+ All Categories
Home > Documents > %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG...

%LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG...

Date post: 17-Apr-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
154
Dissertation for the degree of philosophiae doctor (PhD) at the University of Bergen Dissertation date: atherosclerosis
Transcript
Page 1: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Dissertation for the degree of philosophiae doctor (PhD)

at the University of Bergen

Dissertation date:

atherosclerosis

Page 2: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

© Copyright Rita Vik

The material in this publication is protected by copyright law.

Year: 2015

Title: Bioactive Proteins and Peptides Influence Lipid Metabolism and Inflammation in Relation to Atherosclerosis

Author: Rita Vik

Print: AIT OSLO AS / University of Bergen

Page 3: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

3

Scientific environment

This study was carried out at the Lipid Research Group, at the Department of Clinical

Science, University of Bergen from November 2011 until April 2014.

The work was funded by Nordforsk under the Nordic Centers of Excellence program

in Food, Nutrition and Health; Project (070010) “Mitohealth”, The Research Council

of Norway (grant no. 190287/110), The European Community’s Seventh Framework

Programme (FP7/2007-2013) AtheroRemo (grant no. 201668), The Board of

Nutrition Programmes – University of Bergen, Norilia AS and NFR project 212984.

Page 4: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

4

Acknowledgements

I would like to thank to my main supervisor Professor Rolf K. Berge for introducing

me to the field of fatty acid metabolism, mitochondrial function and atherosclerosis. I

am very thankful for your enthusiastic support, guidance and encouragement during

my PhD period.

A special thanks to my co-supervisor Dr. Bodil Bjørndal for helping me throughout

these years. Your intellectual guidance, contribution, cooperation and kindness have

been like an essential amino acid; indispensable!

I appreciate my second co-supervisor Dr. Ottar Nygård for his critical thoughts and

useful feedback on manuscript preparations and on my thesis.

I am so grateful to the laboratory technicians Kari Williams, Randi Sandvik, Svein

Krüger, Pavol Bohov, Liv Kristine Øysæd and Kari Helland Mortensen for your

skillful technical support and patience with me during these years. Thank you for

assisting me with my laboratory work and always being ready to help me in times of

trouble! I have appreciated your support and conversations during lunchtime.

In addition, I give my thanks to Dr. Trond Brattelid for teaching me aorta dissection.

Here I will also like to thank Eline M. Nævdal for teaching me animal care and

handling. Thanks to the rest of the staff at The Laboratory Animal facility.

A big thanks to our collaborators abroad; Cinzia Parolini, Marco Busnelli, Stefano

Manzini, Giulia S. Ganzetti, Federica Dellera, Cesare R. Sirtori and Giulia Chiesa at

the Department of Pharmacological and Biomolecular Sciences, Università degli

Studi di Milan, Italy, Veronika Tillander and Stefan E. H. Alexson at Department of

Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institutet,

Karolinska University Hospital, Sweden, and Terhi Vihervaara and Kim Ekroos at

Zora Biosciences Oy, Finland.

I own thanks to all the other co-authors for their interesting insights and contribution

to the papers.

Page 5: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

5

Thanks to my fellow PhD-students for creating a friendly atmosphere.

Finally, a warm thanks to my husband Christian. When I first met you, I had just

started my PhD fellowship and I remember my first words to you: I have just started

my PhD degree, so I’m not planning on having a life the next three years… Well, we

both know how that turned out! I am so grateful to you for the consolation,

encouragement and having faith in me. Thank you for finding the time to assist me

with the figures and for taking care of our son during the process of finalizing of my

dissertation. Also, I really appreciated you keeping the apartment neat and bringing

me coffee. I could not have done this without you!

Rita Vik

Bergen, April 2015

Page 6: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

6

Abstract

The prevalence of lifestyle diseases emerging from the metabolic syndrome is

increasing and atherothrombotic cardiovascular disease leading to atherosclerosis is

the primary cause of death in the Western world. The development of atherosclerosis

is linked to mitochondrial dysfunction, lipid metabolism and inflammation.

It has been increasingly clear that bioactive protein can affect lipid metabolism and

hydrolyzed peptides possess bioactive potential.

Our aim was to investigate bioactive protein and peptides from salmon (SPH) and

chicken (CP) and determine their ability to influence lipid metabolism, inflammatory

markers and plaque development in rodent models. Further, we wanted to elucidate if

peptides from SPH or protein from CP could ameliorate atherosclerotic development

and inflammation in an apolipoprotein (apo)-E knockout mice model.

In the first study we investigated three different fractions of SPH in C57BL/6 mice.

Two of the fractions displayed plasma and liver lipid-lowering effect linked to

reduced fatty acid synthesis. The opposite was shown in another fraction. Thus,

bioactive peptides with distinct properties could be isolated from salmon protein.

The fraction of SPH showing most lipid-lowering effect in the previous study was

evaluated in apoE-/- mice to investigate atherosclerotic development. After 12 weeks

we detected less atherosclerotic plaque area in aortic arch and sinus of mice fed SPH,

but there was no change in plague composition. Circulating levels of inflammatory

markers were reduced, whereas plasma TAG and cholesterol levels were unchanged.

This study showed that SPH attenuated atherosclerosis in apoE-/- mice both at

vascular and systemic levels, independent of plasma lipids.

CP was used to elucidate potential effects on lipid metabolism in Wistar rats. Rats fed

CP had lower body weight despite a high feed intake. CP displayed TAG and

cholesterol-lowering capabilities in plasma as well as in liver, in addition to

stimulated mitochondrial fatty acid oxidation. Fatty acid synthesis appeared to be

Page 7: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

7

reduced as hepatic enzyme activities and gene expression of lipogenesis was

decreased and plasma bile acids were increased. These results suggest a

hypotriglyceridemic property of CP and ability to reduced cholesterol levels, which

could be linked to increase in bile formation.

CP was further evaluated in apoE-/- mice. After 12 weeks of CP intervention, we

detected a small reduction in plasma TAG levels linked to increased energy

combustion from fat during inactive state. There was no change in plasma

cholesterol, TAG or fatty acid composition. En-face analysis of aorta revealed no

change in plaque development compared to controls, accompanied by unaltered

cytokine levels.

We concluded with the assumption that protein from different sources may possess

bioactive potential and hydrolyzing methods could liberate peptides with different

properties. SPH was also able to counteract atherosclerotic development in apoE-/-

mice by reducing plaque area and systemic cytokine levels despite unchanged plasma

lipid levels.

CP revealed promising effect on energy metabolism in rats, which could be related to

the amino acid profile of CP.

CP was not able to influence systemic or local inflammation, nor plaque area in apoE-

/- mice. Although CP displayed great influence on lipid metabolism in rats, it was not

capable of counteract the disrupted lipoprotein metabolism in the apoE-/- mouse

model.

Page 8: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

8

List of publications

Paper I Vik, R., Tillander, V., Skorve, J., Vihervaara, T., Ekroos, K., Alexson, S.E.H., Berge, R.K., and Bjørndal, B., Three differently generated salmon protein hydrolysates reveal opposite effects on hepatic lipid metabolism in mice fed a high-fat diet. Food Chemistry, 2015. 183: p. 101-110.

Paper II Parolini, C., Vik, R., Busnelli, M., Bjørndal, B., Holm, S., Brattelid, T., Manzini, S., Ganzetti G.S., Dellera, F., Halvorsen, B., Aukrust, P., Sirtori, C.R., Nordrehaug, J.E., Skorve, J., Berge R.K., Chiesa, G., A salmon protein hydrolysate exerts lipid-independent anti-atherosclerotic activity in ApoE-deficient mice. Plos One, 2014. 9(5): e97598.

Paper III Vik, R., Bjørndal, B., Bohov, P., Brattelid, T., Svardal, A., Nygård, O.K., Nordrehaug, J.E., Skorve, J., and Berge, R.K., Hypolipidemic effect of dietary water-soluble protein extract from chicken: impact on genes regulating hepatic lipid and bile acid metabolism. European Journal of Nutrition, 2014. 54(2): p. 193-204.

Paper IV Vik, R., Brattelid, T., Skorve, J., Nygård, O.K., Nordrehaug, J.E., Berge, R.K., and Bjørndal, B., A water-soluble extract of chicken reduced plasma triacylglycerols, but showed no anti-atherosclerotic activity in apoE-/- mice. (Accepted, In Press)

Page 9: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

9

Table of Contents

SCIENTIFIC ENVIRONMENT ......................................................................................................... 3

ACKNOWLEDGEMENTS ................................................................................................................. 4

ABSTRACT........................................................................................................................................... 6

LIST OF PUBLICATIONS ................................................................................................................. 8

TABLE OF CONTENTS ..................................................................................................................... 9

ABBREVIATIONS ............................................................................................................................. 12

LIST OF FIGURES ............................................................................................................................ 14

LIST OF TABLES .............................................................................................................................. 15

1. INTRODUCTION .................................................................................................................... 16

1.1 OBESITY AND CARDIOMETABOLIC SYNDROME ....................................................................... 16

1.2 OBESITY AND INFLAMMATION ............................................................................................... 18

1.3 ATHEROSCLEROSIS ................................................................................................................ 20

1.4 DIETARY PROTEINS ................................................................................................................ 23

1.4.1 Dietary proteins can affect lipid metabolism .............................................................. 24

1.4.2 Amino acid profile ....................................................................................................... 25

1.4.3 Amino acid ratios ........................................................................................................ 26

2. AIMS OF STUDY .................................................................................................................... 28

3. THEORETICAL BACKGROUND ........................................................................................ 29

3.1 LIPIDS .................................................................................................................................... 29

3.1.1 Fatty acids ................................................................................................................... 29

3.1.2 Glycerolipids ............................................................................................................... 29

3.1.3 Digestion, Absorption and Transport ......................................................................... 31

3.1.4 Cholesterol, cholesteryl esters and bile acids ............................................................. 33

3.1.5 Activation to acyl-CoA ................................................................................................ 38

Page 10: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

10

3.1.6 Mitochondrial β-oxidation ......................................................................................... 38

3.1.7 Peroxisomal β-oxidation ............................................................................................ 41

3.1.8 Lipogenesis ................................................................................................................. 41

3.1.9 Elongation and desaturation of fatty acids ................................................................ 42

3.1.10 Ceramides and sphingolipids ..................................................................................... 44

3.2 PROTEIN ................................................................................................................................ 44

3.2.1 Catabolism of protein and amino acids...................................................................... 45

3.2.2 Enzymatically breakdown of dietary proteins ............................................................ 45

3.2.3 Transamination and oxidative deamination of amino acids....................................... 46

3.2.4 Nitrogen excretion and pathways of amino acid degradation.................................... 47

3.2.5 Bioactive peptides ...................................................................................................... 47

3.3 TRANSCRIPTIONAL REGULATION OF LIPID AND AMINO ACID METABOLISM ............................ 48

3.3.1 Peroxisome proliferator-activated receptors ............................................................. 48

3.3.2 Sterol regulatory element-binding factors.................................................................. 49

4. EXPERIMENTAL MODELS ................................................................................................ 51

4.1 WISTAR RATS ........................................................................................................................ 51

4.2 C57BL/6J MICE ..................................................................................................................... 51

4.3 APOE-/- MICE ......................................................................................................................... 52

5. SUMMARY OF RESULTS .................................................................................................... 53

6. DISCUSSION ........................................................................................................................... 57

6.1 MARINE BIOACTIVE PEPTIDES................................................................................................ 57

6.1.1 SPH reduced body weight .......................................................................................... 58

6.1.2 TAG-reducing effect of SPH ....................................................................................... 59

