (Table 5).

DISCUSSION

In this study of overweight women, participants in the structured weight loss program lost

an average of 7% of baseline body weight at two-year follow-up. Weight loss resulted in

an improvement in numerous cardiometabolic risk factors, including biochemical values

and a functional test of cardiopulmonary fitness. Importantly, the improvements in most

risk factors were evident at both 12 and 24 months despite modest recidivism in weight

loss over time. These results are supported by several shorter-term studies, including an

8-week study by Te Morenga et al. in 83 adults, wherein weight loss ranged from 3.3 to

4.5 kg and resulted in significant reductions in LDL-cholesterol, triglycerides, and

glucose.8 A weight loss intervention by Muzio et al. resulted in significant reductions in

weight, waist circumference, triglycerides and LDL-cholesterol after 5 months.11

Short-

term studies have also demonstrated a reduction in CRP,24

but seldom to the degree

necessary to reduce clinical risk as observed in this trial. In a 12-week weight loss study

by Clifton et al. in 79 overweight/obese adult females, significant weight loss was also

demonstrated, but importantly, this is one of only a few trials that re-assessed the

Page 178: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

177

intervention efficacy longer term.10

As with the present study, long-term improvements in

glucose, insulin, CRP, and LDL-cholesterol were demonstrated.10

Longer-term studies are limited, but a few have evaluated changes in cardiometabolic

parameters over time in relation to weight loss. Four-year data from the LookAHEAD

trial, a study performed in individuals with type 2 diabetes, support our results and show

that a lifestyle intervention aimed at weight reduction results in significant reductions in

triglycerides and LDL-cholesterol.25

In one of the larger samples studied, Foster et al.

showed an 11 kg average weight loss at year 1 (n=307; 68% female) and 7 kg loss at year

2 (n=113),26

weight loss effects similar to what our sample achieved. Consistent with our

findings, triglycerides improved at year 1 and were not significantly different from

baseline at year 2 in that study. In contrast to our findings, their results suggested no

significant reduction in LDL-cholesterol at 24 months.26

In a study of 176

overweight/obese adults of which 87% were female, Burke et al. showed significant

improvements in total cholesterol and LDL:HDL ratio as well as HOMA-IR, a measure

of insulin resistance, after 12 months of intervention, changes that were generally

sustained at six-month post-intervention follow-up.27

These results, taken in

consideration with our results, suggest that efforts to favorably modulate cardiometabolic

risk factors through modest weight loss in numerous short-term trials generally translate

to longer-term follow-up, even with modest weight regain between 12 and 24 months.

Page 179: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

178

Improvements in several CVD risk factors were demonstrated in response to participation

in this weight loss study. WHtR is an adiposity index that more accurately predicts

cardiometabolic risk than BMI or waist circumference.28,29

In this study, WHtR was

reduced regardless of the degree of weight lost, though this effect was more pronounced

in women who lost ≥10% of their initial body weight. Recent evidence also indicates that

non-HDL cholesterol more accurately predicts risk of coronary heart disease than LDL-C

as it encompasses all cholesterol-containing atherogenic particles including LDL-C and

VLDL-C.30

Women with elevated baseline non-HDL cholesterol (≥160 mg/dL)

demonstrated reductions to within a normal range after 24 months, and non-HDL

cholesterol values were reduced at 12 and 24 months, regardless of the degree of weight

loss. The literature regarding lifestyle interventions for weight loss and non-HDL

cholesterol response is extremely limited, though our findings suggest that non-HDL

cholesterol is responsive to a weight loss intervention and could be included as an

outcome in future studies targeting cardiometabolic risk via weight reduction.

Previous work has demonstrated that reductions in insulin resistance are observed with

increases in physical activity, independent of weight loss.31

Other intervention studies for

weight reduction agree that exercise is a necessary component, in addition to diet

modification, to reduce insulin resistance.32

In this study, diet was the major lifestyle

modification addressed, but regular physical activity was also promoted through

counseling. In response, insulin levels and, perhaps more importantly, HOMA-IR values,

were significantly reduced at 12 months, but only in women demonstrating a weight loss

Page 180: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

179

≥10% of their initial body weight. We cannot ascertain if this effect was due solely to

changes in diet or physical activity.

This study also had the benefit of evaluating cardiopulmonary fitness, an indicator of

health also known to improve with physical activity.33

Performance improved over time

regardless of percent weight loss but was not associated with cardiometabolic factors.

These results suggest that efforts to expand outcome measures in weight loss studies

targeting improved cardiometabolic risk should consider biochemical biomarkers as well

as functional measures. Improvements in cardiopulmonary fitness have been reported in

other weight loss studies,32,34

although not consistently.35

While control for baseline values of the respective outcome (i.e. weight, biomarker) when

conducting regression analysis is sometimes applied, estimation of the predictive nature

of the baseline value is seldom evaluated.36

Our study suggests that one important

predictor of change in cardiometabolic biomarkers is the baseline level of the individual

biomarker. These data suggest that lifestyle interventions targeting weight loss are

effective for improving cardiometabolic risk in individuals presenting with unfavorable

measures. Our data suggest that these measures should be expected to show significant

and sustained improvement if a degree of weight loss is achieved and maintained.

Limitations to this study include a lack of assessment of lipoprotein particle size, known

to be associated with cardiovascular risk37

and responsive to dietary intervention.9

Page 181: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

180

Further, we did not conduct hemodynamic measures, such as blood pressure, during the

three-minute step test. These measures likely would have provided more robust data

regarding cardiopulmonary fitness.33

However, cardiopulmonary fitness was not a

primary endpoint of this study, and although less accurate than measuring maximal

oxygen uptake (VO2max) and hemodynamics during test administration, the three-minute

step test has high reliability, is sensitive to change, and has been used broadly to assess

change in cardiopulmonary fitness in clinical trials.23

Finally, we did not collect specific

information on diet or physical activity that can influence these biomarkers; however, it is

apparent that women were generally in negative energy balance given the substantial

weight loss demonstrated by so many study participants. It is well recognized that

negative energy balance can modulate many of these targeted risk factors whether that

occurs through dietary intervention or in combination with physical

activity.11,24,25,30,35,36,38,39

Strengths of the trial include the 24-month intervention and follow-up period in a

relatively large sample of women with robust and repeated measures of cardiometabolic

risk factors. The sample size and variance in weight loss also afforded an opportunity to

ascertain differences in risk factors over time in women who were more (≥10% body

weight loss) versus less (<10% body weight loss) responsive to the weight loss

interventions.

Page 182: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

181

In conclusion, participation in a weight loss intervention resulted in significant reductions

in body weight, BMI, and waist circumference after 12 and 24 months. This weight loss

was associated with significant improvements in several of the cardiometabolic risk

factors that are commonly assessed in the clinical setting. Moreover, weight loss of

≥10% after 12 months produced significant reductions in these risk factors, often

lowering the risk factor from a category of high risk to that of medium or low risk (i.e.

CRP and insulin). These data also show that baseline cardiometabolic risk factors

(cholesterol, fasting glucose, insulin, triglycerides, and CRP) have significant predictive

value in determining favorable risk reduction response to the weight loss intervention,

suggesting that these risk factors could be used to identify women more likely to respond

favorably to weight loss interventions.

Page 183: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

182

References (for Appendix E)

1. Gregg EW, Cheng YJ, Cadwell BL, Imperatore G, Williams DE, Flegal KM,

Narayan KM, Williamson DF. Secular trends in cardiovascular disease risk

factors according to body mass index in US adults. JAMA : the journal of the

American Medical Association. Apr 20 2005;293(15):1868-1874.

2. Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic

disease. J Clin Invest. Jun 2011;121(6):2111-2117.

3. Taylor R. Pathogenesis of type 2 diabetes: tracing the reverse route from cure to

cause. Diabetologia. Oct 2008;51(10):1781-1789.

4. Layman DK, Evans EM, Erickson D, Seyler J, Weber J, Bagshaw D, Griel A,

Psota T, Kris-Etherton P. A moderate-protein diet produces sustained weight loss

and long-term changes in body composition and blood lipids in obese adults. J

Nutr. Mar 2009;139(3):514-521.

5. Harvie MN, Pegington M, Mattson MP, Frystyk J, Dillon B, Evans G, Cuzick J,

Jebb SA, Martin B, Cutler RG, Son TG, Maudsley S, Carlson OD, Egan JM,

Flyvbjerg A, Howell A. The effects of intermittent or continuous energy

restriction on weight loss and metabolic disease risk markers: a randomized trial

in young overweight women. Int J Obes (Lond). May 2011;35(5):714-727.

6. Laddu D, Dow C, Hingle M, Thomson C, Going S. A review of evidence-based

strategies to treat obesity in adults. Nutr Clin Pract. Oct 2011;26(5):512-525.

7. Johnson WD, Brashear MM, Gupta AK, Rood JC, Ryan DH. Incremental weight

loss improves cardiometabolic risk in extremely obese adults. Am J Med. Oct

2011;124(10):931-938.

8. Morenga LT, Williams S, Brown R, Mann J. Effect of a relatively high-protein,

high-fiber diet on body composition and metabolic risk factors in overweight

women. Eur J Clin Nutr. Nov 2010;64(11):1323-1331.

9. Varady KA, Bhutani S, Klempel MC, Kroeger CM. Comparison of effects of diet

versus exercise weight loss regimens on LDL and HDL particle size in obese

adults. Lipids Health Dis. 2011;10:119.

10. Clifton PM, Keogh JB, Noakes M. Long-term effects of a high-protein weight-

loss diet. Am J Clin Nutr. Jan 2008;87(1):23-29.

11. Muzio F, Mondazzi L, Harris WS, Sommariva D, Branchi A. Effects of moderate

variations in the macronutrient content of the diet on cardiovascular disease risk

factors in obese patients with the metabolic syndrome. Am J Clin Nutr. Oct

2007;86(4):946-951.

12. Rock CL, Flatt SW, Sherwood NE, Karanja N, Pakiz B, Thomson CA. Effect of a

free prepared meal and incentivized weight loss program on weight loss and

weight loss maintenance in obese and overweight women: a randomized

controlled trial. JAMA. Oct 27 2010;304(16):1803-1810.

13. 1983 metropolitan height and weight tables. Stat Bull Metrop Life Found. Jan-Jun

1983;64(1):3-9.

14. Services. UDoHaH. 2008 physical activity guidelines for Americans. . In:

Services. UDoHaH, ed. Atlanta, GA.2008.

Page 184: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

183

15. Fukuyama N, Homma K, Wakana N, Kudo K, Suyama A, Ohazama H, Tsuji C,

Ishiwata K, Eguchi Y, Nakazawa H, Tanaka E. Validation of the Friedewald

Equation for Evaluation of Plasma LDL-Cholesterol. J Clin Biochem Nutr. Jul

2008;43(1):1-5.

16. Kirk E, Reeds DN, Finck BN, Mayurranjan SM, Patterson BW, Klein S. Dietary

fat and carbohydrates differentially alter insulin sensitivity during caloric

restriction. Gastroenterology. May 2009;136(5):1552-1560.

17. Ridker PM. High-sensitivity C-reactive protein and cardiovascular risk: rationale

for screening and primary prevention. Am J Cardiol. Aug 21 2003;92(4B):17K-

22K.

18. Lichtenstein AH, Appel LJ, Brands M, Carnethon M, Daniels S, Franch HA,

Franklin B, Kris-Etherton P, Harris WS, Howard B, Karanja N, Lefevre M, Rudel

L, Sacks F, Van Horn L, Winston M, Wylie-Rosett J. Diet and lifestyle

recommendations revision 2006: a scientific statement from the American Heart

Association Nutrition Committee. Circulation. Jul 4 2006;114(1):82-96.

19. Wilson PW, D'Agostino RB, Levy D, Belanger AM, Silbershatz H, Kannel WB.

Prediction of coronary heart disease using risk factor categories. Circulation. May

12 1998;97(18):1837-1847.

20. Executive Summary of The Third Report of The National Cholesterol Education

Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High

Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA : the journal of

the American Medical Association. May 16 2001;285(19):2486-2497.

21. Cornier MA, Donahoo WT, Pereira R, Gurevich I, Westergren R, Enerback S,

Eckel PJ, Goalstone ML, Hill JO, Eckel RH, Draznin B. Insulin sensitivity

determines the effectiveness of dietary macronutrient composition on weight loss

in obese women. Obes Res. Apr 2005;13(4):703-709.

22. Kirk EP, Klein S. Pathogenesis and pathophysiology of the cardiometabolic

syndrome. J Clin Hypertens (Greenwich). Dec 2009;11(12):761-765.

23. McArdle WD, Katch FI, Pechar GS, Jacobson L, Ruck S. Reliability and

interrelationships between maximal oxygen intake, physical work capacity and

step-test scores in college women. Med Sci Sports. Winter 1972;4(4):182-186.

24. Camhi SM, Stefanick ML, Ridker PM, Young DR. Changes in C-reactive protein

from low-fat diet and/or physical activity in men and women with and without

metabolic syndrome. Metabolism. Jan 2010;59(1):54-61.

25. Wing RR. Long-term effects of a lifestyle intervention on weight and

cardiovascular risk factors in individuals with type 2 diabetes mellitus: four-year

results of the Look AHEAD trial. Arch Intern Med. Sep 27 2010;170(17):1566-

1575.

26. Foster GD, Wyatt HR, Hill JO, Makris AP, Rosenbaum DL, Brill C, Stein RI,

Mohammed BS, Miller B, Rader DJ, Zemel B, Wadden TA, Tenhave T,

Newcomb CW, Klein S. Weight and metabolic outcomes after 2 years on a low-

carbohydrate versus low-fat diet: a randomized trial. Ann Intern Med. Aug 3

2010;153(3):147-157.

Page 185: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

184

27. Burke LE, Hudson AG, Warziski MT, Styn MA, Music E, Elci OU, Sereika SM.

Effects of a vegetarian diet and treatment preference on biochemical and dietary

variables in overweight and obese adults: a randomized clinical trial. Am J Clin

Nutr. Sep 2007;86(3):588-596.

28. Ashwell M, Gunn P, Gibson S. Waist-to-height ratio is a better screening tool

than waist circumference and BMI for adult cardiometabolic risk factors:

systematic review and meta-analysis. Obes Rev. Mar 2012;13(3):275-286.

29. Ashwell M, Hsieh SD. Six reasons why the waist-to-height ratio is a rapid and

effective global indicator for health risks of obesity and how its use could

simplify the international public health message on obesity. Int J Food Sci Nutr.

Aug 2005;56(5):303-307.

30. Rana JS, Boekholdt SM, Kastelein JJ, Shah PK. The role of non-HDL cholesterol

in risk stratification for coronary artery disease. Curr Atheroscler Rep. Apr

2012;14(2):130-134.

31. Earnest CP, Poirier P, Carnethon MR, Blair SN, Church TS. Autonomic function

and change in insulin for exercising postmenopausal women. Maturitas. Mar

2010;65(3):284-291.

32. Blumenthal JA, Babyak MA, Sherwood A, Craighead L, Lin PH, Johnson J,

Watkins LL, Wang JT, Kuhn C, Feinglos M, Hinderliter A. Effects of the dietary

approaches to stop hypertension diet alone and in combination with exercise and

caloric restriction on insulin sensitivity and lipids. Hypertension. May

2010;55(5):1199-1205.

33. Church TS, Earnest CP, Skinner JS, Blair SN. Effects of different doses of

physical activity on cardiorespiratory fitness among sedentary, overweight or

obese postmenopausal women with elevated blood pressure: a randomized

controlled trial. JAMA : the journal of the American Medical Association. May 16

2007;297(19):2081-2091.

34. Borel AL, Nazare JA, Smith J, Almeras N, Tremblay A, Bergeron J, Poirier P,

Despres JP. Visceral and not subcutaneous abdominal adiposity reduction drives

the benefits of a 1-year lifestyle modification program. Obesity (Silver Spring).

Jun 2012;20(6):1223-1233.

35. Christiansen T, Paulsen SK, Bruun JM, Pedersen SB, Richelsen B. Exercise

training versus diet-induced weight-loss on metabolic risk factors and

inflammatory markers in obese subjects: a 12-week randomized intervention

study. Am J Physiol Endocrinol Metab. Apr 2010;298(4):E824-831.

36. Albu JB, Heilbronn LK, Kelley DE, Smith SR, Azuma K, Berk ES, Pi-Sunyer

FX, Ravussin E. Metabolic changes following a 1-year diet and exercise

intervention in patients with type 2 diabetes. Diabetes. Mar 2010;59(3):627-633.

37. Krauss RM. Lipoprotein subfractions and cardiovascular disease risk. Curr Opin

Lipidol. Aug 2010;21(4):305-311.

38. Due A, Larsen TM, Mu H, Hermansen K, Stender S, Astrup A. Comparison of 3

ad libitum diets for weight-loss maintenance, risk of cardiovascular disease, and

diabetes: a 6-mo randomized, controlled trial. Am J Clin Nutr. Nov

2008;88(5):1232-1241.

Page 186: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

185

39. Lavoie ME, Faraj M, Strychar I, Doucet E, Brochu M, Lavoie JM, Rabasa-Lhoret

R. Synergistic associations of physical activity and diet quality on

cardiometabolic risk factors in overweight and obese postmenopausal women. Br

J Nutr. May 9 2012:1-10.

Page 187: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

186

Tables

Table 1. Baseline Characteristics of Overweight / Obese Women Enrolled in a

Weight Loss Study (n=442)

Characteristic Baseline Value, Mean (SD)

Age (years) 44(10)

Weight (kg) 92.1(10.8)

Height (cm) 164.9(6.3)

BMI (kg/m2) 33.9(3.4)

Waist Circumference (cm) 108.6(9.5)

Hip Circumference (cm) 120.2(8.2)

Waist to Hip Ratio 0.9(0.06)

Postmenopausal (%) 159 (36%)

Education (n)

High School or less 52

Some College 189

College Graduate 95

Graduate School 106

Page 188: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

187

Table 2: Change in Cardiometabolic Risk Factors in Relation to Baseline Values, Stratified

by Clinical Risk

Risk Factor Baseline

Mean (SD)

n=442

n1 12 Months

Mean (SD)

n=407

n1 24 Months

Mean (SD)

n=383

Glucose, Mean (SD) 94 (11) 406 95 (18) 383 94 (13)

<100 mg/dL 89 (7) 310 91 (9) ‡

293 91 (10) †

≥100 mg/dL 110 (19) 96 107 (32) 90 101 (18) ‡

Insulin, Mean (SD) 17.8 (8.9) 407 15.6 (9.8) ‡ 383 18.5 (11.2)*

<15 μU/ml 11.1 (2.7) 180 11.0 (4.5) 174 13.4 (5.5) ‡

≥15 μU/ml 23.1 (8.4) 227 19.3 (11.2) ‡ 209 22.7 (12.8)

HOMA-IR2 4.24 (2.53) 405 3.72 (2.58)

‡ 383 4.37 (3.10)*

<3 2.27 (0.54) 138 2.39 (1.11) 135 2.95 (1.38) ‡

≥3 5.26 (2.55) 267 4.41 (2.84) ‡ 248 5.15(3.49)

CRP mg/L, Median (IQR) 3.0 (1.6, 5.6) 407 1.9 (1.0, 4.1) † 383 2.1 (1.0, 4.0)

CRP <1 mg/L

(low risk)

0.6 (0.2) 38 0.8 (0.6) 35 1.0 (0.7) †

CRP 1-2.99 mg/L

(medium risk)

1.9 (0.6) 169 1.9 (3.9) † 164 3.1 (12.5)

CRP ≥ 3 mg/L

(high risk)

6.8 (4.7) 200 5.1(6.8) † 184 5.4 (6.2)*

Total Cholesterol mg/dL

Mean (SD)

196 (36) 406 189 (36) ‡

383 184 (37) ‡

<200 mg/dL 171 (21) 228 172 (28) 215 168 (27)

≥200 mg/dL 229 (21) 178 211 (34) ‡

168 204 (37) ‡

Page 189: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

188

LDL Cholesterol mg/dL

Mean (SD)

117 (33) 405 111 (34) ‡ 382 113 (37)*

<130 mg/dL 99 (20) 266 99 (27) 255 104 (33)*

≥130 mg/dL 154 (17) 139 133 (35) ‡ 127 132 (38)

HDL Cholesterol mg/dL,

Mean (SD)

57 (16) 405 57 (16) 383 53 (15) ‡

<60 mg/dL 47 (7) 252 52 (13) ‡ 239 47 (10)

≥60 mg/dL 73 (12) 153 66 (15) ‡ 144 63 (17)

Non-HDL Cholesterol

mg/dL

134 (36) 405 132 (35) ‡ 383 131 (38)

<160 mg/dL 123 (34) 294 121 (28) 280 120 (30)

≥160 mg/dL 186 (18) 111 162 (38) ‡ 103 159 (42)

Triglycerides mg/dL,

Mean (SD)

110 (57) 406 106 (55)* 383 117 (93)

<150 mg/dL 91 (26) 337 93 (34) 317 100 (43) ‡

≥150 mg/dL 206 (69) 69 168 (89) ‡ 66 195 (186)

Waist cm, Mean (SD) 108.3 (9.4) 409 99.0 (10.1) ‡ 392 101.1 (10.7)

<88 cm 86.7 (1.2) 4 86.0 (11.3) 4 82.9 (6.5)

≥88 cm 108.8 (9.3) 405 99.2 (10.0) ‡ 388 101.3 (10.5)

Waist/Height Ratio

<0.5 N/A

≥0.5 0.66 (0.06) 409 0.60 (0.06) ‡ 392 0.61 (0.07)

*P<.05, †P<.01,

‡P <.001

1Sample size represents n for paired t tests at 12 or 24 months and varies based on completion of 12- or 24-

month clinic activities. 2Homeostasis model for insulin resistance. Insulin sensitive is <3; insulin resistant is ≥3.

Page 190: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

189

Table 3: Predictors of Cardiometabolic Biomarker Change

Models Predicting 12-Month Biomarker Values (n=406)

Insulin

R2 = .37

Cholesterol

R2 = .46

Glucose

R2 = .26

TG

R2 = .66

CRP

R2 = .16

Baseline level of

biomarker

0.551‡

0.597‡

0.786‡

0.752‡

0.470‡

Baseline weight -0.057 -0.093 0.069 0.286 0.041

% change in weight 0.274‡ 0.403* 0.351

‡ 1.514

‡ 0.091

Baseline waist 0.294* 0.409 0.211 -0.585 0.013

Age -0.025 0.520‡ 0.016 0.301 -0.021

White

race/ethnicity

-2.022* 5.980 -0.964 1.428 -0.420

Models Predicting 24 Month Biomarker Values (n=383)

Insulin

R2 = .52

Cholesterol

R2 = .39

Glucose

R2 = .31

TG

R2 = .56

CRP

R2 = .07

Baseline level of

biomarker

0.770‡ 0.505

‡ 0.525

‡ 1.150

‡ 0.430

Baseline weight 0.073 -0.300 0.104 0.673 -0.060

% change in weight 0.399‡ 0.544

† 0.406

‡ 1.788

‡ 0.064

Baseline waist -0.173 0.847 -0.306 -2.147 -0.029

Age 0.067 0.637‡ 0.064 0.817* -0.098*

White

race/ethnicity

-0.744 5.592 -1.137 -3.287 -2.545*

*P<.05, †P<.01,

‡P <.001. Values shown are beta coefficients in a regression model for each biomarker at

each time period, adjusted for randomization assignment, step test heart rate, and waist/height ratio. CRP

refers to C-Reactive Protein; TG refers to triglycerides.

Page 191: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

190

Table 4: Change in Cardiometabolic Risk Factors, Stratified by Percent Weight Loss

after 12 Months

Lost 10% weight after 12

months (n=167)

Did not lose 10% weight

after 12 months (n=240)

Baseline 12 Months Baseline 12 Months

Weight kg, Mean (SD) 93.2(11.3) 77.3(10.7)‡ 91.0(10.3) 88.3(11.2)

Waist cm, Mean (SD) 109.1(9.5) 93.8(8.5)‡ 107.8(9.4) 102.7(9.5)

Waist/Height Ratio 0.66(0.06) 0.58(0.05) ‡ 0.66(0.06) 0.63(0.06)

Body Mass Index,

Mean (SD)

33.9(3.4) 28.2(3.4)‡ 33.7(3.4) 32.7(3.8)

CRP mg/L, Median (IQR) 2.9(1.5, 5.7) 1.2(0.7, 2.6)† 3.1(1.7, 5.5) 2.4(1.3, 4.9)

CRP <1 mg/L

(low risk %)

9.0 40.4‡ 9.2 15.8

CRP 1-2.99 mg/L

(medium risk %)

41.9 40.1 39.2 39.4

CRP ≥3 mg/L

(high risk %)

49.1 19.3‡ 51.6 44.8*

Total Cholesterol mg/dL,

Mean (SD)

194(38) 185(38)† 198(34) 192(34)

LDL mg/dL, Mean (SD) 115(34) 109(37)* 119(32) 112(32)‡

HDL mg/dL, Mean (SD) 57(15) 57(15) 56(16) 57(16)

Non-HDL mg/dL, Mean(SD) 136 (38) 128 (39)† 142 (35) 135 (34)

TG mg/dL, Mean (SD) 107(48) 94(39)‡ 113(64) 114(63)

Page 192: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

191

Glucose mg/dL, Mean (SD) 94(11) 93(24) 94(11) 96(13)†

Insulin uU/mL, Mean (SD) 16.9(9.1) 12.9(10.0)‡ 18.2(8.7) 17.5(9.2)

HOMA-IR, % Insulin

Resistant1

62% 35%‡ 68% 65%

Step Test Heart

Rate/30 sec, Mean (SD)

54(10) 46(9)‡ 54(9) 50(9)

Step test poor to

fair (%)

68.5 30.7‡ 70.2 58.4

Step test good to

outstanding (%)

31.5 69.3‡ 29.8 41.6

*P<.05, †P<.01,

‡P <.001.