6.1.3 Various protein hydrolysates may influence fatty acid composition .......................... 59

6.1.4 Elevated liver ceramide levels did not affect fatty acid catabolism ........................... 61

Page 11: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

11

6.1.5 SPH reduced atherosclerotic plaque area in apoE-/- mice .......................................... 62

6.1.6 Plaque stability was unaffected after SPH-treatment ................................................. 63

6.1.7 SPH decreased systemic inflammation ....................................................................... 63

6.2 CP AS A PROTEIN SOURCE ...................................................................................................... 65

6.2.1 CP reduced body weight and lipogenesis in rats ........................................................ 65

6.2.2 Elevated plasma level of bile acids after CP intervention .......................................... 66

6.2.3 Cellular toxicity could be reduced .............................................................................. 67

6.2.4 CP showed no anti-atherosclerotic effect in apoE-/- mice ........................................... 67

7. CONCLUSIONS ....................................................................................................................... 70

8. FUTURE PERSPECTIVES..................................................................................................... 72

9. REFERENCES ......................................................................................................................... 73

Page 12: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

12

Abbreviations

AADE Amino Acid Degrading Enzyme

ABCA1 Adenosine Triphosphate-Binding Cassette 1

ACAT Acyl-Coenzyme A Cholesterol Acyltransferase

ACC Acetyl-Coenzyme A Carboxylase

ACE Adenosine-Converting Enzyme

ACOX Acyl-Coenzyme A oxidase

ANOVA Analysis of Variance

APO Apolipoprotein

ASC Acyl-Coenzyme A Synthase

ATP Adenosine Triphosphate

CACT Carnitine Acyltransferase

CoA Coenzyme A

CP Chicken Protein

CPT Carnitine Palmitoyltransferase

CVD Cardiovascular Disease

CYP7A1 Cytochrome P450, family 7, subfamily A, polypeptide 1, or cholesterol

7 alpha-hydroxylase

DAG Diacylglycerol

DGAT Diacylglycerol Acyltransferase

DHA Docosahexaenoic Acid

DPA Docosapentaenoic Acid

EPA Eicosapentaenoic Acid

ER Endoplasmic Reticulum

FAD Flavin Adenine Dinucleotide

FADS Fatty Acid Desaturase

FAS Fatty Acid Synthase

FFA Free Fatty Acid

GPAT Glycero-3-Phospohate Acyltransferase

HDL High-Density Lipoprotein

Page 13: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

13

HMG-CoA 3-Hydroxy-3-Methylglutaryl-Coenzyme A

HNF-4α Hepatocyte Nuclear Factor 4 alpha

ICAM Intercellular Adhesion Molecule

IDL Intermediate-Density Lipoprotein

IL Interleukin

IRS1 Insulin Receptor Substrate 1

LDL Low-Density Lipoprotein

LDLr Low-Density Lipoprotein Receptor

LNAA Large Neutral Amino Acids

LPL Lipoprotein Lipase

MCP1 Monocyte chemoattractant protein 1

MUFA Mono-unsaturated Fatty Acid

NADH Nicotinamide Adenine Dinucleotide

NAFLD Non-Alcoholic Fatty Liver Disease

NEFA Non-Esterified Fatty Acid

NF-kB Nuclear Factor KappaB

NOS2 Nitric Oxide Synthase 2

PPAR Peroxisome Proliferator-Activated Receptor

PK Protein Kinase

PUFA Poly-unsaturated Fatty Acid

RER Respiratory Exchange Ratio

SCD1 Stearoyl-Coenzyme A desaturase 1

SFA Saturated Fatty Acid

SPH Salmon Protein Hydrolysate

SREBP Sterol Regulatory Element-Binding Protein

TAG Triacylglycerol

TCA Tricarboxylic Acid

VCAM Vascular Cell Adhesion Molecule

VSMC Vascular Smooth Muscle cell

VLDL Very Low-Density Lipoprotein

Page 14: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

14

List of Figures

Figure 1 - Metabolic syndrome ................................................................................ 18

Figure 2 - Development of Atherosclerosis .............................................................. 22

Figure 3 - Glycerolipid synthesis ............................................................................. 30

Figure 4 - Lipoprotein metabolism ........................................................................... 33

Figure 5 - Cholesterol synthesis .............................................................................. 34

Figure 6 - Enterohepatic circulation ......................................................................... 37

Figure 7 - Carnitine shuttle ...................................................................................... 39

Figure 8 - Mitochondrial vs. peroxisomal β-oxidation ............................................... 40

Figure 9 - Fatty acid synthesis ................................................................................ 42

Figure 10 - Elongation and desaturation of fatty acids ............................................. 43

Page 15: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

15

List of Tables

Table 1 - Essential and non-essential amino acids in humans .................................27

Page 16: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

16

1. INTRODUCTION

1.1 Obesity and cardiometabolic syndrome

The prevalence of overweight and obesity has increased dramatically worldwide and

according to the World Health Organization (WHO) 1.4 billion adults were obese in

2008, when children and adolescent under the age of 20 were omitted [1]. A number

of impairments are closely associated with the rise in overweight and obesity-related

illnesses like atherothrombotic cardiovascular disease (hereafter referred to as CVD),

described as disruption of atherosclerotic lesions, which are the leading cause of

death in the industrialized world. Risk factors for developing lifestyle diseases

attributed obesity are commonly known under the generic term; metabolic syndrome

[2]. The metabolic syndrome is a cluster of conditions occurring in one individual [3],

comprising abdominal obesity, dyslipidemia, hypertension and insulin resistance [4],

all predisposes for developing cardiovascular diseases [5]. An overview of the

metabolic syndrome is given in Figure 1.

Malfunction of lipid metabolism is an originator strongly associated with the criteria

of metabolic syndrome [6], in addition to neurodegenerative and age-related diseases,

like Alzheimer´s [7, 8]. Cardiometabolic syndrome, emerging from the metabolic

syndrome, is a configuration of maladaptive cardiovascular, renal, metabolic,

prothrombotic and inflammatory abnormalities, and defines a well-established cluster

of risk factor for premature CVD and stroke [9]. Since mitochondria is the main

organelle regulating energy metabolism [10-13], attention has been drawn to

mitochondrial dysfunction as being the basal cause of obesity-related conditions [14-

19]. Chronic inflammatory diseases like rheumatoid arthritis [20] and atherosclerosis

[21], are also linked to obesity, and investigators now appreciate that these

inflammatory states may arise from metabolic imbalance [22]. Although genetics are

involved in the development of both metabolic syndrome and cardiometabolic

syndrome, dietary behaviours and a modifiable lifestyle pattern are the main

contributors to metabolic-related illness.

Page 17: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

17

A likely explanation for the severe increase in overweight and obesity is simply

energy imbalance. The combination of excess energy intake and a sedentary lifestyle

is related to a developed society. Unlimited access to all sorts of food, in particular

highly processed foods used in the fast food industry, and requirement of

substantially less physical activity is undoubtedly the main causes of the growing

obese epidemic [23, 24]. Besides a large food consumption, the increasing economic

welfare among the Western population leads to great amounts of food being

discarded every year. Hydrolyzation processes of waste products yield useful

products, which can contribute to improve protein and food quality. Protein quality is

of particular importance with regard to protein malnutrition in third world countries

where access to food is limited. Protein hydrolyzed from natural sources are

considered more health-enhancing as nutraceuticals, in addition to a better safety

profile compared to synthetic manufactured supplements and pharmaceutics.

Page 18: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

18

Figure 1 - Metabolic syndrome Criteria for the metabolic syndrome are listed in the green box. Metabolic syndrome is diagnosed when 3 out of 5 of these risk factors occur in one individual. Metabolic syndrome is associated with chronic low-grade inflammation and mitochondrial dysfunction predisposing for developing CVD, diabetes and non-alcoholic liver disease. *Waist circumference. CVD, atherothrombotic cardiovascular disease; FFA, free fatty acid; HDL, high-density lipoprotein; TAG, triacylglycerol; VLDL, very low-density lipoprotein.

1.2 Obesity and inflammation

Obesity-induced inflammation is distinctive from classical inflammation

characterized by redness, swelling, itch, heat and pain, as it is asymptomatic. Obesity-

induced inflammation causes systemic reactions, in particular observed in the

vascular system, gastrointestinal tract or liver, thus invisible on the outside of the

body. Another striking different is that the behaviour of classic inflammation is

associated with increased metabolic rate, representing a rapid immune response to

injury, which neutralizes and resolves the inflammatory state, whereas in obesity-

induced inflammation abnormalities in metabolism is the primary cause, in particular

Page 19: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

19

excess energy intake. Adipocytes, or fat cells, are specialized cells involved in lipid

storage and metabolic signalling. In response to nutrient overload, the adipocytes are

also responsible for initiating and sustaining the inflammatory program [25]. Besides

being a storage of fatty acids, the adipose tissue also serves as an endocrine organ

secreting hormones, called adipokines [26], i.e lipoprotein lipase (LPL), leptin,

angiotensin and cytokines [27]. The first hallmark of an inflammatory state in adipose

tissue is increased levels of cytokines due to engagement in inflammatory pathways,

involving the nuclear factor kappaB (NF-κB) pathway, by metabolic signals [28, 29].

In response to elevated cytokine levels, inflammatory kinases are activated mediating

a modest low-grade inflammation. Phosphorylation of serine residues in insulin

receptor substrate (IRS)-1 by activated Jun N-terminal kinase (JNK) [30],

extracellular signal regulated kinase (ERK) [31], protein kinase (PK)-Cθ [32] and

mechanistic target of rapamycin (mTOR) [33] may target insulin IRS1 causing

impairment of the insulin receptor signalling cascade [34, 35]. Serine phosphorylation

of IRS1 by the inhibitor KappaB kinase (IKK) complex links inflammatory pathways

to insulin resistance [36]. Obesity-related inflammation is strongly associated with

CVD probably as an indirect effect through insults like hypertension and

hyperlipidemia, which may induce damage and reshaping of the endothelial cells [37-

40]. Endothelial damage provokes an inflammatory response recruiting growth

factors, cytokines, chemokines and potent vasoactive factors [41, 42].

Page 20: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

20

1.3 Atherosclerosis

The exact mechanisms of initiation and progression of endothelial cell injury are

unknown, but is assumed to involve genetics, environmental cues, tissue injury,

infections and oxidative stress. Elevated plasma lipid levels and high blood pressure

might contribute to the first event in the development of atherosclerosis; endothelial

cell damage [43]. Also, naturally occurring causes like turbulence, in particular where

arteries branch, can induce injury on endothelial cells [44]. Key factors involved in

the progression of endothelial damage are of therapeutic value and investigators are

trying to survey these elements.

Atherothrombosis is characterized by atherosclerotic lesions and Figure 2 gives a

brief description of the progression of atherosclerosis. Generally, it is hypothesized

that the early onset of atherosclerosis is initiated by endothelial dysfunction and once

wounded, the endothelial cells begin to produce surface adhesion molecules, i.e.

vascular cell adhesion molecule (VCAM)-1 and intercellular adhesion molecule

(ICAM)-1 causing adherence of monocytes and T-lymphocytes, recruited by

chemoattractant cytokines, i.e. monocyte chemoattractant protein (MCP)-1.