1Homeostasis model for insulin resistance. Insulin sensitive is <3; insulin

resistant is ≥3.

Page 193: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

192

Table 5: Change in Cardiometabolic Risk Factors, Stratified by Percent Weight Loss

after 24 Months

Lost 10% weight after 24

months (n=130)

Did not lose 10% weight

after 24 months (n=253)

Baseline 24 Months Baseline 24 Months

Weight kg, Mean (SD) 93.3(10.3) 77.3(10.1) ‡ 91.0(10.9) 89.7(11.9)

Waist cm, Mean (SD) 109.0(9.2) 94.8(8.9) ‡ 107.8(9.5) 104.2(9.8)

Waist/Height Ratio 0.66(0.06) 0.57(0,06) ‡ 0.66(0.06) 0.63(0.06)

Body Mass Index,

Mean (SD)

34.0(3.3) 28.2(3.4) ‡ 33.6(3.4) 33.2(4.0)

CRP mg/L, Median (IQR) 2.9(1.6, 5.5) 1.3(0.7, 2.8) ‡ 3.0(1.7, 5.6) 2.5(1.3, 5.2)

CRP <1 mg/L

(low risk %)

7.7 42.3‡ 9.9 13.8

CRP 1-2.99 mg/L

(medium risk %)

44.6 33.1 39.1 43.9

CRP ≥3 mg/L

(high risk %)

47.7 24.6‡ 51.0 42.4

Total Cholesterol mg/dL,

Mean (SD)

194(38) 180(32) ‡ 197(35) 186(33)

LDL mg/dL, Mean (SD) 116(34) 108(34)* 118(32) 116(39)

HDL mg/dL, Mean (SD) 57(15) 56(14) 57(16) 52(15) ‡

Non-HDL mg/dL 137(38) 124(43) ‡ 140(35) 134(35)

TG mg/dL, Mean (SD) 104(45) 92(33) ‡ 114(56) 124(61)

Page 194: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

193

Glucose mg/dL, Mean

(SD)

93(10) 89(11) ‡ 94(11) 96(14)

Insulin uU/mL, Mean (SD) 16.3(8.1) 13.3(6.2) ‡ 18.1(8.6) 21.1(12.2)

HOMA-IR, % Insulin

Resistant1

61% 39%‡ 67% 78%

Step Test Heart

Rate/30 sec, Mean (SD)

54(9) 46(8) ‡ 54(10) 50(9)

Step test poor to

fair (%)

68.5 36.8‡ 46.1 87.1

Step test good to

outstanding (%)

31.5 63.8‡ 53.9 12.9

*P<.05, †P<.01,

‡P <.001.

1Homeostasis model for insulin resistance. Insulin sensitive is <3; insulin

resistant is ≥3.

Page 195: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

194

APPENDIX F

ADDITIONAL PUBLICATION #4:

USE OF HERBAL, BOTANICAL, AND NATURAL PRODUCT SUPPLEMENTS IN

ONCOLOGY AND DIABETES POPULATIONS

Authors: Caitlin Dow, MS and Maureen Leser, MS, RD, CSO, LD

In press: Oncology Nutrition Connection

Introduction

It is well recognized that herbal and botanical products exert biologic effects. Over 50%

of commercial drugs are derived from bioactive compounds of non-human origin, many

of which are plants (1). As increasing numbers of Americans have became disillusioned

with perceived over-use, adverse side effects, and high cost of prescription drugs, they

have turned to herbal and botanical supplements, which they consider natural and safer

than synthetic medications (2). A new medical diagnosis can also stimulate lifestyle

changes including an increase in the use of dietary supplements (3).

Dietary supplement use, particularly multivitamins, is highly prevalent in the United

States. Use is reportedly higher in older, female, overweight/obese individuals as well as

those with health concerns. Data from the 2003-2006 National Health and Nutrition

Examination Survey (NHANES) indicate that approximately 54% of adults (excluding

pregnant women) and 70% of adults >71 years take multi-vitamin and mineral

supplements while 20% of Americans use a supplement with at least one botanical

ingredient (4). A survey published in 2008 by the National Center for Complementary

and Alternative Medicine (NCCAM) indicated that herbal therapy or use of natural

products other than vitamins and minerals was the second most prevalent alternative

Page 196: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

195

medicine modality (excluding prayer), and used by 18.9% of adults (5). Individuals with

chronic disease, including cancer and diabetes mellitus, frequently supplement their diets

with herbal and natural therapies, possibly due to real or perceived limitations of

conventional treatments (6).

The evidence base for herbal/dietary supplements used to prevent or improve therapeutic

outcomes in diabetes and cancer remains limited and concern over potential harm

remains (6-7). Because of the widespread use of these products, it is important for the

registered dietitian (RD) to understand and recognize potential benefits as well as

potential adverse events associated with supplementation. This paper briefly reviews

usage of supplements in oncology and diabetes patients, potential biological benefits and

risks of select supplements used by each population, the current legislative framework for

dietary supplements, and the role of the RD in counseling patients on supplement usage.

Chronic Disease and Dietary Supplements

Adults with cancer or other chronic conditions are more likely to report use of

supplements than healthy populations (8). Supplement usage is widespread amongst

oncology patients, and data indicate that usage typically ranges from 30-75% in these

patients (9). One pilot study of 140 oncology patients indicated that 52% were taking

some kind of dietary supplements and, of those, 23% were using herbal supplements.

Demographic factors associated with herbal supplement use were female gender, age,

fatigue, cancer pain, and presence of metastasis (7). A subset of participants enrolled in

Page 197: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

196

the American Cancer Society’s Study of Cancer Survivors-I (SCS-I) self-administered a

dietary supplement survey. Of 827 surveys included in the review, approximately 97% of

participants had undergone conventional therapy for their cancer, yet 69.3% also reported

using dietary supplements after their cancer diagnosis. Garlic, echinacea, ginseng, black

cohosh, and gingko biloba were included in the list of supplements (other than

multivitamins) taken by 10 or more people in this study (10).

Similar to patients diagnosed with cancer, supplementation with herbal/dietary agents

also is common in people diagnosed with diabetes. Those with diabetes often report use

of dietary supplements and other alternative medicines because of a desire to take an

active role in their health, to improve their quality of life, or due to a belief that

conventional therapies do not work or are too expensive (11). Data from the 2002 and

2007 National Health Interview Study found that, of the 4,150 individuals diagnosed with

Type 1 or Type 2 Diabetes, approximately 34% were taking at least one non-vitamin/non-

mineral dietary supplement (11). A recent retrospective study in 459 adults with diabetes

indicated that 55% used some form of vitamin, mineral, or herbal supplement on a daily

basis (12). Interestingly, the use of non-mineral/non-vitamin supplements was twice as

common among type 2 diabetics as compared to type 1 diabetics (39% vs. 20%,

respectively) (12). Among the more frequently consumed dietary supplements were

cinnamon, coenzyme Q10, chromium, and alpha-lipoic acid.

Target Mechanisms for Diet Supplementation in Cancer and Diabetes

The use of dietary supplements is often driven by an assumption that such products may

Page 198: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

197

modify health by targeting specific pathology associated with disease. For cancer patients

and/or those with a diagnosis of diabetes this may include modulation of inflammation,

oxidative stress, insulin resistance, and/or hyperglycemia. Evidence exists to suggest that

some compounds do favorably alter these biological responses. Curcumin may reduce

inflammation through its ability to inhibit cyclooxygenase-2 (COX-2) (13). Alpha-lipoic

acid is an endogenously produced antioxidant, though there is some data that

supplementation also improves insulin sensitivity (14). Evidence indicates that ginseng

and magnesium may improve insulin secretion/sensitivity (15-16) while chromium and

oat bran may improve glycemic control in patients with Type 2 Diabetes (17-18).

Supplements with anti-cancer properties often modulate cell cycles and help eliminate

abnormal cells that may become cancerous. Allyl sulfides, a variety of which make up

94% of compounds in garlic, are believed to suppress in vitro and in vivo growth of

multiple types of cancer cells via apoptosis and cell cycle arrest (19). Unfortunately,

evidence remains inconclusive for the use of many supplements in individuals with health

issues, as a large number of studies in this field are limited by small sample sizes and/or

poor designs.

Legislation

Though plants have long been considered the “medicine of mankind,” it was reasoned

that scientific discoveries, growth and sophistication of the pharmaceutical industry and

government oversight would give consumers what nature could not - safe and effective

medicines that are consistent from dose to dose. But a series of events in the early to mid

Page 199: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

198

20th

century cast doubt on this assumption and prompted Congress to pass regulations

(20-23) intended to establish a pharmaceutical industry based on “purity, truth in

labeling, and effectiveness” (20). The Dietary Supplement and Health Education Act

(DSHEA) was passed almost 20 years ago to define and provide a regulatory framework

for dietary supplements. To prevent manufacturers from making unsubstantiated claims

regarding supplements, DSHEA established specific guidelines for dietary supplement

labels. Table 1 summarizes those guidelines.

Table 1: Permissible Dietary Supplement Label Claims and Guidance as established

by DSHEA

Three Categories of Claims Allowed for Dietary Supplements (24):

Health Claims Describe the connection between a nutrient or food substance and a disease or health

related condition. FDA reviews scientific evidence or statements from authoritative

scientific bodies before approving health claims.

Nutrient Content

Claims

Describe the level of a nutrient in a food or dietary supplement.

Structure/function

Claims

Describe the role of a nutrient or dietary supplement intended to affect the structure

or function of the body, the mechanism of how it maintains that structure or

function, or general well being. FDA does not pre-approve structure/function claims

so they must include the disclaimer “This statement has not been evaluated by the

Food and Drug Administration. This product is not intended to diagnose, treat, cure,

or prevent any disease.” Structure/function claims must be substantiated with

“competent and reliable scientific evidence”.

Page 200: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

199

Health Risks of Herbal Supplements

Critics of DSHEA argue that herbal supplements suffer from the same safety concerns

that existed when pharmaceutical medicines were first developed; that a lack of

standardization and quality control poses risk to the integrity of the product and consumer

safety. In fiscal year 2011, FDA records indicate that 1,777 mandatory adverse event

reports were submitted from the dietary supplement industry (25), while US Poison

Control Centers recorded 29,000 calls regarding use of dietary supplements in 2009, with

500 related to moderate to severe events (26). Potential safety risks posed by dietary

supplements may be mitigated by evidence-based patient education provided by RDs.

Herbal Supplements, Medical Practice, and the role of the RD

Significant numbers of Americans do not share information regarding use of dietary

supplements with their health care team; one study suggested that 70% of patients

surveyed failed to disclose their use of herbal medicine during preoperative assessment

(27). Failure to communicate with health professionals regarding use of dietary

supplements may result in unidentified adverse interactions between prescribed

medication, over-the-counter medication, foods, and supplements. As distinctions

between fortified foods, functional foods, medical foods and dietary supplements

continue to narrow, the need for patient education becomes more important. RDs

routinely evaluate the use of dietary supplements within a comprehensive nutrition

assessment, and should integrate education and counseling on supplements into their

nutrition practices.

Page 201: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

200

The Academy of Nutrition and Dietetics describe RDs as “consumers’ bridge

between evidence-based research and optimal health” (28). The Academy’s Position

Paper on Functional Foods states that RDs should incorporate functional food

assessment into evidence-based nutrition practice.

RD knowledge of DSHEA provides a strong background for answering legislative

questions on herbal and other dietary supplements.

RDs should provide patient education and report (MedWatch) any potential

interactions between herbal supplements and prescription drugs.

RDs already examine interrelationships between food, functional foods, and

prescribed drugs on nutrition outcomes. Assessing effects of herbal supplements,

either on promoting or managing nutritional health, is an appropriate extension of

that role.

Conclusion:

Individuals with cancer and/or diabetes commonly consume dietary supplements. Some

have demonstrated efficacy in nutritional therapy, others do not, and still others may be

associated with adverse health consequences. The unique knowledge and skill set of the

RD positions her/him as a provider of assessment and education on evidence-based

information on benefits, risks and potential interactions between dietary supplements,

food, and prescription as well as non-prescription medicines. Tables 2 and 3 summarize

Page 202: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

201

current evidence of select supplements commonly used for the prevention and/or

treatment of cancer and diabetes.

Page 203: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

202

Table 2: Evidence of Efficacy of Select Herbal Supplements used by Cancer Survivors Agent

Reported Benefits/Side

Effects

Evidence RD Message

Black Cohosh

(Cimmicifua

Racemosa):

Rhizome and roots used

in herbal treatments;

Remifemin®, a

commercial preparation

of black cohosh,

provides 10 mg

root/rhizome per tablet.

Reported Benefits: Suppress hot flashes in

menopausal women;

cancer treatment

Potential Side Effects: May be toxic to the liver

(29); may interfere with

some cancer treatments

Meta-analysis concludes that evidence is

insufficient for recommending black

cohosh as a treatment for menopausal

symptoms (30).

In a six month study, black cohosh was

more effective than fluoxetine for treating

hot flushes and night sweats associated

with menopause (31)

German Commission E has found black

cohosh to be effective at treating nervous

symptoms associated with menopause (32)

No evidence suggests black cohosh may be

effective as an anticancer treatment, but it

may interfere with conventional cancer

treatments including tamoxifen (33), and

may increase toxic effects of doxorubicin

and docetaxel (34).

Some studies suggest that Black Cohosh

may reduce menopausal symptoms, but

the body of evidence does not support its

use.

No evidence supports efficacy in cancer

treatment, and black cohosh may

interfere with some conventional cancer

treatments as well as atorvastatin

azathioprine, and ctclosporine.

Garlic:

Perennial bulb used in

cooking and as an

herbal treatment

Reported Cancer

Benefit: Garlic may

stimulate apoptosis and

control the cell cycle

Potential Side Effects: May interfere with

function of some

prescription drugs,

including saquinavir and

antiplatelet medications

200 milligrams (mg) synthetic allitridum

and 100 micrograms (mcg) selenium given

every other day reduced risk for all tumors

by 33% and risk of stomach cancer by 52%

as compared to placebo (35).

Ajoene, a compound found in crushed

garlic, stimulated apoptosis in

promyeloleukemic cells (36).

Allyl sulfides, which comprise 94% of

compounds in garlic, may promote

apoptosis, cell cycle arrest, and inhibit

growth of tumor cells (19).

Garlic contains compounds that show

potential as anti-cancer treatments.

However, evidence is insufficient for

recommending garlic supplements for

cancer treatment. Garlic may interfere

with the activity of some medications, in

particular anticoagulant drugs.

The World Health Organization (WHO)

describes an average daily dose of garlic

as 1-5 grams fresh garlic; 0.4-1.2 grams

(g) dried powder; 205 mg garlic oil, and

300-1000 mg garlic extract (37).

Ginger (Zingiber

officinale):

Rhizome is used as an

Reported Cancer

Benefit: Manage nausea

associated with

0.5-1.0 g ginger/day significantly reduced

severity of acute chemotherapy-induced

nausea in adult cancer patients (38).

Some but not all studies suggest that

ginger may reduce nausea and vomiting

associated with chemotherapy. However,

Page 204: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

203

herbal treatment.

chemotherapy

Potential Side Effects: GI complaints reported

with powdered ginger;

may prolong effect

platelet activity

1.0-2.0 g ginger daily for 3 days, given with

antiemetic medicine, did not reduce the

prevalence or severity of acute or delayed

nausea (39).

evidence is insufficient for

recommending ginger supplements for

anti-emetic treatment.

Ginger may interfere with the activity of

some medications, in particular

anticoagulant drugs.

Gingko Biloba:

Seeds and leaves are

used in herbal

treatments.

Reported Cancer

Benefit: May inhibit

proliferation of cancer

cells

Potential Side Effects:

Side effects are

uncommon; may

potentiate effects of

monoamine oxidase

inhibitors (MAO-I),

anticoagulants, and anti-

platelets

Gingko Evaluation of Memory (GEM)

study found that those who received gingko

(as opposed to a placebo) were not less

likely to develop cancer over a 6-year

period (40).

Treatment of pancreatic cell lines with

70uM kaempferol (an active component of

gingko) for 4 days significantly inhibited

cell proliferation.

Combining kaempferol with 5-fU increased

the inhibitory effect on cell growth (41).

Cell studies suggest that researchers

should explore the anticancer potential

of gingko but at this time evidence is

insufficient for recommending gingko

for cancer treatment

Gingko may interfere with the activity of

some medications, in particular

anticoagulant and anticonvulsant drugs

and MAO-I inhibitors.

St. John’s Wort

(Hypericum

perforatum):

Bush that blooms

around June 24th

, the

birthday of St. John the

Baptist. Yellow flowers

from this bush are used

as herbal remedies.

Reported Cancer

Benefit:

May make caner cells

more sensitive to light

therapies

Potential Side Effects:

May interact with many

medications including

irinotecan, imatinib, and

immunosuppressant

medication.

In VITamins And Lifestyle cohort (VITAL

study) use of St. John’s Wort was inversely

associated with risk of colorectal cancer

(42).

Cell and animal studies suggest that St.

John’s wort may make cancer cells more

sensitive to light therapies, and therefore

may have potential to improve anti-cancer

effects of photodynamic therapy (43).

Most studies do not suggest a role for St.

John’s wort in cancer treatment, and this

herb may interfere with the activity of

some chemotherapy agents.

Curcumin (cucurma

longa):

Found in turmeric; the

rhizome is used in

herbal treatments.

Reported Cancer

Benefit: May inhibit

growth of cancer cells;

has anti-inflammatory

properties

Potential Side Effects:

May interact with

Inhibits growth of esophageal cancer cells

in vitro, (44-45) and modulates signaling

factors that decrease cell proliferation of

prostate cancer cells (46).

4 grams of curcumin daily for 30 days is

commonly used in clinical trials.

8,000 mg/day was maximal tolerated dose

Emerging research suggests that

curcumin should undergo further study

for anticancer effects, but at this time

evidence is insufficient for

recommending curcurmin for cancer

treatment

Curcumin may interfere with the activity

Page 205: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

204

cyclophosphamide and

doxorubicin; also may

interact with medicines

used to manage blood

coagulation.

in Phase I trial for advanced and metastatic

breast cancer Some biological and clinical

responses were seen, but many patients

dropped out because of side effects (47).

of some anticancer medications.

The bioavailability of curcumin is poor,

which increases pill burden and potential

for side effects.

Page 206: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

205

Table 3. Evidence of Efficacy of Select Herbal Supplements used by Patients with Diabetes

Agent

Reported

Benefit /

Concern

Evidence RD Message

Fenugreek

(Trigonella

foenugraecum):

Most research studies

administer fenugreek

seed powders.

Glycemic

control

10 grams (g)/day to 100 g/day results in significant

reductions in fasting blood glucose (FBG) in Type 2

Diabetes Mellitus (T2DM) (48) and Type 1 Diabetes

Mellitus (T1DM) patients (49).

May also reduce FBG and hemoglobin A1C

(HbA1C) when taken with a sulfonylurea (50).

No impact on postprandial glucose values compared

to placebo conditions (51).

Study designs and dosages have been

inconsistent. The effectiveness of

fenugreek supplementation in glycemic

control is still questionable. Further,

fenugreek may interact with

anticoagulants and MAOIs.

American Ginseng

(Panax

quinquefolius):

Dried roots of

ginseng plants are

used in herbal

supplements.

Glycemic

control, insulin

secretion

3g dosage reduced FBG and HbA1C (52) and

postprandial glucose in T2DM patients (53). Higher

dosages demonstrated no further benefit (53).

In vitro studies indicate that American ginseng

stimulates insulin production and reduces pancreatic

beta cell apoptosis (15), though human studies

demonstrate no change in fasting insulin values with

American ginseng supplementation (52).

Research results are limited by small

sample sizes (n=9-24), though preliminary

evidence suggests that American ginseng

may play a role in glucose metabolism.

Delayed onset of hypoglycemia may be a

side effect of American ginseng, so blood

glucose should be monitored closely.

American ginseng may interact with

warfarin, MAOIs, estrogens, nifedipine,

and loop diuretics..

Cinnamon

(Cinnamomum

aromaticum):

A common spice used

in culturally diverse

cuisines, cinnamon is

typically

administered as an

encapsulated powder.

Glycemic

control

1-6 g/daily intake of cinnamon capsules significantly

reduced FBG (54), while 1 g/daily reduced HbA1C

by 0.83% (55) in T2DM patients.

Other research has shown no effect of cinnamon

supplementation on HbA1C, FBG, or insulin

sensitivity in T2DM patients (56-57).

A Cochrane systematic review and meta-analysis,

which included a total of 577 participants, found no

overall benefit of cinnamon supplementation on

diabetes endpoints (58).

Collectively, it appears that cinnamon

likely has no substantial effect on

glycemic control. In addition, cinnamon is

high in coumarin, which may cause

hepatotoxicity in patients with liver

disease.

Page 207: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

206

Prickly Pear Cactus

(Opuntia

streptocantha or

Opuntia ficus

indica):

A common foodstuff

in Arizona and

Mexico. Prickly pear

is typically consumed

as a dehydrated

extract or by broiling

the stems of the

young plant.

Glycemic

control

Preliminary trials indicate that the stems, but no other

part of the plant, may have hypoglycemic effects (59-

61).

All results are limited by study sample

size (n=8-32), and the only studies of

prickly pear and hypoglycemia in humans

were performed over two decades ago.

Prickly pear is likely safe when consumed

orally as a food.

Oat Bran:

High in fiber, and

commonly used for

treating

hypercholesterolemia.

Usually consumed as

oat bran flour.

Glycemic

control

(postprandial

and 24-hour)

Studies in T2DM patients indicate that long term and

acute oat bran flour consumption reduces

postprandial glucose (18,62-63).

Oat bran is not approved by the German

Commission E for diabetes treatment,

though it has Generally Recognized as

Safe (GRAS) status in the United States.

Chromium:

Essential in

carbohydrate and

lipid metabolism.

Brewer’s Yeast is

commonly used as a

chromium

supplement.

Glycemic

control

40 to 1000 micrograms (μg) daily supplementation

results in reductions in HbA1C, FBG, and

postprandial glucose in T2DM patients (64,20,65).

T2DM patients with well-controlled diabetes due to

oral hypoglycemic agents do not benefit from 400

μg/day chromium supplementation (66).

Studies examining effects of chromium on

glucose control have been somewhat

limited by sample size (n=3-180), so

conclusive results are difficult to draw.

Chromium supplements have not provided

added benefit when glucose control is

already well controlled with oral

hypoglycemic agents. Use of chromium

supplements (up to 1000 μg/day) have

been found to be safe when used short

term (up to 6 months of supplementation).

Page 208: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

207

Magnesium (Mg2+

):

Essential cofactor in

enzymes/proteins

involved in glucose

metabolism,

including the insulin

receptor. Typically

administered as

MgCl2 liquid

solution.

Insulin

sensitivity,

glycemic

control

A meta-analysis including over 536,000 prospective

cohort study participants suggests an inverse

relationship between risk of T2DM development and

Mg2+

intake (67). A dose-response analysis resulted

in a 14% reduction in risk of developing T2DM with

100 milligrams (mg)/day incremental increases in

Mg2+ intake (67).

Some studies suggest that Mg2+

supplementation can

reduce FBG and improve insulin sensitivity (16,68),

whereas others show no effect (69-70).

Higher Mg2+

intake may reduce risk of

developing T2DM, though study sample

size limits the ability to draw conclusions

regarding glycemic control once diabetes

has developed. However, Mg2+

supplementation is likely safe at doses

below the tolerable upper limit of 350

mg/day.

Selenium:

An essential cofactor

in glutathione

peroxidases.

Typically

administered as

selenized yeast.

Increased risk

of developing

T2DM

Epidemiological evidence suggests that higher serum

selenium values are associated with an increased risk

of T2DM (71).

Results from a large clinical trial suggest indicate an

increased risk of developing T2DM in those

randomized to consume 200 μg selenium/day for 7.7

years compared to those in the placebo group (72).

Unless advised by an MD or an RD,

supplements providing more than the DRI

of 55 μg of selenium should be avoided.

Alpha-Lipoic Acid

(ALA):

A cofactor of many

insulin sensitizing

and glucose

metabolism enzymes.

Administered as a

capsule.

Insulin

senstivity,

peripheral

neuropathy

300 to 1800 mg of daily ALA supplementation for

four weeks improves insulin sensitivity in T2DM

patients (73-74,14).