Cytokines induce epithelia injury including tight junction dismantling and epithelial

barrier permeability alterations involving the epidermal growth factor receptor

(EGFR) -dependent mitogen activated protein/extracellular signal-regulated kinase

(MAP/ERK) 1/2 -signalling pathway [45]. Although all lipoproteins, with the

exception of the large chylomicrons, are able to cross a normal, intact endothelial

layer circulating low-density lipoprotein (LDL) can infiltrate the arterial wall to a

greater extent in inflammation [37]. Accumulated LDL in the intima, the innermost

layer of the artery wall, is more prone to oxidation. Monocytes in the intima mature

into macrophages secreting interleukin (IL), granulocyte macrophage colony-

stimulating factor (GM-CSF) and granulocyte colony-stimulating factor (G-CSF),

among others. The oxidized LDL is not recognized by LDL receptors (LDLr), but

binds to scavenger receptors [46]. The clustering of oxidized LDL particles in

macrophages, converts them into foam cells, creating a lipid-rich core, and eventually

foam cells undergo apoptosis. Oxidized LDL can also induce migration and

Page 21: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

21

proliferation of vascular smooth muscle cells (VSMC) [47]. The T-lymphocytes (T-

cells) secrete cytokines provoking VSMC migration into intima, in addition to

smooth muscle cell proliferation. VSMC are responsible for the synthesis of the

collagen developing a fibrous cap, protecting the plaque from rupture. Recently, our

knowledge on the critical role of VSMC function and plasticity in atherosclerosis has

been substantially extended, indicating that a large portion of foam cells probably are

of VSMC origin [48]. Atherosclerotic plaque can develop without comprising the

lumen due to compensating changes of vessel size expanding the external elastic

membrane, known as positive remodelling [49]. Plaques developing slowly,

containing calcification which creates a mature fibrous cap, are stable and not prone

to rupture [50]. More rapidly growing plaques with a thin fibrin cap, are instable and

more likely to burst [51]. It Is assumed that positive remodelling creates a more

unstable plague compared to negative remodelling [52]. Ruptured plaque triggers

thrombosis and activates platelets and clotting factors, and this clotting cascade can

lead to blood clots resulting in heart attack or stroke [53, 54].

Page 22: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

22

Figure 2 - Development of Atherosclerosis 1. The arterial wall. 2. Endothelial cells are prone to damage and circulating LDL enters the intima. Once in the intima, LDL is susceptible to oxidation. 3. Accumulation of oxidized LDL recruits monocytes binding to adhesion molecules and are, together with chemoattractant proteins, responsible for their entrance into intima. 4. Oxidized LDL is recognized by scavenger receptors located on monocytes, which mature into macrophages. 5. Macrophages overloaded with oxidized LDL become foam cells releasing inflammatory cytokines. 6. Migration and proliferation of VSMC synthesize collagen and together with foam cells cluster into fatty streaks,

Page 23: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

23

known as atherosclerotic lesions. Remodeling of the vessel causes lesions progression without affecting luminal size. The fibrous cap shoulder is prone to rupture (Adapted from [52]). GM-CSF, granulocyte macrophage colony-stimulating factor; ICAM1, intercellular adhesion molecule 1; IL, interleukin; MCP1, monocyte chemoattractant protein 1; NOS2, nitric oxide synthase 2; VCAM1, vascular cell adhesion molecule 1; VSMC, vascular smooth muscle cell.

1.4 Dietary proteins

Protein is present in every cell throughout the body and is essential for building and

maintaining bones, muscle and skin. Proteins also serve as hormones and cytokines

synthesized, and released, to act as cell-cell signals, transcription factors, enzymes

and more. There are limited data pertaining protein and amino acid metabolism in

humans, thus the exact essential protein requirements is assessed from a minimum

level of protein necessary for maintaining a short-term nitrogen balance under

controlled conditions based on use for body protein synthesis and urea production.

Dietary protein is the source of essential amino acids required for growth and

maintenance. An overview of the essential and non-essential amino acids in humans

is given in Table 1. Also, protein digestibility should be accounted for in a balanced

diet. Differences in the rate of hydrolysis of protein from different sources could

affect digestion, gastric emptying, absorption and rate of removal, thus providing

variation in energy supply.

Whereas some proteins are suggested to suppress appetite [55], proteins from various

meat sources are assumed to affect satiety in different degrees [56]. Amino acid

precursors of the neurotransmitters serotonin and catecholamines, concentrations of

circulating hormones and metabolites (insulin, glucose, amino acids), as well as gut

factors are all believed to influence satiety. In a study comparing beef, chicken and

fish protein, fish was found to give the highest level of satiety [56]. This effect was

assumed to be attributed the tryptophan/large neutral amino acid (LNAA) ratio

suggesting involvement of the neurotransmitter serotonin, and consequently

serotoninergic activity [57-59]. A low plasma ratio of tryptophan/LNAA has shown

less desire to binge in vulnerable women after ingestion of a protein-rich meal [60].

Page 24: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

24

1.4.1 Dietary proteins can affect lipid metabolism

It is well known that dietary fatty acids influence lipid metabolism, as well as

carbohydrate and protein metabolism, through cell-specific gene expression [61].

This regulatory mechanism is mainly influenced by the fatty acid composition of

membranes, body fat distribution and excessive consumption of fats [62-64], but

more recent work shows that also dietary protein affects lipid metabolism in rodents

[65, 66] and humans [67]. Diets high in protein causes less lipid accumulation in the

liver due to a rise in very low-density lipoprotein (VLDL) production rate

independent of fat content of the diet [65]. In one study in mice, enhanced expression

of genes involved in protein catabolic processes and transamination was shown, after

ingestion of a high protein diet [65]. Also, increased mitochondrial biosynthesis was

detected and increased expression of genes encoding enzymes participating in the

tricarboxylic acid (TCA) cycle, or citric acid cycle, thus energy utilization was

increased [65].

Proteins from soy are known to reduce cholesterol levels and reduce oxidative

damage to lipids [68], however uncertainty around the mechanisms behind these

effects remain. Also which constituents of the protein the effects are attributed to, the

amino acid content or isoflavones, is also somewhat debated. However, the

cholesterol reducing effect of soy has been linked to increased faecal excretion of

neutral sterols and cholesterol through bile, thus upregulating bile acid synthesis and

LDLr activity in rats [69]. The digestibility of the protein could affect lipid

absorption, and peptides high in hydrophobic amino acids are suggested to reduce

lipid-carrying capacity of micelles by binding bile acids [70], in this way decreasing

fat absorption. To compensate for the increased loss of cholesterol through bile

excretion, increased hepatic cholesterol biosynthesis has been reported, through

enhanced activity of the rate-limiting enzyme in cholesterol synthesis; 3-hydroxy-3-

methylglutaryl-coenzyme A (HMG CoA) reductase [71, 72]. However, this can also

be a compensatory mechanism to reduce plasma cholesterol, thus removal of

cholesterol, in particular LDL from the blood [73]. It is assumed that this effect could

Page 25: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

25

be linked to the isoflavone content of soy protein (i.e. genistein and daidzein) [74,

75].

Recently protein from marine sources have emerged as cholesterol and lipid-lowering

diet components [76, 77] affecting VLDL secretion [76]. Meat and fish are the main

sources of protein in the Western world and contain bioactive peptides offering

antioxidant, antihypertensive, antimicrobial and antiproliferative properties [78-81],

all of which can reduce the risk of CVD. Antihypertensive and antioxidant capacities

are of particular interest in this regard. Antioxidants are demonstrated to inhibit LDL

oxidation and cellular lipid peroxidation, thus attenuating cell-mediated LDL

oxidation [82]. Several protein hydrolysates are shown to possess antioxidant effects

[79, 81, 83], and consequently may protect LDL from oxidation [84]. Hypertension is

a major risk factor for coronary disease, myocardial infarction, congestive heart

failure and stroke [85]. Protein from animal muscles [86], a chicken collagen

hydrolysate [87] and a fish protein hydrolysate [88] have appeared antihypertensive

through an angiotensin-converting enzyme (ACE) -inhibiting effect.

1.4.2 Amino acid profile

Taurine is a sulphur-containing organic acid, included among the amino acids, and is

involved in osmoregulation, calcium modulation and stabilization of membranes [89].

Taurine is one of the most abundant amino acid in the body, where it is synthesized

from methionine and cysteine, and is also found in high amounts in seafood. The

most documented effect of taurine is the conjugation with bile, and studies in humans

have shown that taurine levels are lower in obese subject [90] and in persons with

diabetes [91]. Taurine has also been linked to a cholesterol-lowering effect through

its involvement in the catabolism of cholesterol into bile [92], probably due to

increased expression and activity of the rate-limiting enzyme in bile acid synthesis,

cholesterol 7 alpha-hydroxylase (CYP7A1) [93], as well as upregulation of LDLr

activity increasing cholesterol clearance from the circulation [94], and cholesterol

excretion through bile by upregulation of the adenosine triphosphate-binding cassette

(ABC) transporters ABCG5 and ABCG8 [93].

Page 26: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

26

1.4.3 Amino acid ratios

The ratios between the amino acids lysine/arginine and methionine/glycine have also

been purposed to influence lipid and cholesterol levels [95]. The low lysine/arginine

ratio of soy has been demonstrated to decrease and increase secretion of insulin and

glucagon, respectively [63], thus favoring lipolysis instead of lipogenesis. Protein

synthesis and proper folding of protein are essential for both cellular function and

avoiding cellular toxicity. Lysine is an indispensable amino acid, as well as a

precursor for other amino acids and is involved in collagen formation via its

hydroxylation to hydroxylysine [96, 97]. A low level of this amino acid may lead to

impaired protein synthesis [97] affecting high-turnover-constituents, which are

dependent on proper protein functions, like the apolipoproteins. Incorrect folding

leads to weakened receptor-mediated uptake of these particles, loss of sensitivity

towards proteolysis and increased susceptibility to oxidation [98], probably

increasing the risk for atherosclerosis. Carnitine is synthesized from lysine (and

methionine) [99], and is essential for fatty acid transportation into mitochondria for β-

oxidation, which could contribute to the lipid-lowering effect of lysine. Arginine is

linked to improved endothelial cell function, as it is the main substrate for nitric oxide

synthase (NOS) synthesizing nitric oxide (NO), which is an important cellular signal

molecule [100].

Low ratio of methionine/glycine has also demonstrated to decrease liver cholesterol

mainly through increasing HMG-CoA reductase activity [101]. High level of

homocysteine is associated with increased risk of endothelial damage and vascular

inflammation, resulting in atherosclerosis [102]. Homocysteine is synthesized from

the amino acid methionine in a multiple-step reaction where methionine is

demethylated ultimately giving homocysteine [103]. Activated methionine is the most

important methyl donor in the body formed by the transfer of an adenosyl group from

adenosine triphosphate (ATP) -yielding S-adenosylmethionine (SAM), catalyzed by

methionine adenosyltransferase (MAT). The methylation reaction takes place when

SAM donates a methyl group, yielding S-adenosylhomocysteine (SAH). SAH is an

inhibitor of methylation reactions, thus rapidly degraded. In this step homocysteine is

Page 27: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

27

formed, under the action of SAH hydrolase removing the adenosyl group. It has been

suggested that the methyl group of methionine is responsible for the cholesterol-

elevating effect of methionine [104]. Glycine is associated with lower plasma

cholesterol levels [105].

Although the sulphur of amino acids have been linked to a cholesterol-increasing

effect, cysteine is suggested to reduce oxidative stress [106]. Thus, specific amino

acids have been shown to reduce cholesterol and oxidative stresses related to CVD,

and improve cellular conditions preventing ischemic injury.

Indications of beneficial effects from specific combinations of amino acids, rather

than whole proteins, have caused a growing interest of hydrolyzation of protein from

raw materials aiming for an optimized nutritional value. Hidden within the parental

protein appears to be bioactive peptides offering health improving potentials, which

are released during gastrointestinal digestion or food processing. Hydrolyzation of

proteins seems to have a more prominent effect, perhaps due to increase

bioavailability of the bioactive peptides.

Table 1 - Essential and non-essential amino acids in humans

Essential amino acids Non-essential amino acids

Histidine* Alanine

Isoleucine Arginine

Leucine Asparagine

Lysine Aspartic acid

Methionine Cysteine

Phenylalanine Glutamic acid

Threonine Glutamine

Tryptophan Glycine

Valine Histidine*

Proline

Serine

Tyrosine

*Indispensable in infants

Page 28: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

28

2. AIMS OF STUDY

The general purpose of this thesis was to survey the effect of: 1) bioactive peptides

from hydrolyzed salmon and 2) chicken meat as a protein source, and investigate

their influence on lipid metabolism using rodent models. Further, we wanted to

elucidate if peptides from salmon and/or chicken protein (CP) could attenuate

atherosclerotic development and inflammation in an apolipoprotein (apo)-E knockout

mice model.

More specifically:

Study I: In the first experiment we assessed different fractions of a salmon

protein hydrolysate (SPH) and their lipid-lowering effect in male

C57BL/6 mice.

Study II: Based on study I, we used the SPH fraction (E1) showing most lipid-

lowering potential to investigate atherosclerosis and inflammation in

female apoE-/- mice.

Study III: The effect of chicken as a protein source on lipid metabolism was

evaluated in male Wistar rats.

Study IV: CP was further investigated in apoE-/- mice. Our working hypothesis was

that CP would lead to less atherosclerosis.

Page 29: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

29

3. THEORETICAL BACKGROUND

3.1 Lipids

Lipids are a group of organic compounds insoluble in water, varying in size and

polarity from hydrophobic triacylglycerols (TAG) to more soluble phospholipids.

Lipids also include cholesterol, cholesteryl esters and phytosterols. The water-

insolubility of lipids forces them into specialized processes during digestion,

absorption, transport, storage and utilization.

3.1.1 Fatty acids

Fatty acid is a common term of aliphatic molecules consisting of a hydrocarbon

backbone with a carboxylic acid end. The hydrocarbon chain length of dietary fatty

acids vary from 12-24 carbons, and can be subdivided into short-chained fatty acids

(SCFA, > 6 carbons), medium-chained fatty acids (MCFA, 6-12 carbons), long-

chained fatty acids (LCFA, 12-22 carbons) and very long-chained fatty acids

(VLCFA, > 22 carbons). The diversity of fatty acids is further increased by the

insertion of double bonds in the hydrocarbon chain. Saturated fatty acids (SFA),

consist exclusively of single bonds, mono-unsaturated fatty acids (MUFA) with a

single double bond, and poly-unsaturated fatty acids (PUFA) containing multiple

double bonds.

3.1.2 Glycerolipids

Glycerol contains three hydroxyl groups, which can be esterified typically by one,

two or three different fatty acids creating a glycerolipid. Diacylglycerols (DAG) have

two fatty acids esterified to the glycerol and have important biological functions, like

activating PK-C [107], whereas TAG have three fatty acids esterified to a glycerol.

Glycerolipid synthesis includes four steps, as shown in Figure 3. Glycerol-3-

phosphate is generated from phosphorylation of glycerol by glycerol kinase, or from

the reduction of dihydroxyacetone phosphate derived from glucose via glycolysis.

Page 30: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

30

The latter one is the only source for adipose glycerol-3-phosphate due to the lack of

glycerol kinase. Consequently, the adipose tissue can only store fat in fed state when

glycolysis is active. TAGs are the main storage form of lipids in living organisms,

and are also the largest proportion of dietary fat consumed by humans.

In contrast to glycogen, which is the glucose storage in animals, TAG do not bind

water and the energy density is much higher making it an excellent compound for

energy storage. TAG synthesis takes place mainly in liver in both endoplasmic

reticulum (ER) and mitochondria [108, 109], but also in adipose tissue and mammary

glands. TAG are synthesized from three potential sources; de novo lipogenesis,

plasma non-esterified fatty acids (NEFA) or lipoprotein remnants.

Phospholipids contain a DAG, a phosphate group and an organic molecule such as

choline. Phospholipids are the main constituents of lipid bilayer of cells and involved

in metabolism and cell signaling.

Figure 3 - Glycerolipid synthesis Glycerolipid synthesis includes four steps; acylation of glycerol-3-phosphate to form LPA by the action of GPAT [110]. GPAT exists in four isoforms, GPAT1 and 2 account for half of the activity in liver, whereas GPAT3 and 4 in ER predominates in adipose tissue. The next step involves an acylation of LPA to PPA followed by a hydrolyzation of the phosphate group by PAP forming DAG. The final step in glycerolipid synthesis is the acylation yielding TAG, catalyzed by the enzyme DGAT (Adapted from [111]). DAG, diacylglycerol; DGAT, diacylglycerol acyltransferase; GPAT, glycerol-3-phosphate acyltransferase; LPA, lysophosphatidic acid; PPA, phosphatidic acid; PAP, phosphatidic acid phosphatase; TAG, triacylglycerol.

Page 31: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

31

3.1.3 Digestion, Absorption and Transport

A challenge in fatty acid digestion and absorption is the solubility; fatty acids are

hydrophobic whereas the digestive enzyme lipase is hydrophilic. To overcome this

barrier, emulsification is necessary. Bile, combined with motility in the small

intestine, aid the process of emulsification breaking down large fat globules into

smaller particles surrounded by bile salts and phospholipids [112]. These emulsifying

droplets increase the surface area where the enzyme lipase can digest TAG into

monoglycerides and free fatty acids (FFA), which also associate with bile salts and

phospholipids, making micelles. The micelles assist in the absorption of fatty acids by

providing monoglycerides and FFAs into a pool, then fatty acids uses their

hydrophobic advantage diffusing passively through the membrane of enterocytes.

Once inside the enterocytes, TAG are reconstructed from monoglycerols and FFAs,

and then packed together with dietary cholesterol into lipoproteins.

Lipoproteins are macromolecular complexes of specific carrier proteins,

apolipoprotein, with various combinations of phospholipids, cholesterol, cholesteryl

esters and TAG. Each type of lipoprotein has specific functions depending on point of

synthesis, lipid composition, and apolipoprotein content. The largest of the

lipoproteins are the chylomicrons transporting dietary TAG from the intestine to

peripheral tissues, displayed in Figure 4. Chylomicrons travel through exocytosis

from the enterocyte surface and pass into the lymphatic capillaries before entering the

blood [112]. The apolipoproteins in chylomicrons include apoB-48, apoE and apoC-2

[113]. The latter one activates LPL in tissues like skeletal muscle, adipose tissue,

heart and mammary glands releasing the FFAs of the chylomicrons [114], creating

smaller lipoproteins, known as chylomicron remnants. When depleted of most of its

fatty acids, the chylomicrons remnants move to the liver where receptors recognize

apoE and mediates uptake by endocytosis [115]. Excess dietary fats, or carbohydrates

not immediately needed for fuel, are converted to TAG and packed into lipoproteins

called VLDL. VLDLs also contain cholesterol, cholesteryl esters, apoB-100, apoC-1,

apoC-2, apoC-3 and apoE [116]. VLDLs circulate the blood from liver to muscle and

adipose tissue, where apoC-2 activates LPL [117], and FFAs are removed. The

Page 32: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

32

VLDL remnants are cleared from the blood stream through receptor-mediated uptake

by apoE of the VLDL particle and liver receptors. Some of the VLDL remnants are

converted to intermediate density lipoprotein (IDL), rich in cholesterol and

cholesteryl esters. Further removal of TAG leads to the formation of LDL [118], as

illustrated in Figure 4. This lipoprotein, also very rich in cholesterol and cholesteryl

esters, contains apoB-100 as the major apolipoprotein [119]. LDL carries cholesterol

to extrahepatic tissue, which have receptors recognizing apoB-100.

High-density lipoprotein (HDL) originates from protein rich particles in the liver and

intestine, and contains little cholesterol and cholesteryl esters [120]. HDL contains

the enzyme lecithin-cholesterol acyl transferase (LCAT), which catalyzes the

synthesis of cholesteryl esters from phosphatidylcholine and cholesterol [121, 122].

Newly synthesized HDL is disk-shaped, but once the cholesterol ester content

increases creating a core, the HDL matures into a spherical HDL particle. This

particle is now rich in cholesterol and returns to the liver, known as reverse

cholesterol transportation, unloading the cholesterol, illustrated in Figure 4.

Page 33: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

33

Figure 4 - Lipoprotein metabolism TAG-rich chylomicrons are assembled in the intestine after lipid absorption. Chylomicrons and VLDL, which is synthesized in the liver, are stripped of TAG in peripheral tissues by LPL. TAG-deprived VLDL is remodeled in the liver to LDL, which delivers cholesterol to cells for incorporation into membranes or steroid synthesis. LDL binds to liver LDLr. HDL brings back excess cholesterol through reverse cholesterol transport. HDL, high-density lipoprotein; LDL, low-density lipoprotein; LDLr, low-density lipoprotein receptor; LPL, lipoprotein lipase; TAG, triacylglycerol; VLDL, very low-density lipoprotein.

3.1.4 Cholesterol, cholesteryl esters and bile acids

Cholesterol, like fatty acids, is synthesized from acetyl-CoA [123], but the assembly

is different. Cholesterol synthesis takes place in numerous stages, described in Figure

5.

Page 34: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

34

Figure 5 - Cholesterol synthesis Cholesterol synthesis takes place in multiple steps: First, two molecules of acetyl-CoA condense to form acetoacetyl-CoA, catalyzed by thiolase, then a third molecule of acetyl-CoA condense with acetoacetyl-CoA by the action of HMG-CoA synthase, forming a six carbon compound HMG-CoA. Three phosphate groups are transferred to mevalonate, two of these, in addition to a carboxyl group, leave in the next step, producing isopentenyl pyrophosphate, the first activated isoprene, which is isomerized to dimethylallyl pyrophosphate, the second activated isoprene. These two substrates undergo a head-to-tail condensation, forming geranyl pyrophosphate, which undergoes another head-to-tail condensation with isopentenyl pyrophosphate, forming the intermediate farnesyl pyrophosphate. Two molecules of farnesyl pyrophosphate join head-to-head, eliminating both pyrophosphate groups and yield squalene. The enzyme squalene synthase is regulated by the cholesterol content of cells. The addition of

Page 35: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

35

an oxygen atom to the squalene chain forms squalene epoxide and the linear squalene epoxide is converted to a cyclic structure, lanosterol, which contains the characteristic four rings of a steroid nucleus. After 19 reactions, lanosterol is finally converted to cholesterol (Adapted from [124]). CoA, coenzyme A; HMG-CoA, 3-hydroxy-3-methylglutaryl-Coenzyme A.

Cholesterol synthesis was believed to takes place mainly in cytosol of hepatocytes as

all of the enzymes involved are present there, except HMG-CoA reductase, which is

found in ER. However, recently all enzymes, with the exception of one, are also

discovered in the peroxisome [125]. Cholesterol is incorporated into membranes

modulating rigidity and permeability [126], and is an important precursor for steroid

hormones [127]. Some of the cholesterol synthesized in liver, is used in membranes

of hepatocytes. The fate of cholesterol in ER is divided into two different pathways;

bile synthesis and cholesteryl ester synthesis.

The major path of cholesterol catabolism is bile acid synthesis where cholesterol is a

substrate for the enzyme CYP7A1 [128]. The majority of liver cholesterol is

converted to bile acid; cholic acid is synthesized by the action of CYP7A1 inserting a

hydroxyl group at C7 of the steroid ring of cholesterol, and is the rate-determining

step of bile synthesis [129]. The primary bile acids, cholic acid and chenodeoxycholic

acid, are conjugated with either taurine or glycine [130], giving eight active

conjugated forms referred to as bile salts. Conjugation of the carboxylic end of bile

acids, gives a negative charge, fully ionized. Bile salts are more efficient detergent,

more secretable and less cytotoxic due to the enhanced amphipathic structure. Biliary

salts are the major solutes in human bile and aids in lipid digestion, as previously

described.