Improvements in symptoms of peripheral neuropathy

including burning, pain, and numbness of the feet and

legs is observed after 3 weeks of 600 mg/daily ALA

supplementation (75-76).

600 mg/daily supplementation is safe and reduces

progression of neuropathy in diabetic patients (77).

Evidence suggests that ALA may improve

insulin sensitivity and reduce progression

of peripheral neuropathy, long and short

term. Patients wit T2DM should consult a

medical professional regarding use of this

product. However, ALA is expensive, and

Vitamin E produces similar results as an

antioxidant at reduced cost.

Coenzyme Q10

(CoQ):

Increased risk

of developing

T2DM

Some studies show reductions in HbA1C in

individuals with T2DM who consume 200 mg CoQ

for 12 weeks (78-79).

Study providing 200 mg CoQ for 12 weeks found no

effect on HbA1C or any other diabetes-associated

outcomes (80).

Few studies have tested the role of CoQ in

parameters associated with diabetes

outcomes, though it is used in patients

with neurological disorders, heart failure,

hypertension, and in those taking statins.

There is insufficient data to make a

recommendation for the supplementation

of CoQ in diabetics seeking HbA1C

reduction.

Page 209: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

208

References (for Appendix F)

1. Alves R and Rosa IML. Biodiversity, traditional medicine and public health: where

do they meet? J Ethnobiol Ethnomedicine. 2007;3:14 Published online 2007 March 21.

doi: 10.1186/1746-4269-3-14 2. Wachtel-Galor and Benzie IF. Herbal Medicine (ch 1): An Introduction to its history,

usage, regulation, current trends, and research needs. IN Herbal Medicine:

Biomolecular and Clinical Aspects, 2nd

ed. Benzie IFF and Wachtel-Galor S, editors,

CRC Press, Boca Raton (FL), 2011.

3. Rock CL. Multivitamin-multimineral supplements: who uses them? Am J Clin Nutr.

2007; 85(1):277S-279S.

4. Bailey RL, Gahche JJ, Lentino CV, Dwyer JT, Engel JS, Thomas PR, et al. Dietary

supplement use in the United States, 2003-2006. J Nutr. 2011;141(2):261-266.

5. Barnes PM, Bloom B, Nahin RL. Complementary and alternative medicine use

among adults and children: United States, 2007. Natl Health Stat Report 2008;10:1–

23.

6. van Tonder E, Herselman MG, Visser J. The prevalence of dietary-related

complementary and alternative therapies and their perceived usefulness among cancer

patients. J Human Nutr Diet. 2009;22(6);528-535.

7. Bardia A, Greeno E, Bauer BA. Dietary supplement usage by patients with cancer

undergoing chemotherapy: does prognosis or cancer symptoms predict usage? J

Support Oncol. 2007;5(4):195-198.

8. Miller MF, Bellizzi KM, Sufian M, Ambs AH, Goldstein MS, Ballard-Barbash R.

Dietary supplement use in individuals living with cancer and other chronic

conditions: a population-based study. J Am Diet Assoc. 2008;108(3):483-494.

9. Richardson MA. Biopharmacologic and herbal therapies for cancer: research update

from NCCAM. J Nutr. 2001;131(11 Suppl):3037S-3040S.

10. Ferrucci LM, McCorkle R, Smith T, Stein KD, Cartmel B. Factors related to the use

of dietary supplements by cancer survivors. J Altern Complement Med.

2009;15(6):673-680.

11. Nahin RL, Byrd-Clark D, Stussman BJ, Kalyanaraman N. Disease severity is

associated with the use of complementary medicine to treat or manage type-2

diabetes: data from the 2002 and 2007 National Health Interview Study. BMC

Complement Altern Med. 2012;12:193. doi: 10.1186/1472-6882-12-193

12. Odegard PS, Janci MM, Foeppel MP, Beach JR, Trence DL. Prevalence and

correlates of dietary supplement use in individuals with diabetes mellitus at an

academic diabetes care clinic. Diabetes Educ. 2011;37(3):419-425.

13. Menon VP and Sudheer AR. Antioxidant and anti-inflammatory properties of

curcumin. Adv Exp Med Biol. 2007;595:105-125.

14. Ansar H, Mazloom Z, Kazemi F, Hejazi N. Effect of alpha-lipoic acid on blood

glucose, insulin resistance and glutathione peroxidase of type 2 diabetic patients.

Saudi Medical Journal, 2011;32(6):584-588.

Page 210: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

209

15. Luo JZ and Luo L. American ginseng stimulates insulin production and prevents

apoptosis through regulation of uncoupling protein-2 in cultured beta cells. Evidence-

based complementary and alternative medicine: eCAM, 2006;3(3):365-372.

16. Rodriguez-Moran M and Guerrero-Romero F. Oral magnesium supplementation

improves insulin sensitivity and metabolic control in type 2 diabetic subjects: a

randomized double-blind controlled trial. Diabetes Care. 2003;26(4):1147-1152.

17. Sharma S, Agrawal RP, Choudhary N, Jain S, Goyal S, Agarwal V. Beneficial effect

of chromium supplementation on glucose, HbA1C and lipid variables in individuals

with newly onset type-2 diabetes. J Trace Elem Med Biol. 2011;25(3):149-153.

18. Tapola N, Karvonen H, Niskanen L, Mikola M, Sarkkinen E. Glycemic responses of

oat bran products in type 2 diabetic patients. Nutr Met Cardiovasc Dis. 2005;15(4):

255-261.

19. Wang H-C, Pao J, Lin S-Y, Sheen L-Y. Molecular mechanisms of garlic-derived allyl

sulfides in the inhibition of skin cancer progression. Ann NY Acad Sci.

2012;1271(1):44-52.

20. Talalay P and Talalay P. The importance of using scientific principles in the

development of medicinal agents from plants. Acad Med. 2001;76:238-247.

21. Barkan ID.Industry invites regulation: the passage of the Pure Food and Drug Act of

1906. Am J Public Health.1985;75:18-26.

22. The new federal laws relating to foods, drugs, and cosmetics. Am J Public Health and

the Nation’s Health. 1938;28(9):1114-1116.

23. Greene JA and Podolsky SH. Reform, regulation, and pharmaceuticals – The

Kefauver-Harris amendments at 50. N Engl J Med. 2012;36:1481-1483.

24. S.784 Dietary Supplement Health and Education Act of 1994. Public Law 103–417,

1994.

25. Federal Register volume 77, number 57 (Friday, March 23, 2012). From the Federal

Register Online via the Government Printing Office (www.gpo.gov).

26. Bronstein AC, Spyker DA, Cantilena LR Jr, et al. 2009 Annual Report of the

American Association of Poison Control Centers’ National Poison Data System

(NPDS): 27th Annual Report. Clinical Toxicology 2010; 48:979–1178.

27. Kaye AD, Clarke C, Sabar R, Vig S, Dhawan KP, Hofbauer R. Herbal medications:

current trends in anesthesiology practice-a hospital survey. J Clin Anesth.

2000;12:468-471.

28. Hasler CM, Brown AC, Burns JA. Position of the American Dietetic Association:

Functional Foods. J Am Diet Assoc. 2009;109:735-746.

29. Mahady GB, Dog TL, Barrett ML, Chaavez ML, Gardiner P, Ko R. United States

Pharmacopeia review of the black cohosh case reports of hepatotoxicity Menopause.

2008;15(4 Pt 1):628-638.

30. Leach MJ, Moore V. Black cohosh (Cimicifuga spp.) for menopausal symptoms.

Cochrane Database Syst Rev. 2012 Sep 12;9:CD007244.

31. Oktem M, Eroglu D, Karahan HB, Taskintuna N, Kuscu E, Zeynelogluy HB. Black

cohosh and fluoxetine in the treatment of postmenopausal symptoms: a prospective,

randomized trial. Adv Ther. 2007;24(2):448-461.

32. Bundesanzeiger (Cologne, Germany): January 5, 1989

Page 211: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

210

33. Li J, Gödecke T, Chen SN, Imai A, Lankin DC, Farnsworth NR, et al. In vitro

metabolic interactions between black cohosh (Cimifuga racemosa) and tamoxifen via

inhibition of cytochromes P450 2D6 and 3A4. Xenobiotica.2011;41(12):1021-1030.

34. Rockwell S, Liu Y, Higgins SA. Alteration of the effects of cancer therapy agents on

breast cancer cells by the herbal medicine black cohosh. Breast Cancer Res Treat

2005;90(3):233-239.

35. Li H, Li HQ, Wang Y, et al. An intervention study to prevent gastric cancer by micro-

selenium and large dose of allitridum. Chinese Medical Journal (English).

2004;117(8):1155-1160.

36. Dirsch VM, Gerbes AL, Vollmar AM. Ajoene, a compound of garlic, induces

apoptosis in human promyeloleukemic cells, accompanied by generation of reaction

oxygen species and activation of nuclear factor kB. Mol Pharmacol.1998;53(3):402-

407.

37. WHO monographs on medicinal plants commonly used in Newly Independent States

(NIS). Bulbus Allii Sativi. p 9-28.

http://www.who.int/medicines/areas/traditional/monograph_eng.pdf Accessed

January 12, 2013.

38. Ryan JL, Heckler CE, Roscoe JA, Dakhil SR, Kirshner J, Flynn PJ. Ginger (zingiber

officinale) reduces acute chemotherapy-induced nausea: A URCC CCOP study of

576 patients. Support Care Cancer. 2012;20(7):1479-1489.

39. Zick SM, Ruffin MT, Lee J, Normolle DP, Siden R, Alrawi S, et al. Phase II trial of

encapsulated ginger as a treatment for chemotherapy-induced nausea and vomiting.

Support Care Cancer. 2009;17(5):563-572.

40. Biggs ML, Sorkin BC, Nahin RL, Kuller LH, Fitzpatrick AL. Ginkgo biloba and risk

of cancer: secondary analysis of the ginkgo evaluation of memory (GEM) study.

Pharmacoepidemiol Drug Saf. 2010;19(7):694-698.

41. Zhang Y, Chen AY, Chen C, Yao Q. Ginkgo biloba extract kaempferol inhibits cell

proliferation and induces apoptosis in pancreatic cancer cells. J Surg Res.

2008;148(1):17-23.

42. Satia JA, Littman A, Slatore CG, Galanko JA, White E. Associations of herbal and

specialty supplements with lung and colorectal cancer risk in the VITamins And

Lifestyle(VITAL) study. Cancer Epidemiol Biomarkers Prev. 2009;18(5):1419-1428.

43. Wessels JT, Busse AC, Rave-Fränk M, Zänker S, Hermann R, Grabbe E, et al.

Photosensitizing and radiosensitizing effects of hypericin on human renal carcinoma

cells in vitro. Photochem Photobiol. 2008;84(1):228-35.

44. Subramaniam D, Ponnurangam S, Ramamoorthy P, Standing D, Battafarano RJ,

Anant S, et al. Curcumin induces cell death in esophageal cancer cells through

modulating notch signaling. PLoS One. 2012; 7(2): e30590. Published online 2012

February 17. doi: 10.1371/journal.pone.0030590.

45. Sundram V, Chauham SC, Ebeling M, Jaggi M. Curcumin attenuates B-catenin

signaling in prostate cancer cells through activation of protein kinase D1. PLoS One.

2012; 7(4): e35368. Published online 2012 April 16.

doi: 10.1371/journal.pone.0035368.

46. Ye F, Zhang G-H, Guan B-X, Xu X-C. Suppression of esophageal cancer cell growth

Page 212: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

211

using curcumin, epigallocatechin-3-gallate and lovastatin. World J Gastroenterol.

2012;18(2):126-135.

47. Bayet-Robert M, Kwiatkowski F, Leheurteru M, Gachon F, Planchat E, Abrial C, et

al. Phase I dose escalation trial of docetaxel aplus curcumin in patients with advanced

and metastatic breast cancer. Cancer Biology & Therapy. 2010;9(1):8-14.

48. Kassaian N, Azadbakht L, Forghani B, Amini M. Effect of fenugreek seeds on blood

glucose and lipid profiles in type 2 diabetic patients. Intl J Vit Nutr Res.

2009;79(1):34-39.

49. Sharma RD, Raghuram TC, Rao NS. Effect of fenugreek seeds on blood glucose and

serum lipids in type 1 diabetes. Eur J Clin Nutr. 1990;44(4):301-306.

50. Lu FR, Shen L, Qin Y, Gao L, Li H, Dai Y. Clinical observation on trigonella

foenum-graecum L. total saponins in combination with sulfonylureas in the treatment

of type 2 diabetes mellitus. Chinese J Integrative Med. 2008;14(1):56-60.

51. Losso JN, Holliday DL, Finley JW, Martin RJ, Rood JC, Yu Y, et al. Fenugreek

bread: a treatment for diabetes mellitus. J Medicinal Food. 2009;12(5):1046-1049.