The turnover of the exchangeable pool of cholesterol participates in an incomplete

enterohepatic circulation, represented in Figure 6. Both free cholesterol and

cholesterol converted into bile is excreted through the bile duct into duodenum

mixing with dietary cholesterol. Partially reabsorption takes place in the jejunum,

whereas the majority of bile acid reabsorption takes place in the distal ileum. The

unabsorbed portion of cholesterol and bile acids derived from cholesterol, are

excreted through faeces, thus there is a continuous loss of cholesterol. The loss is

replaced by endogenous synthesis and reabsorption of dietary cholesterol, and the

Page 36: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

36

turnover is assumed to be such that 2% is renewed pr. day. As the majority of

cholesterol loss is through intestinal excretion, faecal output of bile acids and neutral

steroids determine the rate of cholesterol turnover.

In the opposite pathway, cholesterol is used for cholesteryl ester synthesis. The acyl-

coenzyme A cholesterol acyl transferase (ACAT) catalyzes the liver production of

cholesteryl esters [131]. Cholesteryl esters synthesized in the liver, is packed into

lipoprotein particles and secreted to cholesterol-demanding tissues, as formerly

described.

Page 37: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

37

Figure 6 - Enterohepatic circulation Cholesterol is metabolized to cholic acid, conjugated with taurine or glycine and released from the liver/gallbladder through the common bile duct into the small intestine. 95% of the bile is reabsorbed in the distal intestine, 5% excreted through the faeces. 5% of the reabsorbed bile enters the circulation. Plasma and newly synthesized cholesterol can be used for bile acid synthesis (Modified from [132]).

Page 38: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

38

3.1.5 Activation to acyl-CoA

Both dietary and de novo synthesized fatty acids requires esterification with a

coenzyme A (CoA) to be activated. This hydrolysis is carried out by the enzyme acyl-

coenzyme A synthase (ASC) present in the outer mitochondrial membrane, in

membranes of ER and peroxisomes. The insertion of this energy-rich thioester bond

is necessary for all fates of fatty acids, ranging from β-oxidation, synthesis of

glycerolipids, phospholipids and cholesteryl esters, desaturation and elongation. The

fatty acyl-CoAs are transported by fatty acyl-binding proteins (FABP).

3.1.6 Mitochondrial β-oxidation

Mitochondria are tubular-shaped, membrane-bound organelles located in cytoplasm

of eukaryotic cells. Mitochondria are the power station of the cell consisting of an

outer and an inner membrane surrounding the core of the organelle known as matrix.

Mitochondria are crucial in ATP synthesis [133] and are present in great numbers in

highly energy demanding tissues like cardiac- and skeletal muscles.

Mitochondria are the principle site of fatty acid oxidation, which takes place in the

matrix [134]. Although SCFAs and MCFAs can passively diffuse through the

mitochondrial membranes, LCFAs, which constitute the majority of dietary fatty

acids, requires a transport system. This challenge is overcome by the action of the

carnitine palmitoyl transferase (CPT) system, a three-step reaction termed carnitine

shuttle [135], shown in Figure 7. After the activation by ASC in the outer membrane,

the fatty acyl-CoA ester formed is transiently attached to the hydroxyl group of a

carnitine giving fatty acyl-carnitine, catalyzed by carnitine acyltransferase (CACT)-1.

After synthesis, the acyl-CoA ester moves into the matrix by facilitated diffusion

under the action of CPT1. The final step in this process is the enzymatically transfer

of the fatty acyl group from carnitine to intra-mitochondrial CoA by CPT2 [136].

Page 39: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

39

Figure 7 - Carnitine shuttle Fatty acid transportation across the mitochondrial membranes as fatty esters of carnitine starts off with activation by ASC in the outer membrane. The fatty acyl-CoA ester formed is transiently attached to the hydroxyl group of a carnitine giving fatty acyl-carnitine, catalyzed by CACT1. After synthesis, the acyl-CoA ester moves into the matrix by facilitated diffusion under the action of CPT1. The final step in is the enzymatically transfer of the fatty acyl group from carnitine to intra-mitochondrial CoA by CPT2 [136] (Modified from [137]). ASC, acyl-coenzyme A synthetase; CACT1, carnitine acyltransferase 1; CoA, coenzyme A; CPT, carnitine palmitoyltransferase, IMM, inner mitochondrial membrane; OMM, outer mitochondrial membrane.

Mitochondrial β-oxidation takes place in three stages and results in the removal of

two carbons in the form of acetyl-CoA, and is demonstrated in Figure 8.

Page 40: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

40

Figure 8 - Mitochondrial vs. peroxisomal β-oxidation The first step in mitochondrial β-oxidation is catalyzed by ACAD located in the inner mitochondria membrane, using FAD as a prosthetic group receiving the electrons from the fatty acyl-CoA. The electrons enter the respiratory chain contributing to ATP synthesis. After the insertion of a double bond between the α and β carbons, H2O is added to the double bond of trans Δ2-enoyl-CoA to form β-hydroxyacyl-CoA dehydrogenase, catalyzed by enoyl-CoA hydratase. The splitting of the double bond between these two carbons forms L-β-hydroxyacyl-CoA. In the next step, H2O is removed through a dehydrogenation reaction, yielding β-ketoacyl-CoA. In this reaction NAD+ is reduced to NADH by accepting electrons from this reaction and passing them on to NADH dehydrogenase, the first electron carrier in the respiratory chain. The shortening of the fatty acyl chain by the reaction between β-ketoacyl-CoA and a free CoA represent the final step in β-oxidation, catalyzed by thiolase (ACOT), and the resulting fatty acyl is reduced by two carbons, by releasing acetyl-CoA. After finishing one passage through the β-oxidation, the fatty acyl-chain recycles until it is completely oxidized. The first reaction in peroxisomal oxidation does not contribute to ATP synthesis. The two next reactions are similar to mitochondrial oxidation, but involve other enzymes. Both oxidations result in a release of a fatty acyl-CoA and acetyl-CoA (Adapted from [138]). ACAD, acyl-coenzyme A dehydrogenase; ACOT, acyl-coenzyme A thioesterase; ATP, adenosine triphosphate; FAD, flavin adenine dinucleotide; NAD, nicotinamide adenine dinucleotide.

Page 41: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

41

The acetyl-CoAs released can enter the TCA cycle to generate ATP, or it can be used

for ketone body synthesis forming acetoacetate, β-hydroxybutyrate and acetone.

Although acetoacetate and β-hydroxybutyrate are synthesized in the liver, they cannot

be used as an energy source here, but are transported to other tissues, like the brain,

reconverted to acetyl-CoA and then used as fuel. Acetone is a waste product of the

decarboxylation reaction of ketone body production and exhaling secretes most of the

acetone. Starvation, high-fat-low-carbohydrate diets or prolonged endurance exercise

leads to production of ketone bodies from fatty acids in the liver [139].

3.1.7 Peroxisomal β-oxidation

β-oxidation is also carried out in the peroxisome [140], as described in Figure 8,

shortening LCFAs to MCFAs making them suitable for mitochondrial β-oxidation

[141]. Oxidation in peroxisomes and mitochondria is quite similar; a comparable

membrane shuttle of fatty acids [142] and activation by addition of acyl-CoA, but

some details differ. The first reaction in peroxisomal oxidation does not contribute to

ATP synthesis [143] and is therefore a less efficient energy-providing source. The

two next enzymes differ from the two in mitochondria, although they catalyze the

same reactions. Both oxidations result in a release of a fatty acyl-CoA and acetyl-

CoA. The shortened fatty acyl is subsequently shuttled to mitochondria for further

oxidation.

3.1.8 Lipogenesis

The body can synthesize a diversity of fatty acids from the substrates acetyl-CoA and

malonoyl-CoA under the action of the rate-limiting enzyme in fatty acid synthesis,

acetyl-coenzyme A carboxylase (ACC). Biosynthesis of fatty acids is a four-step

repeating cycle, catalyzed by the enzyme fatty acid synthase (FAS) complex,

involving the addition of a two-carbon unit, explaining why most natural occurring

fatty acids are even-numbered. Notably, some fatty acids from cow milk are odd-

numbered due to the ruminal bacteria of cattle [144].

Page 42: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

42

Figure 9 - Fatty acid synthesis

Biosynthesis of fatty acids is a four-step repeating cycle consisting of a condensation, reduction, dehydration and another reduction, involving the addition of a two-carbon unit (Inspired by [145]). ACP, acyl carrier protein; ADP, adenosine diphosphate; ATP, adenosine triphosphate, BHBA, beta-hydroxy-butyric acid, CoA, coenzyme A, CPT, carnitine palmitoyl transferase, FAS, fatty acid synthase, MUFA, mono-unsaturated fatty acid; NADP, nicotinamide adenine dinucleotide phosphate, SFA, saturated fatty acid

Fatty acid synthesis is outlined in Figure 9 and takes place mainly in the cytosol of

hepatocytes, but also to some extent in adipose tissue. The protein acyl carrier protein

(ACP) activates fatty acid synthesis, a part of the fatty acid synthase (FAS) complex,

which includes all enzymes participating in the pathway. The first reaction is a

carboxylation of acetyl-CoA to malonyl-CoA, catalyzed by the enzyme ACC. The

acetyl-CoA is generated from catabolism of glucose and the carbon skeleton of amino

acids. Malonyl-CoA is considered a precursor of fatty acids synthesis and an inhibitor

of fatty acid oxidation [71].

3.1.9 Elongation and desaturation of fatty acids

Fatty acids synthesis usually produce 16:0 (palmitic acid), and in minor amounts 18:0

(stearic acid). These fatty acids are highly represented in membranes, in brain

Page 43: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

43

however, there is a substantial requirement for LCFAs. Mainly liver and brain

mitochondria, or the surface of ER, have the capacity to carry out elongation, ER

being the most active [146, 147]. Elongation and desaturation are shown in Figure 10.

Figure 10 - Elongation and desaturation of fatty acids

Elongation of fatty acids resembles fatty acid synthesis, but the activities are separable involving different enzymes. Malonyl-CoA is the source of the two carbons added. Desaturation involves the removal of two hydrogens from the fatty acid yielding a carbon-carbon double bond.

Elongation in ER is similar to fatty acid biosynthesis, whereas mitochondrial

elongation is essentially the reversal of β-oxidation, but both involve addition, and

subsequent, reduction of acetyl groups. CoA derivatives must be activated for the

donation of a two-carbon unit by malonyl-CoA. The additional steps are followed by

reduction, dehydration and another reduction reaction. Mitochondria mainly elongate

SCFAs.

Page 44: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

44

Desaturation of fatty acid is another essential event in lipogenesis. Stearoyl-

coenzyme A desaturase (SCD)-1 is located in the membrane of ER and responsible

for the insertion the first cis-double bond between C-9 and C-10 in the hydrocarbon

chain, therefore often referred to as Δ9 desaturase [148, 149]. SCD1 is the rate-

determining enzyme in the oxidative desaturation of MUFAs and the preferred

substrates are 16:1n-7 (palmitoyl-CoA) and 18:1n-7 (oleoyl-CoA) [150]. The fatty

acid desaturase (FADS) family includes FADS1 and FADS2, also called Δ5

desaturase and Δ6 desaturase, respectively, which catalyze biosynthesis of highly

unsaturated fatty acids.

3.1.10 Ceramides and sphingolipids

Relatively similar to the glycerolipids are the sphingolipids, but sphingolipids are

derived from sphingosine instead of glycerol. Ceramides are the precursors of

sphingolipids and are converted to sphingolipids in the liver [151]. Sphingolipids

serve as structural components of the plasma membrane, however recently their role

in inflammatory disease has been identified. Activity of sphingolipid enzymes have

been shown to increase in response to elevated cytokine levels [152]. Glycerolipids

and sphingolipids cross talk as second messengers to control vesicle movement, cell

division and cell death.

3.2 Protein

Protein is formed when amino acids are joined together by peptide bonds initially

forming peptides. Peptides containing more than 60 amino acids are designated

proteins. The amino acid sequence of a protein is called the primary structure and

further creates helixes or sheets, posing the secondary structure, and subsequent fold

into three-dimensional structures, known as tertiary structure [153]. Protein

containing two or more separate polypeptide chains arranged into a three-dimensional

structure creates a quaternary structure.