52. Vuksan V, Sievenpiper JL, Xu Z, Wong EY, Jenkins AL, Beljan-Zdravkovic U, et al.

Konjac-Mannan and American ginseng: emerging alternative therapies for type 2

diabetes mellitus. J Am Coll Nutr. 2001;20(5Suppl):370S-380S; discussion 381S-

383S.

53. Vuksan V, Stavro MP, Sievenpiper JL, Beljan-Zdravkovic U, Leiter LA, Josse RG.

Similar postprandial glycemic reductions with escalation of dose and administration

time of American ginseng in type 2 diabetes. Diabetes Care. 2000;23(9):1221-1226.

54. Khan A, Safdar M, Ali Khan MM, Khattak KN, Anderson RA. Cinnamon improves

glucose and lipids of people with type 2 diabetes. Diabetes Care. 2003;26(12):3215-

3218.

55. Crawford P. Effectiveness of cinnamon for lowering hemoglobin A1C in patients

with type 2 diabetes: a randomized, controlled trial. J Am Board Fam Med.

2009;22(5):507-512.

56. Baker WL, Gutierrez-Williams G, White CM, Kluger J, Coleman CI. Effect of

cinnamon on glucose control and lipid parameters. Diabetes Care. 2008;31(1):41-43.

57. Vanschoonbeek K, Thomassen BJ, Senden JM, Wodzig WK, van Loon LJ. Cinnamon

supplementation does not improve glycemic control in postmenopausal type 2

diabetes patients. J Nutr. 2006;136(4):977-980.

58. Leach MJ and Kumar S. Cinnamon for diabetes mellitus. Cochrane database of

systematic reviews. 2012. 9:p.CD007170.

59. Frati-Munari AC, Gordillo BE, Altamirano P, Ariza CR. Hypoglycemic effect of

Opuntia streptacantha Lemaire in NIDDM. Diabetes Care. 1988;11(1):63-66.

60. Frati-Munari AC, et al. Hypoglycemic action of different doses of nopal (Opuntia

streptacantha Lemaire) in patients with type II diabetes mellitus. Arch Invest Med

(Mex). 1989;20(2):197-201.

61. Frati Munari AC, Lastra VO, Andraca CRA. Evaluation of nopal capsules in diabetes

mellitus. Gaceta Medica de Mexico. 1992;128(4):431-436.

62. Pick ME, Hawrysh ZJ, Gee MI, Toth E, Garg ML, Hardin RT. Oat bran concentrate

bread products improve long-term control of diabetes: a pilot study. J Am Diet Assoc.

Page 213: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

212

1996;96(12):1254-1261.

63. Chandalia M, Garg A, Lutjohann D, von Bergmann K, Grundy SM, Brinkley LJ.

Beneficial effects of high dietary fiber intake in patients with type 2 diabetes mellitus.

New Eng J Med. 2000;342(19):1392-1398.

64. Anderson RA, Cheng N, Bryden NA, Polansky MM, Cheng N, Chi J, et al. Elevated

intakes of supplemental chromium improve glucose and insulin variables in

individuals with type 2 diabetes. Diabetes. 1997;46(11):1786-1791.

65. Racek J, Trefil L, Rajdo D, Mudrova V, Hunter D, Senft V. Influence of chromium-

enriched yeast on blood glucose and insulin variables, blood lipids, and markers of

oxidative stress in subjects with type 2 diabetes mellitus. Biol Trace Elem Res.

2006;109(3):215-230.

66. Kleefstra N, Houweling ST, Bakker SJ, Verhoeven S, Gans RO, Meyboom-de Jong

B, et al. Chromium treatment has no effect in patients with type 2 diabetes in a

Western population: a randomized, double-blind, placebo-controlled trial. Diabetes

Care. 2007;30(5):1092-1096.

67. Dong JY, Xun P, He K, Qin LQ. Magnesium intake and risk of type2 diabetes: meta-

analysis of prospective cohort studies. Diabetes Care. 2011;34(9):2116-2122.

68. Yokota K, Kato M, Lister F, Ii H, Hayakawa R, Kikuta T. Clinical efficacy of

magnesium supplementation in patients with type 2 diabetes. J Am Coll Nutr.

2004;23(5):506S-509S.

69. De Valk HW, Verkaaik R, van Rijn HJ, Geerdink RA, Struyvenberg A. Oral

magnesium supplementation in insulin-requiring type 2 diabetic patients. Diabet Med.

1998;15(6):503-507.

70. Gullestad L, Jacobsen T, Dolva LO. Effect of magnesium treatment on glycemic

control and metabolic parameters in NIDDM patients. Diabetes Care.

1994;17(5):460-461.

71. Bleys J, Navas-Acien A, Guallar E. Serum selenium and diabetes in U.S. adults.

Diabetes Care. 2007;30(4):829-834.

72. Stranges S, Marshall JR, Natarajan R, Donahue RP, Trevisan M, Combs GF, et al.

Effects of long-term selenium supplementation on the incidence of type 2 diabetes: a

randomized trial. Ann Intern Med. 2007;147(4):217-223.

73. Jacob S, Ruus P, Hermann R, Tritschler HJ, Maerker E, Renn W. Oral administration

of RAC-alpha-lipoic acid modulates insulin sensitivity in patients with type-2

diabetes mellitus: a placebo-controlled pilot trial. Free Rad Biol Med. 1999;27(3-

34):309-314.

74. Konrad T, Vicini P, Kusterer K, Hoflich A, Assadkhani A, Bohles HJ. Alpha-Lipoic

acid treatment decreases serum lactate and pyruvate concentrations and improves

glucose effectiveness in lean and obese patients with type 2 diabetes. Diabetes Care.

1999;22(2):280-287.

75. Ziegler D, Hanefeld M, Ruhnau KJ, Hasche H, Lobisch M, Schutte K. Treatment of

symptomatic diabetic polyneuropathy with the antioxidant alpha-lipoic acid: a 7-

month multicenter randomized controlled trial (ALADIN III Study). ALADIN III

Study Group. Alpha-Lipoic Acid in Diabetic Neuropathy. Diabetes Care.

1999;22(8):1296-1301.

Page 214: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

213

76. Ruhnau KJ, Meissner HP, Finn JR, Reljanovic M, Lobisch M, Schutte. Effects of 3-

week oral treatment with the antioxidant thioctic acid (alpha-lipoic acid) in

symptomatic diabetic polyneuropathy. Diab Med. 1999;16(12):1040-1043.

77. Ziegler D, Low PA, Litchy WJ, Boulton AJ, Vinik AI, Freeman R. Efficacy and

safety of antioxidant treatment with alpha-lipoic acid over 4 years in diabetic

polyneuropathy: the NATHAN 1 trial. Diabetes Care. 2011;34(9):2054-2060.

78. Hodgson JM, Watt GF, Playford DA, Burke V, Croft KD. Coenzyme Q10 improves

blood pressure and glycaemic control: a controlled trial in subjects with type 2

diabetes. Eur J Clin Nutr. 2002;5(11):1137-1142.

79. Playford DA, Watts GF, Croft KD, Burke V. Combined effect of coenzyme Q10 and

fenofibrate on forearm microcirculatory function in type2 diabetes. Atherosclerosis.

2003;168(1):169-179.

80. Eriksson JG, Forsen TJ, Mortensen SA, Rohde M. The effect of coenzyme Q10

administration on metabolic control in patients with type 2 diabetes mellitus.

Biofactors.1999;9(2-4):315-318.

Page 215: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

214

REFERENCES

1. Celermajer DS, Chow CK, Marijon E, et al. Cardiovascular disease in the

developing world: prevalences, patterns, and the potential of early disease

detection. Journal of the American College of Cardiology 2012, 60:1207-16.

2. Mathieu P, Lemieux I,Despres JP. Obesity, inflammation, and cardiovascular risk.

Clinical pharmacology and therapeutics 2010, 87:407-16.

3. Bakris GL. Current perspectives on hypertension and metabolic syndrome. J

Manag Care Pharm 2007, 13:S3-5.

4. Ogden CL, Carroll MD, Kit BK, et al. Prevalence of obesity in the United States,

2009-2010. NCHS data brief 2012, 1-8.

5. Pearson TA, Blair SN, Daniels SR, et al. AHA Guidelines for Primary Prevention

of Cardiovascular Disease and Stroke: 2002 Update: Consensus Panel Guide to

Comprehensive Risk Reduction for Adult Patients Without Coronary or Other

Atherosclerotic Vascular Diseases. American Heart Association Science Advisory

and Coordinating Committee. Circulation 2002, 106:388-91.

6. Meyers MR,Gokce N. Endothelial dysfunction in obesity: etiological role in

atherosclerosis. Current opinion in endocrinology, diabetes, and obesity 2007,

14:365-9.

7. Tchernof A,Despres JP. Pathophysiology of human visceral obesity: an update.

Physiological reviews 2013, 93:359-404.

8. Taylor AE, Ebrahim S, Ben-Shlomo Y, et al. Comparison of the associations of

body mass index and measures of central adiposity and fat mass with coronary

heart disease, diabetes, and all-cause mortality: a study using data from 4 UK

cohorts. The American journal of clinical nutrition 2010, 91:547-56.

9. Ghandehari H, Le V, Kamal-Bahl S, et al. Abdominal obesity and the spectrum of

global cardiometabolic risks in US adults. Int J Obes (Lond) 2009, 33:239-48.

10. Canoy D. Coronary heart disease and body fat distribution. Current

atherosclerosis reports 2010, 12:125-33.

11. Despres JP. Abdominal obesity and cardiovascular disease: is inflammation the

missing link? The Canadian journal of cardiology 2012, 28:642-52.

12. Trayhurn P,Wood IS. Adipokines: inflammation and the pleiotropic role of white

adipose tissue. The British journal of nutrition 2004, 92:347-55.

13. Pou KM, Massaro JM, Hoffmann U, et al. Visceral and subcutaneous adipose

tissue volumes are cross-sectionally related to markers of inflammation and

oxidative stress: the Framingham Heart Study. Circulation 2007, 116:1234-41.

14. Bosanska L, Michalsky D, Lacinova Z, et al. The influence of obesity and

different fat depots on adipose tissue gene expression and protein levels of cell

adhesion molecules. Physiological research / Academia Scientiarum

Bohemoslovaca 2010, 59:79-88.

15. Campia U, Tesauro M,Cardillo C. Human Obesity and Endothelium-Dependent

Responsiveness. Br J Pharmacol 2011,

16. Esposito K, Ciotola M,Giugliano D. Mediterranean diet, endothelial function and

vascular inflammatory markers. Public health nutrition 2006, 9:1073-6.

Page 216: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

215

17. Lumeng CN,Saltiel AR. Inflammatory links between obesity and metabolic

disease. J Clin Invest 2011, 121:2111-7.

18. Silver HJ, Dietrich MS,Niswender KD. Effects of grapefruit, grapefruit juice and

water preloads on energy balance, weight loss, body composition, and

cardiometabolic risk in free-living obese adults. Nutr Metab (Lond) 2011, 8:8.

19. Fujioka K, Greenway F, Sheard J, et al. The effects of grapefruit on weight and

insulin resistance: relationship to the metabolic syndrome. J Med Food 2006,

9:49-54.

20. Cho KW, Kim YO, Andrade JE, et al. Dietary naringenin increases hepatic

peroxisome proliferators-activated receptor alpha protein expression and

decreases plasma triglyceride and adiposity in rats. Eur J Nutr 2011, 50:81-8.

21. Orallo F, Camina M, Alvarez E, et al. Implication of cyclic nucleotide

phosphodiesterase inhibition in the vasorelaxant activity of the citrus-fruits

flavonoid (+/-)-naringenin. Planta Med 2005, 71:99-107.

22. Erlund I, Silaste ML, Alfthan G, et al. Plasma concentrations of the flavonoids

hesperetin, naringenin and quercetin in human subjects following their habitual

diets, and diets high or low in fruit and vegetables. Eur J Clin Nutr 2002, 56:891-

8.

23. Benavente-Garcia O,Castillo J. Update on uses and properties of citrus

flavonoids: new findings in anticancer, cardiovascular, and anti-inflammatory

activity. J Agric Food Chem 2008, 56:6185-205.

24. Joshipura KJ, Ascherio A, Manson JE, et al. Fruit and vegetable intake in relation

to risk of ischemic stroke. JAMA : the journal of the American Medical

Association 1999, 282:1233-9.

25. Dauchet L, Ferrieres J, Arveiler D, et al. Frequency of fruit and vegetable

consumption and coronary heart disease in France and Northern Ireland: the

PRIME study. The British journal of nutrition 2004, 92:963-72.