Page 45: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

45

3.2.1 Catabolism of protein and amino acids

Dietary protein and intracellular protein, mainly from the breakdown of muscles can

be degraded to amino acids to provide energy. Inflammation has shown to increase

muscle breakdown [154] and the amine group of the amino acid is separated from the

carbon skeleton and used in biosynthetic pathways, or excreted as urea. The

remaining carbon skeleton is converted to α-keto acids subsequently entering the

citric acid cycle, and ultimately the carbon skeletons are diverted to gluconeogenesis,

ketogenesis or completely oxidized to CO2 and H2O [155, 156].

Glutamine is the most abundant amino acid in the blood and a general collection

point of amino groups, donating an amino group in both biosynthesis and elimination

of nitrogenous products. In skeletal muscles, the amino groups are transferred to

pyruvate to form alanine, important in transportation of amino groups to the liver,

which is the only organ carrying out a complete urea cycle [157], and the major site

for amino acid metabolism.

3.2.2 Enzymatically breakdown of dietary proteins

In response to ingested protein, the gastric mucosa lining the stomach secretes the

hormone gastrin, stimulating the cascade in which hydrochloric acid and pepsinogen

are secreted as well. The acidity of the gastric juice unfolds globular proteins and

exposes their peptide bond making them available for enzymatic hydrolysis by

proteinases. Through the action of pepsin, polypeptide chains are broken down to

smaller peptides by hydrolysis of peptide bonds on the amino-terminal of the

aromatic amino acid residues; phenylalanine, tryptophan and tyrosine. As the acidic

chyme of the stomach enters the small intestine, the low pH triggers release of

secretin in the blood, which promotes pancreatic deliver of bicarbonate into the upper

part of the small intestine, duodenum, to neutralize the gastric acid. In response to the

presence of amino acids in the duodenum, cholecystokinin is released, stimulating

secretion of the inactive precursors [158] trypsinogen, chymotrypsinogen and

procarboxypeptidases, synthesized and released by the exocrine cells of the pancreas.

Further hydrolysis of the peptides produced under the action of pepsin in the stomach

Page 46: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

46

is efficiently performed by trypsin and chymotrypsin due to amino acid specificities

of these three enzymes. The various zinc-containing carboxypeptidases cleave basic

and acidic C-terminal amino acids [159], to complete the degradation of peptides in

the small intestine, together with aminopeptidase, which hydrolyzes the amino-

terminal residues. Breakdown of dietary protein appears to yield more small peptides

than free amino acids [160]. The resulting mixture of free amino acids, di- and some

tripeptides are then transported to the epithelial cells through secondary active

transport via the Na+K+ -pump. The accumulation of amino acids, di- and tripeptides

in the epithelial cells during digestion, results in passive transport of these molecules

via solute carrier to the blood capillaries and subsequent to the liver [137].

Hydrolyzed proteins in the diet are believed to be absorbed more easily, since

peptides are smaller than proteins and the peptides bond more exposed, thus the

peptidases are thought to gain more access to breakage of the peptide bonds. Also,

some peptides could be resistant to digestion, only increasing satiety, thus passing

through the digestive tract without contributing with energy [56].

3.2.3 Transamination and oxidative deamination of amino acids

The catabolism of most amino acids starts with transamination of the amino groups to

α-ketoglutarate by the enzyme pyridoxal phosphate, yielding glutamate. The removal

of the amino group of glutamate in the hepatic mitochondrial matrix for excretion is

called oxidative deamination, catalyzed by glutamate dehydrogenase. and is a

convenient manner to convert amino acids to its α-keto acid forms [161]. The α-

ketoglutarate produced can enter the citric acid cycle or gluconeogenesis whereas the

NH4 can be used for urea production.

In skeletal muscles glucogenic amino acids can be broken down for use as fuel and

the amino group in the form of glutamate can be further transferred to pyruvate by

alanine transferase forming alanine. Blood alanine travels to the liver where pyruvate

and glutamate is reformed. Thus, the glucogenic amino acids are converted glucose in

the liver for use as fuel, while ketogenic acids are converted to ketone bodies as brain

fuel [162].

Page 47: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

47

3.2.4 Nitrogen excretion and pathways of amino acid degradation

Urea production takes place mainly in hepatocytes [163]; more precisely it starts off

in mitochondria and continues in the cytosol. Ornithine accept a carbamoyl group

from carbamoyl phosphate forming citrulline, which passes from the matrix to the

cytosol further accepting a second amino group from aspartate, which yields

argininosuccinate. The proceeding of the urea cycle involves two steps of cleavage;

argininosuccinate is cleaved into fumarate and arginine, which is further cleaved into

urea and ornithine. Urea passes into the bloodstream and travels to the kidney for

excretion as urine. All urea enzymes are shown to correspond with protein

consumption in rats [164] As for the carbon skeleton of amino acids, some are

converted to glucose, others to ketone bodies, as previously described. The major

focus of this thesis is on arginine, lysine, methionine and glycine, which all are

glucogenic acids, except lysine which is ketogenic. When degraded, arginine is

converted to glutamate and succinyl-CoA, lysine is converted to acetoacetyl-CoA and

further to acetyl-CoA, methionine is converted to propionyl-CoA and subsequently to

succinyl-CoA, and glycine is converted to pyruvate and further to acetyl-CoA, thus

all is converted to substrates of the citric acid cycle.

3.2.5 Bioactive peptides

In addition to their nutritional value, bioactive peptides exert a physiological

effect in the body. Meat and fish are valuable sources of proteins and appear potential

as novel sources of bioactive peptides displaying antihypertensive, antioxidant,

antimicrobial and antiproliferative effects [80, 81, 165, 166]. Upon hydrolysis by

digestive enzymes or bacterial fermentation, peptides are fractionated based on size,

usually performed by ultrafiltration. To separate individual peptides, reverse-phase

high performance liquid chromatography (RP-HPLC) or gel permeation

chromatography is used. To identify individual peptides, a combination of mass

spectrometry and protein sequencing are performed.

The most established effect of food-derived bioactive peptides is the inhibitory effect

on the vasoconstrictor ACE by binding directly to this enzyme, inhibiting the

Page 48: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

48

conversion to its active form [82] , thus reducing hypertension, an important risk

factor for CVD. In order to perform ACE-inhibitory effect, peptides need to arrive at

the target organ in an intact form, thus being resistant to digestion.

Discarded bi-products of food processing are of major concern and are mostly

incorporated to animal feed. Adding value to these side-products could be aided by

the demonstration that they may be used to generate bioactive peptides [167]. Salmon

muscle has also been shown to contain peptide sequences contributing to ACE-

inhibitory effect [168]. Of the peptides showing an ACE inhibitory effect, short

chained, polar and low hydrophobic amino acid content displayed the most prominent

effect [169].

Antioxidant potential from natural foodstuff has emerged as a preferred source

compared to synthetic antioxidants due to less side effects [170]. Antioxidant

potential is dependent on peptide size, amino acid profile, and the content of free

amino acids within the hydrolysate.

3.3 Transcriptional regulation of lipid and amino acid metabolism

Fatty acids regulate gene expression, activity and abundance of the transcription

factors peroxisome proliferator-activated receptors (PPAR), liver X receptor (LXR),

hepatocyte nuclear factor (HNF)-4α, NF-κB and sterol regulatory element-binding

proteins (SREBP) [61]. Also, it has been reported that dietary peptides control amino

acid oxidation through PPARα [171].

3.3.1 Peroxisome proliferator-activated receptors

PPARs are nuclear hormone-receptors regulating lipid and glucose metabolism, and

recent investigation indicate that PPARs participate in the regulation of amino acid

metabolism as well [172, 173]. Three isoforms of PPARs have been discovered; α,

β/δ, and γ. PPARs are ligand-dependent, and are typically activated by fatty acids and

their eicosanoids [174]. Once activated, PPARs form a heterodimer with retinoid-X-

Page 49: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

49

receptor (RXR), which bind peroxisome proliferating receptor elements (PPREs),

stimulating, or suppressing, transcription of target genes [175, 176]. PPARα is highly

expressed in energy demanding tissues like liver, heart, skeletal muscle, kidney and

brown fat [177], and regulates genes involved in fatty acid uptake and catabolism,

inflammation and vascular function [178]. PPARα also regulates amino acid

oxidation in the liver, and is assumed to be a key factor regulating intermediate

metabolism during fasting state [173]. PPARγ is primarily limited to brown and white

adipose tissue [179], but also found in the large intestine [180] regulating genes

engaged in fatty acid uptake and storage, inflammation and glucose homeostasis

[180, 181]. PPARδ is moderately expressed in most tissue regulating genes

participating in fatty acid metabolism, inflammation and macrophage lipid

homeostasis [182]. PPARα activation facilitates fatty acid oxidation by increased

expression of LPL and apoA5 [183], and a decrease in apoC3 [184]. This leads to

liberation of fatty acids available for storage in adipocytes or metabolized in muscles.

Once activated, PPARα raises the HDL level due to an increase in hepatic apoA1

synthesis and ABCA1 expression, stimulating HDL-mediated cholesterol efflux

through macrophages [185, 186]. PPAR agonists contribute to increased fatty acid

oxidation and decreased inflammation [187, 188], and are therefore an interesting

field to investigate. Also, it has been suggested that PPARα reduces amino acid

catabolism [189] by regulating hepatic amino acid degrading enzymes (AADE)

through HNF4α activity, preserving amino acids by stimulating fatty acid oxidation

during fasting state [171]. Catabolism of amino acids, catalyzed by AADEs generates

metabolic energy, as well as substrates for gluconeogenesis.

3.3.2 Sterol regulatory element-binding factors

Lipids have many important functions in an organism, and SREBPs are considered a

major regulator of cholesterol synthesis and transportation, and synthesis of fatty

acids, TAG and phospholipids as these transcription factors have the ability to

activate a cascade of enzymes involved in lipogenesis and cholesterol synthesis [190].

In addition, SREBP stimulate synthesis of NADPH [191], which is needed for several

synthesis processes.

Page 50: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

50

SREBPs are synthesized in ER as inactive precursors and activated in response to

sterol deprivation, or high circulating insulin levels [192]. SREBP-cleavage

activating protein escorts SREBP precursors to Golgi where cleavage of the amino-

terminal takes place [193-195]. After translocation to the nucleus, SREBPs bind

promotor regions, SREBP response elements, of genes involved in fatty acid and

cholesterol synthesis and transportation [196]. Liver and adipose tissue are the main

tissues involved in export and storage of lipids, thus SREBPs are most active in these

organs. There are three isoforms of SREBP, referred to as SREBP1a, SREBP1c and

SREBP2. SREBP1c is primarily expressed in liver regulating genes involved in fatty

acid synthesis, whereas SREBP2 is mainly present in liver and adipose tissue

modulating genes engaged in cholesterol synthesis. Bioactive compounds can

influence hormones and cholesterol influencing ligands ability to bind receptors,

hence some genes can be preferentially activated. Insulin and glucagon can modulate

gene expression in this manner, and insulin and glucagon promotes and inhibits

SREBP-1c, respectively [197]. Circulating levels of insulin and glucagon are

influenced by diet constituents. Soy protein display a much lower short term insulin

concentration [198, 199], and higher long-term glucagon [200] concentration as

compared to a casein diet, thus SREBP is supressed and lipogenic gene expression is

reduced. Soy proteins are also known to reduce plasma and hepatic cholesterol in

animals and humans [198, 201-204], and in response, SREBP2 expression is

enhanced increasing cholesterol biosynthesis, in particular HMC-CoA reductase

activity, and cholesterol uptake, mainly LDLr activity. The ability of soy protein to

modulate insulin/glucagon levels has been linked to low lysine/arginine ratio

corresponding to a higher glucagon levels. LDLr is under control of SREBP2

transcription and upregulation of LDLr could be linked to the reduced plasma and

hepatic cholesterol levels observed in soy protein feeding. Glycine-containing

peptides, β-conglycinin and glycinin are found to upregulate LDLr activity [205].