26. Mink PJ, Scrafford CG, Barraj LM, et al. Flavonoid intake and cardiovascular

disease mortality: a prospective study in postmenopausal women. The American

journal of clinical nutrition 2007, 85:895-909.

27. Zelman K. The Grapefruit Diet: What It is. Healthy Eating & Diet [cited 2011

May 27, 2011].

28. Cunningham E,Marcason W. Is it possible to burn calories by eating grapefruit or

vinegar? J Am Diet Assoc 2001, 101:1198.

29. Duyff, Food Folklore: Tales and Truths About What We Eat. 1 ed, ed. A.D.

Association1999: Wiley.

30. Flood-Obbagy JE,Rolls BJ. The effect of fruit in different forms on energy intake

and satiety at a meal. Appetite 2009, 52:416-22.

31. Goldwasser J, Cohen PY, Yang E, et al. Transcriptional regulation of human and

rat hepatic lipid metabolism by the grapefruit flavonoid naringenin: role of

PPARalpha, PPARgamma and LXRalpha. PloS one 2010, 5:e12399.

32. Wilcox LJ, Borradaile NM, de Dreu LE, et al. Secretion of hepatocyte apoB is

inhibited by the flavonoids, naringenin and hesperetin, via reduced activity and

expression of ACAT2 and MTP. Journal of lipid research 2001, 42:725-34.

Page 217: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

216

33. Assini JM, Mulvihill EE, Sutherland BG, et al. Naringenin prevents cholesterol-

induced systemic inflammation, metabolic dysregulation, and atherosclerosis in

Ldlr-/- mice. Journal of lipid research 2013, 54:711-24.

34. Gorinstein S, Caspi A, Libman I, et al. Red grapefruit positively influences serum

triglyceride level in patients suffering from coronary atherosclerosis: studies in

vitro and in humans. J Agric Food Chem 2006, 54:1887-92.

35. Demonty I, Lin Y, Zebregs YE, et al. The citrus flavonoids hesperidin and

naringin do not affect serum cholesterol in moderately hypercholesterolemic men

and women. J Nutr 2010, 140:1615-20.

36. Rizza S, Muniyappa R, Iantorno M, et al. Citrus Polyphenol Hesperidin

Stimulates Production of Nitric Oxide in Endothelial Cells while Improving

Endothelial Function and Reducing Inflammatory Markers in Patients with

Metabolic Syndrome. J Clin Endocrinol Metab 2011, 96:E782-92.

37. Reshef N, Hayari Y, Goren C, et al. Antihypertensive effect of sweetie fruit in

patients with stage I hypertension. Am J Hypertens 2005, 18:1360-3.

38. Morand C, Dubray C, Milenkovic D, et al. Hesperidin contributes to the vascular

protective effects of orange juice: a randomized crossover study in healthy

volunteers. Am J Clin Nutr 2011, 93:73-80.

39. Landberg R, Sun Q, Rimm EB, et al. Selected dietary flavonoids are associated

with markers of inflammation and endothelial dysfunction in U.S. women. J Nutr

2011, 141:618-25.

40. Dalgard C, Nielsen F, Morrow JD, et al. Supplementation with orange and

blackcurrant juice, but not vitamin E, improves inflammatory markers in patients

with peripheral arterial disease. The British journal of nutrition 2009, 101:263-9.

41. Wu CH, Lin JA, Hsieh WC, et al. Low-density-lipoprotein (LDL)-bound

flavonoids increase the resistance of LDL to oxidation and glycation under

pathophysiological concentrations of glucose in vitro. Journal of agricultural and

food chemistry 2009, 57:5058-64.

42. Burton-Freeman B. Postprandial metabolic events and fruit-derived phenolics: a

review of the science. The British journal of nutrition 2010, 104 Suppl 3:S1-14.

43. Alipour A, Elte JW, van Zaanen HC, et al. Novel aspects of postprandial lipemia

in relation to atherosclerosis. Atherosclerosis. Supplements 2008, 9:39-44.

44. Lacroix S, Rosiers CD, Tardif JC, et al. The role of oxidative stress in

postprandial endothelial dysfunction. Nutrition research reviews 2012, 25:288-

301.

45. O'Keefe JH,Bell DS. Postprandial hyperglycemia/hyperlipidemia (postprandial

dysmetabolism) is a cardiovascular risk factor. The American journal of

cardiology 2007, 100:899-904.

46. Bansal S, Buring JE, Rifai N, et al. Fasting compared with nonfasting

triglycerides and risk of cardiovascular events in women. JAMA : the journal of

the American Medical Association 2007, 298:309-16.

47. Cavalot F, Pagliarino A, Valle M, et al. Postprandial blood glucose predicts

cardiovascular events and all-cause mortality in type 2 diabetes in a 14-year

Page 218: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

217

follow-up: lessons from the San Luigi Gonzaga Diabetes Study. Diabetes care

2011, 34:2237-43.

48. Burton-Freeman B, Linares A, Hyson D, et al. Strawberry modulates LDL

oxidation and postprandial lipemia in response to high-fat meal in overweight

hyperlipidemic men and women. Journal of the American College of Nutrition

2010, 29:46-54.

49. Ghanim H, Sia CL, Upadhyay M, et al. Orange juice neutralizes the

proinflammatory effect of a high-fat, high-carbohydrate meal and prevents

endotoxin increase and Toll-like receptor expression. The American journal of

clinical nutrition 2010, 91:940-9.

50. Huebbe P, Giller K, de Pascual-Teresa S, et al. Effects of blackcurrant-based juice

on atherosclerosis-related biomarkers in cultured macrophages and in human

subjects after consumption of a high-energy meal. The British journal of nutrition

2012, 108:234-44.

51. Jimenez-Gomez Y, Lopez-Miranda J, Blanco-Colio LM, et al. Olive oil and

walnut breakfasts reduce the postprandial inflammatory response in mononuclear

cells compared with a butter breakfast in healthy men. Atherosclerosis 2009,

204:e70-6.

52. Snyder SM, Reber JD, Freeman BL, et al. Controlling for sugar and ascorbic acid,

a mixture of flavonoids matching navel oranges significantly increases human

postprandial serum antioxidant capacity. Nutrition research 2011, 31:519-26.

53. Pinsky PF, Miller A, Kramer BS, et al. Evidence of a healthy volunteer effect in

the prostate, lung, colorectal, and ovarian cancer screening trial. Am J Epidemiol

2007, 165:874-81.

54. Russell GV, Pierce CW,Nunley L. Financial implications of obesity. Orthop Clin

North Am 2011, 42:123-7, vii.

55. Selassie M,Sinha AC. The epidemiology and aetiology of obesity: a global

challenge. Best Pract Res Clin Anaesthesiol 2011, 25:1-9.

56. Heidenreich PA, Trogdon JG, Khavjou OA, et al. Forecasting the future of

cardiovascular disease in the United States: a policy statement from the American

Heart Association. Circulation 2011, 123:933-44.

57. Dallas C, Gerbi A, Tenca G, et al. Lipolytic effect of a polyphenolic citrus dry

extract of red orange, grapefruit, orange (SINETROL) in human body fat

adipocytes. Mechanism of action by inhibition of cAMP-phosphodiesterase

(PDE). Phytomedicine 2008, 15:783-92.

58. Kim SY, Kim HJ, Lee MK, et al. Naringin time-dependently lowers hepatic

cholesterol biosynthesis and plasma cholesterol in rats fed high-fat and high-

cholesterol diet. J Med Food 2006, 9:582-6.

59. Mulvihill EE, Allister EM, Sutherland BG, et al. Naringenin prevents

dyslipidemia, apolipoprotein B overproduction, and hyperinsulinemia in LDL

receptor-null mice with diet-induced insulin resistance. Diabetes 2009, 58:2198-

210.

Page 219: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

218

60. Jeon SM, Kim HK, Kim HJ, et al. Hypocholesterolemic and antioxidative effects

of naringenin and its two metabolites in high-cholesterol fed rats. Transl Res

2007, 149:15-21.

61. Wood N. Hepatolipidemic effects of naringenin in high cornstarch- versus high

coconut oil-fed rats. J Med Food 2004, 7:315-9.

62. Raper N PB, Ingwersen L, Steinfeldt L, Anand J. . An overview of USDA's

Dietary Intake Data System. Journal of Food Composition and Analysis 2004,

17:545-555.

63. Lohman TG RA, Martorell R, Anthropometric Standardization Reference

Manual, ed. S. G1988, Champaign, IL: Human Kinetics Books.

64. Dale RA, Jensen LH,Krantz MJ. Comparison of two point-of-care lipid analyzers

for use in global cardiovascular risk assessments. Ann Pharmacother 2008,

42:633-9.

65. Girennavar B JG, Jifon JL, Patil BS. Variation of bioactive furocoumarins and

flavonoids in different varieties of grapefruit and pummelo. European Food

Research Technology 2008, 226:1269-1275.

66. Wilburn AJ, King DS, Glisson J, et al. The natural treatment of hypertension. J

Clin Hypertens (Greenwich) 2004, 6:242-8.

67. Antoniades C, Shirodaria C, Warrick N, et al. 5-methyltetrahydrofolate rapidly

improves endothelial function and decreases superoxide production in human

vessels: effects on vascular tetrahydrobiopterin availability and endothelial nitric

oxide synthase coupling. Circulation 2006, 114:1193-201.

68. Jung UJ, Kim HJ, Lee JS, et al. Naringin supplementation lowers plasma lipids

and enhances erythrocyte antioxidant enzyme activities in hypercholesterolemic

subjects. Clin Nutr 2003, 22:561-8.

69. Anderson JW, Baird P, Davis RH, Jr., et al. Health benefits of dietary fiber. Nutr

Rev 2009, 67:188-205.

70. Lopez-Jimenez F,Cortes-Bergoderi M. Update: systemic diseases and the

cardiovascular system (i): obesity and the heart. Rev Esp Cardiol 2011, 64:140-9.

71. Lardizabal JA,Deedwania PC. Benefits of statin therapy and compliance in high

risk cardiovascular patients. Vasc Health Risk Manag 2010, 6:843-53.

72. Mukhtar RY,Reckless JP. Statin-induced myositis: a commonly encountered or

rare side effect? Curr Opin Lipidol 2005, 16:640-7.

73. Fichtlscherer S, Schmidt-Lucke C, Bojunga S, et al. Differential effects of short-

term lipid lowering with ezetimibe and statins on endothelial function in patients

with CAD: clinical evidence for 'pleiotropic' functions of statin therapy. Eur

Heart J 2006, 27:1182-90.

74. Dhaliwal SS,Welborn TA. Measurement error and ethnic comparisons of

measures of abdominal obesity. Prev Med 2009, 49:148-52.

75. Cassidy A, Rimm EB, O'Reilly EJ, et al. Dietary flavonoids and risk of stroke in

women. Stroke; a journal of cerebral circulation 2012, 43:946-51.

76. Liu L, Xu DM,Cheng YY. Distinct effects of naringenin and hesperetin on nitric

oxide production from endothelial cells. Journal of agricultural and food

chemistry 2008, 56:824-9.

Page 220: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

219

77. Dow CA, Going SB, Chow HH, et al. The effects of daily consumption of

grapefruit on body weight, lipids, and blood pressure in healthy, overweight

adults. Metabolism: clinical and experimental 2012, 61:1026-35.

78. Bailey DG,Dresser GK. Interactions between grapefruit juice and cardiovascular

drugs. American journal of cardiovascular drugs : drugs, devices, and other

interventions 2004, 4:281-97.

79. Grundy SM. Metabolic syndrome pandemic. Arteriosclerosis, thrombosis, and

vascular biology 2008, 28:629-36.

80. Blake GJ,Ridker PM. Inflammatory bio-markers and cardiovascular risk

prediction. Journal of internal medicine 2002, 252:283-94.

81. Blankenberg S, Rupprecht HJ, Bickel C, et al. Circulating cell adhesion molecules

and death in patients with coronary artery disease. Circulation 2001, 104:1336-

42.

82. Lotito SB,Frei B. Dietary flavonoids attenuate tumor necrosis factor alpha-

induced adhesion molecule expression in human aortic endothelial cells.

Structure-function relationships and activity after first pass metabolism. The

Journal of biological chemistry 2006, 281:37102-10.

83. Barona J, Aristizabal JC, Blesso CN, et al. Grape polyphenols reduce blood

pressure and increase flow-mediated vasodilation in men with metabolic

syndrome. The Journal of nutrition 2012, 142:1626-32.

84. Pai JK, Pischon T, Ma J, et al. Inflammatory markers and the risk of coronary

heart disease in men and women. The New England journal of medicine 2004,

351:2599-610.