Page 51: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

51

4. EXPERIMENTAL MODELS

4.1 Wistar rats

Wistar rats are an outbred strain from the albino rats belonging to the strain Rattus

norvegicus [206] developed and distributed in 1906 and throughout 1940s at the

Wistar institute [207]. As early as in the 1800 albino rats were separated from other

rat groups and used for rat shows. During this period the albino rats find their way

into the laboratory and became the first animal used in research. Henry Donaldsen

and colleagues put a lot of effort into optimizing the albino rats for use in many

research fields. In 1960 the Wistar Institute in Philadelphia sold the rights to other

commercial companies.

Wistar rats are bred under standard laboratory conditions and are characterized by

long ears and having a tail-length shorter than body length. The Wistar rats are

domestic rats making them more tractable, calmer and less likely to bite compared to

wild types. There are many advantages using rats in scientific research; they are

inexpensive, available, easy to handle and house, and can be used for a broad

spectrum of disciplines including physiology, nephrology, endocrinology, nutrition,

metabolism, drug evaluation, toxicology and transplantable tumors.

4.2 C57BL/6J mice

The most popular animal for laboratory use are the C57BL/6J mice and this model is

widely used in many research areas studying human disease [206]. These mice are an

inbred strain [208], which means that the mice are traceable to a single ancestral pair

in at least 20 generation. The background of C57BL/6J mice contains both

spontaneous and induced mutations, and is also used in production of transgenic

mice. The mouse strain is homozygous for Cdh23ahl, resulting in age-related hearing

loss with onset after 10 months. The C57BL/6J strain was developed in 1921 by CC

Little at the Bussey Institute for Research in Applied Biology [209], and these mice

develop obesity, hyperlipidemia and insulin resistance on a high-fat diet.

Page 52: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

52

4.3 ApoE-/- mice

Mice are highly resistance to atherosclerosis, except the C57BL/6 strain, which

develop plaque when fed a high-fat diet. In 1992 the apoE knockout mice model was

evolved from the C57BL/6 strain and still widely used as an experimental model for

studying atherosclerosis [210]. ApoE is a constituent of chylomicrons and

intermediate density lipoprotein essential for normal TAG catabolism. ApoE is

synthesized in various organs, mainly in liver cells and central nervous system. The

plasma lipoprotein levels and their metabolites are dependent on apolipoproteins. The

apoE-knockout model was developed in the cell line 129P2/olaHsd, derived from cell

line EI4TG2a ES. Two plasmids were used, pJPB63 and pNMC109, founder line T-

89 in the primary reference. ApoEtm1Unc was backcrossed 10 times to C57BL/6J mice

[211] and intercrossed to homozygosity [212]. Insertion of a neomycin resistance

cassette deleted part of the exon 3 and part of intron 3 of the apoE gene, thus the gene

was knocked out. Mice homozygous for the ApoEtm1Unc are hypercholesterolemic

with elevated plasma cholesterol levels independent of age, gender and diet. On a

standard chow diet, apoE mice have a total plasma cholesterol level of 4-500 mg/dL

mainly due to accumulation of VLDL remnants caused by impaired clearance of

these particles due to knockout of the apoE gene [213]. Accordingly, these mice are

prone to develop severe atherosclerotic lesions at a relatively early age. ApoE mice

can be fed a Western diet or a high-fat diet to develop more pronounced lesions at a

shorter time. ApoE-/- mice are widely used in studying atherosclerosis as they give the

opportunity to investigate the involvement of inflammation and immune mechanisms,

pathogenesis and therapy of atherosclerosis, in addition to genetic influence.

Page 53: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

53

5. SUMMARY OF RESULTS

Paper I:

Three differently generated salmon protein hydrolysates reveal opposite effects

on hepatic lipid metabolism in mice fed a high-fat diet

We wanted to explore any possible health benefits from 5% SPH generated using

different enzymatic hydrolyzation processes and microfiltration methods, and found

that C57BL/6 mice fed the peptides E1 and E4 displayed lower body weight, despite

a similar feed intake, as well as a lower total weight gain compared to the casein diet.

In contrast, the E2 group displayed a growth curve and feed intake comparable to

controls. Peptide E1 and E4 groups also showed lower plasma and liver TAG levels.

In the E1 group this was linked to reduced hepatic gene expressions of fatty acid

synthase (Fasn) and the rate-limiting enzyme in fatty acid synthesis, Acaca. Also,

Fads1 and Fads2, involved in Δ5 and Δ6 desaturation, respectively, tended to be

lower in E1.

In the E4 group, mitochondrial β-oxidation was increased, demonstrated by higher

activity of CPT2. FAS activity tended to be lower and Fasn expression was

significantly lower in the E4 group, suggesting a concomitant increase in β-oxidation

and reduction in lipogenesis in this group. As seen in E1, Fads1 was lower expressed

in this group. HMG-CoA reductase (Hmgcr) was reduced in both E1 and E4. In

contrast, mice fed the E2-diet demonstrated elevated hepatic TAG content compared

to controls, in addition to increased plasma TAG, cholesterol and phospholipid levels.

FAS activity was up-regulated in the E2 group, as were gene expression of Acot1,

indicating increased synthesis and liberation of fatty acids in this group.

SFAs were unaffected in all peptide groups. MUFAs tended to be reduced in the E1

group and were significantly reduced in the E4 group, whereas n-3 PUFAs were

increased in the E4 group, in accordance with an increase in Δ6 desaturase index. n-6

PUFAs were increased in the E1 group, as were the Δ5 desaturase index. The

opposite was observed in E2 where MUFAs were increased, corresponding to the

Page 54: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

54

increase in Δ9 desaturase indices, while n-3 and n-6 PUFAs were reduced compared

to controls. The total hepatic lipid level (TAG, cholesterol and phospholipids) was

unchanged in E1 and E4 compared to controls, whereas hepatic lipids were increased

in E2. This was in line with the relative difference (%) of TAG found in TAG

lipidomics. The differences in TAG in E1 compared to control was seen as a

reduction in all species of TAG, whereas the E2 group-diet primarily increased the

TAG species containing SCFAs, SFAs and MUFAs compared to control. The relative

difference of all d18:1 ceramides were increased in the E1 group, in contrast to the E2

group.

We observed diverse effects from three pre-digested peptide fractions on weight gain,

plasma and liver lipids and lipid synthesis, indicating that enzymatically

hydrolyzation of peptides can lead to distinct, and in some cases, opposite effects.

Paper II:

A salmon protein hydrolysate exerts lipid-independent anti-atherosclerotic

activity in apoE-deficient mice.

The SPH-E1 fraction was further investigated and after 12 weeks with dietary

intervention with 5% SPH treatment apoE-/- mice showed reduced atherosclerotic

plaque area in sinus and aortic arch. Also, a reduced gene expression of Icam in the

aortic arch was detected. Immunohistochemical determination of atherosclerotic

lesions in aortic sinus showed no differences in plaque content of connective tissue,

macrophages or lymphocytes, indicating that plaque stability was not affected.

Plasma arachidonic acid (C20:4n-6) and oleic acid (C18:1n-9) were increased and

decreased, respectively, after SPH-treatment. Plasma concentration of inflammatory

markers IL-1β, IL-6, TNF-α and GM-CSF were decreased, whereas plasma

cholesterol and TAG levels was unchanged.

These results showed that a 5% SPH diet reduced atherosclerotic development in

apoE-/- mice, and tended to decrease expression of adhesion molecules in aortic arch,

Page 55: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

55

thus possessed a local anti-atherosclerotic effect, but did not influence the stability of

the plaque area. The decrease of plasma inflammatory markers suggests a systemic

effect of the SPH as well, and that the reduction in plaque was independent of plasma

lipids.

Paper III:

Hypolipidemic effect of dietary water-soluble protein extract from chicken:

Impact on genes regulating hepatic lipid and bile acid metabolism.

After diet intervention with low-fat diets with doses of 6%, 14% or 20% CP for 4

weeks, Wistar rats fed the 20% CP dose displayed the highest feed intake, but gained

less weight. We found a dose-respond effect of CP in lowering the plasma

cholesterol, cholesterol-esters, TAG and phospholipids, measured as delta values.

Dietary supplementation with 20% CP decreased liver contents of TAG, cholesterol

and phospholipids. The 20% CP diet also changed the fatty acid composition in liver,

showing increased levels of α-linoleic acid (C18:3n-3) and docosapentaenoic acid

(DPA) (C22:5n-3). Hepatic β-oxidation was increased, accompanied by increased

CPT2 activity in the 20% CP group compared to the control group. Gene expression

of the transcription factor, Ppara, was increased, which might contribute to the

increase in fatty acid catabolism observed. Enzymes involved in fatty acid synthesis,

ACC and FAS, tended to decrease in the 6% and 14% CP fed groups, but only

reached significance in the 20% group. GPAT, involved in TAG synthesis, was also

significantly decreased in the two highest doses of CP. CP did not show any effect on

cholesterol synthesis, however, the gene expressions of Abcb4 were increased, which

could indicating increased bile transportation.

Carnitine turnover was assessed my measuring carnitine and its precursor and esters,

which all were increased, indicating improved mitochondrial function.

This study demonstrated decreased plasma and liver TAG and cholesterol by diet

intervention with CP. We concluded that the high feed intake and lowered plasma and

Page 56: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

56

hepatic lipids, without a high weight gain, could be due to poorer intestinal

absorption, a change in energy balance and/or increased faecal excretion.

Paper IV:

A water-soluble extract of chicken reduced plasma triacylglycerols, but showed

no anti-atherosclerotic activity in apoE-/- mice.

In apoE-/- mice given a 15% CP diet we detected marginal plasma TAG lowering

accompanied by indication of increased mitochondrial β-oxidation in CP-fed mice

compared to casein-fed controls after 12 weeks. Mice fed CP also displayed a lower

respiratory exchange ratio (RER) during inactive state, supporting higher energy

combustion from fat. There was no change in plasma cholesterol levels or fatty acid

composition, and en-face analysis of aorta displayed no change in plaque

development between the two groups. To further support these data, plasma

inflammatory markers were measured revealing unaltered systemic inflammation.

It appears that chicken protein is less potent in counteracting persistent disruptions on

lipid metabolism present in apoE-/- mice, and does not have the capability to affect

atherosclerotic development.

Page 57: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

57

6. DISCUSSION

The occurrence of overweight and obesity worldwide is growing in spite of numerous

dietary advices, effort and public education from health authorities. Although

consumers are increasingly attentive to food safety, quality and health-related issues,

obesity is of serious concern in the Western populations and CVD annually causes 17

million deaths [214]. The risk factors of metabolic syndrome resulting from

overweight and obesity lead to metabolic dysfunctions, evident as dyslipidemia and

systemic inflammation, and one of its clinical manifestations is atherosclerosis. The

present thesis aimed to elucidate two protein sources and their bioactive potential to

affect lipid metabolism and mitochondrial fatty acid oxidation, cholesterol and TAG

levels, as well as their capability of influencing inflammation, a hallmark in the

development of atherosclerosis. The major focus of the thesis has been centred on the

liver, plasma and heart/aorta, as liver is the main organ regulating both fatty acid

oxidation and synthesis. Plasma and heart pertain to systemic and local inflammation,

respectively.