85. Sanchez-Moreno C, Cano MP, de Ancos B, et al. High-pressurized orange juice

consumption affects plasma vitamin C, antioxidative status and inflammatory

markers in healthy humans. The Journal of nutrition 2003, 133:2204-9.

86. Minuz P, Fava C,Lechi A. Lipid peroxidation, isoprostanes and vascular damage.

Pharmacological reports : PR 2006, 58 Suppl:57-68.

87. Hutcheson R,Rocic P. The metabolic syndrome, oxidative stress, environment,

and cardiovascular disease: the great exploration. Experimental diabetes research

2012, 2012:271028.

88. Mathieu P, Pibarot P,Despres JP. Metabolic syndrome: the danger signal in

atherosclerosis. Vascular health and risk management 2006, 2:285-302.

89. Miyazaki K, Makino K, Iwadate E, et al. Anthocyanins from purple sweet potato

Ipomoea batatas cultivar Ayamurasaki suppress the development of

atherosclerotic lesions and both enhancements of oxidative stress and soluble

vascular cell adhesion molecule-1 in apolipoprotein E-deficient mice. Journal of

agricultural and food chemistry 2008, 56:11485-92.

90. Margioris AN. Fatty acids and postprandial inflammation. Current opinion in

clinical nutrition and metabolic care 2009, 12:129-37.

91. Jackson KG, Walden CM, Murray P, et al. A sequential two meal challenge

reveals abnormalities in postprandial TAG but not glucose in men with increasing

numbers of metabolic syndrome components. Atherosclerosis 2012, 220:237-43.

Page 221: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

220

92. O'Keefe JH, Gheewala NM,O'Keefe JO. Dietary strategies for improving post-

prandial glucose, lipids, inflammation, and cardiovascular health. Journal of the

American College of Cardiology 2008, 51:249-55.

93. Burdge GC,Calder PC. Plasma cytokine response during the postprandial period:

a potential causal process in vascular disease? The British journal of nutrition

2005, 93:3-9.

94. Klop B, Proctor SD, Mamo JC, et al. Understanding postprandial inflammation

and its relationship to lifestyle behaviour and metabolic diseases. International

journal of vascular medicine 2012, 2012:947417.

95. Rudolph TK, Ruempler K, Schwedhelm E, et al. Acute effects of various fast-

food meals on vascular function and cardiovascular disease risk markers: the

Hamburg Burger Trial. The American journal of clinical nutrition 2007, 86:334-

40.

96. Rankin JW, Andreae MC, Oliver Chen CY, et al. Effect of raisin consumption on

oxidative stress and inflammation in obesity. Diabetes, obesity & metabolism

2008, 10:1086-96.

97. Shen W, Xu Y,Lu YH. Inhibitory effects of Citrus flavonoids on starch digestion

and antihyperglycemic effects in HepG2 cells. Journal of agricultural and food

chemistry 2012, 60:9609-19.

98. Daher CF, Abou-Khalil J,Baroody GM. Effect of acute and chronic grapefruit,

orange, and pineapple juice intake on blood lipid profile in normolipidemic rat.

Medical science monitor : international medical journal of experimental and

clinical research 2005, 11:BR465-72.

99. Pruessner JC, Kirschbaum C, Meinlschmid G, et al. Two formulas for

computation of the area under the curve represent measures of total hormone

concentration versus time-dependent change. Psychoneuroendocrinology 2003,

28:916-31.

100. Yu Q, Gao F,Ma XL. Insulin says NO to cardiovascular disease. Cardiovasc Res

2011, 89:516-24.

101. Chiva-Blanch G, Urpi-Sarda M, Llorach R, et al. Differential effects of

polyphenols and alcohol of red wine on the expression of adhesion molecules and

inflammatory cytokines related to atherosclerosis: a randomized clinical trial. The

American journal of clinical nutrition 2012, 95:326-34.

102. Serafini M, Peluso I,Raguzzini A. Flavonoids as anti-inflammatory agents. The

Proceedings of the Nutrition Society 2010, 69:273-8.

103. Rattazzi M, Puato M, Faggin E, et al. C-reactive protein and interleukin-6 in

vascular disease: culprits or passive bystanders? Journal of hypertension 2003,

21:1787-803.

104. Burton-Freeman B, Talbot J, Park E, et al. Protective activity of processed tomato

products on postprandial oxidation and inflammation: a clinical trial in healthy

weight men and women. Molecular nutrition & food research 2012, 56:622-31.

105. Edirisinghe I, Banaszewski K, Cappozzo J, et al. Strawberry anthocyanin and its

association with postprandial inflammation and insulin. The British journal of

nutrition 2011, 106:913-22.

Page 222: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

221

106. Rexrode KM, Pradhan A, Manson JE, et al. Relationship of total and abdominal

adiposity with CRP and IL-6 in women. Annals of epidemiology 2003, 13:674-82.

107. Bastard JP, Maachi M, Lagathu C, et al. Recent advances in the relationship

between obesity, inflammation, and insulin resistance. European cytokine network

2006, 17:4-12.

108. Blankenberg S, Barbaux S,Tiret L. Adhesion molecules and atherosclerosis.

Atherosclerosis 2003, 170:191-203.

109. Derosa G, Cicero AF, Fogari E, et al. Effects of n-3 PUFAs on postprandial

variation of metalloproteinases, and inflammatory and insulin resistance

parameters in dyslipidemic patients: evaluation with euglycemic clamp and oral

fat load. Journal of clinical lipidology 2012, 6:553-64.

110. Pacheco YM, Bemudez B, Lopez S, et al. Minor compounds of olive oil have

postprandial anti-inflammatory effects. The British journal of nutrition 2007,

98:260-3.

111. Schoppen S, Perez-Granados AM, Navas-Carretero S, et al. Postprandial lipaemia

and endothelial adhesion molecules in pre- and postmenopausal Spanish women.

Nutricion hospitalaria : organo oficial de la Sociedad Espanola de Nutricion

Parenteral y Enteral 2010, 25:256-61.

112. Perez-Martinez P, Moreno-Conde M, Cruz-Teno C, et al. Dietary fat differentially

influences regulatory endothelial function during the postprandial state in patients

with metabolic syndrome: from the LIPGENE study. Atherosclerosis 2010,

209:533-8.

113. Deopurkar R, Ghanim H, Friedman J, et al. Differential effects of cream, glucose,

and orange juice on inflammation, endotoxin, and the expression of Toll-like

receptor-4 and suppressor of cytokine signaling-3. Diabetes care 2010, 33:991-7.

114. Andrade CM, Sa MF,Toloi MR. Effects of phytoestrogens derived from soy bean

on expression of adhesion molecules on HUVEC. Climacteric : the journal of the

International Menopause Society 2012, 15:186-94.

115. Benavente-Garcia O,Castillo J. Update on uses and properties of citrus

flavonoids: new findings in anticancer, cardiovascular, and anti-inflammatory

activity. Journal of agricultural and food chemistry 2008, 56:6185-205.

116. Wiswedel I, Hirsch D, Kropf S, et al. Flavanol-rich cocoa drink lowers plasma

F(2)-isoprostane concentrations in humans. Free radical biology & medicine

2004, 37:411-21.

117. Aronne LJ, Brown WV,Isoldi KK. Cardiovascular disease in obesity: A review of

related risk factors and risk-reduction strategies. Journal of clinical lipidology

2007, 1:575-82.

118. Raitakari M, Ilvonen T, Ahotupa M, et al. Weight reduction with very-low-caloric

diet and endothelial function in overweight adults: role of plasma glucose.

Arteriosclerosis, thrombosis, and vascular biology 2004, 24:124-8.

119. Go AS, Mozaffarian D, Roger VL, et al. Executive summary: heart disease and

stroke statistics--2013 update: a report from the American Heart Association.

Circulation 2013, 127:143-52.

Page 223: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

222

120. Craddick SR, Elmer PJ, Obarzanek E, et al. The DASH diet and blood pressure.

Current atherosclerosis reports 2003, 5:484-91.

121. Manduteanu I,Simionescu M. Inflammation in atherosclerosis: a cause or a result

of vascular disorders? Journal of cellular and molecular medicine 2012, 16:1978-

90.

122. Allister EM, Borradaile NM, Edwards JY, et al. Inhibition of microsomal

triglyceride transfer protein expression and apolipoprotein B100 secretion by the

citrus flavonoid naringenin and by insulin involves activation of the mitogen-

activated protein kinase pathway in hepatocytes. Diabetes 2005, 54:1676-83.

123. Sharma AK, Bharti S, Ojha S, et al. Up-regulation of PPARgamma, heat shock

protein-27 and -72 by naringin attenuates insulin resistance, beta-cell dysfunction,

hepatic steatosis and kidney damage in a rat model of type 2 diabetes. The British

journal of nutrition 2011, 106:1713-23.

124. Torronen R, Sarkkinen E, Niskanen T, et al. Postprandial glucose, insulin and

glucagon-like peptide 1 responses to sucrose ingested with berries in healthy

subjects. The British journal of nutrition 2012, 107:1445-51.

125. Wilson T, Singh AP, Vorsa N, et al. Human glycemic response and phenolic

content of unsweetened cranberry juice. Journal of medicinal food 2008, 11:46-

54.

126. Bryans JA, Judd PA,Ellis PR. The effect of consuming instant black tea on

postprandial plasma glucose and insulin concentrations in healthy humans.

Journal of the American College of Nutrition 2007, 26:471-7.

127. Campia U, Tesauro M,Cardillo C. Human obesity and endothelium-dependent

responsiveness. British journal of pharmacology 2012, 165:561-73.

128. Pasceri V, Willerson JT,Yeh ET. Direct proinflammatory effect of C-reactive

protein on human endothelial cells. Circulation 2000, 102:2165-8.

129. Pasceri V, Cheng JS, Willerson JT, et al. Modulation of C-reactive protein-

mediated monocyte chemoattractant protein-1 induction in human endothelial

cells by anti-atherosclerosis drugs. Circulation 2001, 103:2531-4.

130. Biswas P, Delfanti F, Bernasconi S, et al. Interleukin-6 induces monocyte

chemotactic protein-1 in peripheral blood mononuclear cells and in the U937 cell

line. Blood 1998, 91:258-65.

131. Bodet C, La VD, Epifano F, et al. Naringenin has anti-inflammatory properties in

macrophage and ex vivo human whole-blood models. Journal of periodontal

research 2008, 43:400-7.

132. Tsai SJ, Huang CS, Mong MC, et al. Anti-inflammatory and antifibrotic effects of

naringenin in diabetic mice. Journal of agricultural and food chemistry 2012,

60:514-21.

133. Hamalainen M, Nieminen R, Vuorela P, et al. Anti-inflammatory effects of

flavonoids: genistein, kaempferol, quercetin, and daidzein inhibit STAT-1 and

NF-kappaB activations, whereas flavone, isorhamnetin, naringenin, and

pelargonidin inhibit only NF-kappaB activation along with their inhibitory effect

on iNOS expression and NO production in activated macrophages. Mediators of

inflammation 2007, 2007:45673.

Page 224: THE ROLE OF GRAPEFRUIT CONSUMPTION IN CARDIOMETABOLIC HEALTH IN OVERWEIGHT

223

134. Nie YC, Wu H, Li PB, et al. Naringin attenuates EGF-induced MUC5AC

secretion in A549 cells by suppressing the cooperative activities of MAPKs-AP-1

and IKKs-IkappaB-NF-kappaB signaling pathways. European journal of

pharmacology 2012, 690:207-13.

135. Choe SC, Kim HS, Jeong TS, et al. Naringin has an antiatherogenic effect with

the inhibition of intercellular adhesion molecule-1 in hypercholesterolemic

rabbits. Journal of cardiovascular pharmacology 2001, 38:947-55.

136. Lee CH, Jeong TS, Choi YK, et al. Anti-atherogenic effect of citrus flavonoids,

naringin and naringenin, associated with hepatic ACAT and aortic VCAM-1 and

MCP-1 in high cholesterol-fed rabbits. Biochemical and biophysical research

communications 2001, 284:681-8.

137. Gopaul NK, Zacharowski K, Halliwell B, et al. Evaluation of the postprandial

effects of a fast-food meal on human plasma F(2)-isoprostane levels. Free radical

biology & medicine 2000, 28:806-14.

138. Collins T,Cybulsky MI. NF-kappaB: pivotal mediator or innocent bystander in

atherogenesis? The Journal of clinical investigation 2001, 107:255-64.

139. Brook RD, Kansal M, Bard RL, et al. Usefulness of low-density lipoprotein

particle size measurement in cardiovascular disease prevention. Clinical

cardiology 2005, 28:534-7.

140. Green DJ, Jones H, Thijssen D, et al. Flow-mediated dilation and cardiovascular

event prediction: does nitric oxide matter? Hypertension 2011, 57:363-9.