6.1 Marine bioactive peptides

Fish consumption has been of great interest to investigate regarding bioactive effects

and the health benefits of fish intake is dedicated to the omega-3 PUFAs. Lately,

protein and peptides from various sources have been a focus of research as they

possess nutritional and essential properties in organisms, and are important

constituents in food production. Seafood is viable source of proteins, both

economically and environmentally, with balanced amino acid contents and high

nutritional value, showing promising health improving effects in both rat [76, 215]

and human studies [216]. Marine proteins possess antioxidant activity [217], ACE –

inhibitory action, anticancer effect [218] and cholesterol-reducing potential [76].

Peptides are inactive in their parental protein structure, but can be liberated by

digestion or commercial hydrolysis. Controlled hydrolysis of protein is the best way

to recover potent bioactive peptides in term of nutritional properties, namely a

Page 58: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

58

balanced amino acid composition and high digestibility [219]. Currently, most fish

raw material is discarded. This secondary raw material is well suited for recovery and

utilization as functional foods. The bioactive peptides can be extracted using specific

proteolytic enzymes to cleave definite peptide bonds and defined hydrolysis

conditions to generate desired peptides [220]. For the past 60 years, fish protein has

been hydrolyzed mainly for use in animal feed production. Recent work on

hydrolysis processes of fish protein has led to functional hydrolysates with optimal

nutritional availability for use as food supplements. Commercially manufactured

peptides are designed to achieve a defined molecular size, solubility in water and

nutritional balanced amino acid profile.

6.1.1 SPH reduced body weight

Numerous studies with protein and hydrolysates have shown improvements on blood

lipids, and dietary protein is known to affect lipid metabolism [221-223]. This was in

line with results for two of the salmon peptide fractions we assessed (Paper 1).

Among the three macronutrients, protein has the greatest thermic effect and satiety

value compared to both carbohydrate and fat [224-226] assumed to contribute to

lower body weight, both during normal or high dietary intake [227, 228], as observed

in our experiment (Paper 1). Although, given the same amount of protein, body

weights were distinct between the groups in our study.

Protein differs according to its source; proteins from animal origin are considered

complete protein containing all nine essential amino acids that the human body

requires, whereas proteins from plants are incomplete. Further, proteins from

different meat sources possess distinct effects as well, as varying degrees of satiety.

Fish protein has been shown to increase satiety linked to serotoninergic effect, or

differences in digestibility, thus slower gastric emptying [56]. Fish protein

hydrolysate has demonstrated increased hepatic mitochondrial fatty acid oxidation

[229]. Increase in CPT2 activity in group E4 was detected, which could also be

another likely explanation to lower body weight seen in this group (Paper 1).

Page 59: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

59

6.1.2 TAG-reducing effect of SPH

Fish oil has been reported to reduce plasma TAG levels [230], whereas fish protein

lowers cholesterol levels in rats [76, 231-233]. Also, fish protein has been reported to

decrease cholesterol content of LDL particle as compared to casein [234]. However,

in our study in mice we detected plasma TAG-reducing effect in two of the peptide

fractions (E1 and E4) compared to casein, but unchanged plasma cholesterol levels

(Paper 1). The TAG-reducing action of fish oil is attributed to the omega-3 fatty

acids as they are natural PPAR agonists, thus contributing to increased fatty acid

oxidation. However, studies indicate that fish oil lowers TAG through both PPAR-

dependent and independent pathways [235]. It has been proposed that fish oil also

decreases the transcription factor SREBPs, thus down-regulating mRNA expression

of lipogenic genes [236]. In line with studies on n-3 PUFAs [237, 238], we detected

reduced enzyme activity of FAS and decreased gene expression of Fasn and Acaca,

involved in fatty acid synthesis, using fish peptides (Paper 1). Previous studies in rats

have shown reduced plasma cholesterol concentrations connected to elevated plasma

bile acid levels [215], and an increase in both cholesterol clearance through bile and

bile excretion [239]. Despite the decrease in gene expression of Hmgcr in mice fed

the E1 and E4 diets, no reduction in plasma cholesterol levels was observed. This

could be related to the lower dose of SPH, or the animal model. Elevated plasma bile

acid level was detected in group fed the E4 diet (Paper 1). Unfortunately, cholesterol

and bile excretion through faeces were not measured in our study. In contrast, peptide

group E2 displayed increased hepatic TAG, as well as plasma TAG, cholesterol and

phospholipid levels, combined with increased FAS activity and elevated gene

expression of Scd1, indicating stimulated fatty acid synthesis in this group (Paper 1).

Differences in liver and plasma TAG could be explained by opposite regulation of

fatty acid synthesis between the peptide groups.

6.1.3 Various protein hydrolysates may influence fatty acid composition

Bioavailability of a protein is assumed to be varying according to protein source,

some are expected to suppress appetite [55], others increase satiety [56]. Feed intake

Page 60: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

60

in our four groups could not explain the divergent effect on body weight, as protein

unequally affect body weight gain independent of feed intake [240]. The variation in

body weight between our treatment groups could thus be explained by distinct

peptide or amino acid profile of each protein hydrolysate fraction (Paper 1). Dietary

protein may influence fatty acid composition and lipogenesis, and desaturation have

been reported to be affected by the aromatic amino acids tyrosine and phenylalanine

[241]. Elimination of tyrosine and phenylalanine from amino acid mixtures, has

shown increased Δ6 activity [241]. Here, the gene expression of Fads2 was

comparable between all groups since they were all dominated by amino acids from

casein. Arginine and leucine have been suggested to regulate gene expression of Scd1

and Fasn [242]. Arginine levels in E1 and E4 groups were lower compared to the E2

group, and fatty acid pattern was similar in E1 and E4, as well as an increase in

18:3n-6/18:2n-6 and 20:4n-6/20:3n-6 ratios in the E4-fed group compared to control.

These fatty acid ratios are often used as an index for the activities of Δ6 and Δ5

desaturases, respectively. As these indices are affected by several pathways [243],

they did not correspond with the gene expression of Fads1 and Fads2 (Paper 1). In

contrast, the unaltered Scd1 expression reflected with the Δ9 desaturase index in

liver. In rats fed the peptide fraction E2 a simultaneously enhanced liver Scd1

expression, increase in Δ9 desaturase index and elevated MUFAs was detected,

further supporting an enhanced fatty acid synthesis in this group. Oleoyl-CoA

(C18:1) is the main product of Δ9 desaturase and is primarily used for TAG

generation, and this was reflected in an increase in short-chained MUFAs. This could

suggest that increase in Scd1 and a corresponding increase in incorporation of

MUFAs into TAG could lead to an increase in VLDL release. In two of the peptide

fraction groups (E1 and E4) MUFAs decreased whereas PUFA increased, thus

favouring PUFA synthesis instead of MUFAs, suggesting that different peptide

effects on lipid levels could have been reinforced by their opposite regulation of fatty

acids influencing cell signaling.

Page 61: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

61

6.1.4 Elevated liver ceramide levels did not affect fatty acid catabolism

Ceramides are a subgroup of waxy lipids consisting of sphingosine and a fatty acid

present in the cell membrane. In addition to their physiological role in cell

membranes including adhesion, differentiation and cell senescence, ceramides and

their metabolites are also implemented in pathological states like obesity, diabetes

and inflammation. Elevated liver LCFAs are suggested to contribute to adverse effect

of ceramides on insulin and inflammation, in addition to characterize human subjects

with high liver fat content [244]. However, despite elevated ceramide levels in

C57BL/6J mice livers in the peptide E1 group, we detected lower hepatic TAG level

in this group (Paper 1). Accumulation of ceramides leads to an alteration in energy

metabolism slowing anabolism to ensure that catabolism ensues [245], actions

associated with obesity. We did not find an increase in fatty acid oxidation in this

group, suggesting that, at least fatty acid catabolism was not affected by elevated

ceramide levels (Paper 1). As there was no indication of fatty liver and hepatic

expression of Scd1 and lipogenic activity was unchanged, the elevated ceramide

levels observed in the peptide E1 group might be due to the decrease in hepatic TAG.

TAG species are the most prevalent lipid class in the liver, thus it is reasonable to

assume that a decrease in these levels could affect percentage of other lipid classes,

like ceramides, hence an increase in ceramide levels. On the contrary, in the peptide

E2 group there was a general decrease in ceramides. The increase in Scd1 and FAS

activity in the peptide E2 group, with a simultaneously increase in DAG, but not

ceramide concentrations, could indicate early onset of non-alcoholic fatty liver

disease (NAFLD) [246]. Liver TAG levels in this group were also significantly

increased, further supporting the possibility of early onset of fatty liver. The decrease

in DAG in the E1 group could be of importance in regulating PK-K, modulating cell

processes.

In this study we found diverse effects of three peptide fractions from salmon protein

hydrolysates treated with various proteolytic enzymes on body weight, plasma and

liver lipids and synthesis, indicating that different peptide fractions can have potential

Page 62: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

62

for both health-promoting and adverse effects (Paper 1). Digestion of protein leads to

products that can interfere with intestinal absorption and pre-hydrolyzation of protein

is shown to exert a more protruding effect than whole protein [247-249], and gives

the opportunity to optimize nutritional value and bioavailability, resulting in a

specified product regarding its use.

6.1.5 SPH reduced atherosclerotic plaque area in apoE-/- mice

The initiation of CVD is progression of atherosclerotic lesions; whereas the onset of

atherosclerotic development is controversial. Whether, high circulating levels of

inflammatory cytokines represent the degree of atherosclerosis or simply an active

immune system is speculative. There is generally an agreement about interplay

between inflammation and lipid storage, which comes first however, remains unclear

[250-253]. The absent of apoE on chylomicrons and other lipoproteins, cause

accumulation of VLDL remnants in plasma, which contributes to atherosclerotic

development. ApoE-/- knockout mice are widely used in this regard and this animal

model develop atherosclerotic lesions on a standard chow diet. After testing different

SPH-fractions in C57BL/6J mice (Paper 1), accompanied by promising results on

TAG-reducing effects in plasma, in addition to indications of reduced fatty acid

synthesis, the E1 fraction was further evaluated in apoE-/- mice (Paper 2). After 12

weeks, we found reduced atherosclerotic plaque area in aortic sinus as well as in

aortic arch compared to the casein diet (Paper 2). In addition, gene expression of the

atherosclerotic marker in pooled aortic arch, Icam1 was decreased, purposing a local

anti-atherosclerotic potential of SPH (Paper 2).

Metabolites of arachidonic acid (20:4n-6), like prostaglandins and leukotrienes, are

thought to be proatherogenic [254, 255], whereas oleic acid are considered health

beneficial by lowering cholesterol levels and improving insulin sensitivity [256].

Oleic acid (18:1n-9) is proposed to counteract mechanisms of arachidonic acid by

reducing its serum level [256]. Arachidonic acid inhibits Δ9 desaturase, which can

explain the lower level of oleic acid (Paper 2). Additionally, we detected reduced

liver mRNA expression of Δ9 desaturase (Scd1), further contributing to lower level of

Page 63: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

63

oleic acid. However, oleic acid, via linoleic acid, is the precursor of arachidonic acid,

thus an increase in arachidonic could be at the expense of oleic acid. Despite an

increase and decrease in the proatherogenic arachidonic acid and health-promoting

oleic acid, respectively, plaque area decreased, indicating that SPH has the potential

to attenuate atherosclerosis independently of plasma fatty acids (Paper 2).

6.1.6 Plaque stability was unaffected after SPH-treatment

The common feature of acute manifestations of atherosclerosis is rupture of the

plaque, which causes heart attacks or stroke. Plaques with a thin fibrous cap tend to

expend as more lipid accumulate and macrophages release enzymes destroying

collagen resulting in a weak, rupture-prone fibrous cap. As the stability of the plaque

is an important factor contributing to the severity of atherosclerosis, the content of the

atherosclerotic plaque was assessed by immunohistochemistry. However, there was

no change in connective tissue, macrophages or lymphocytes, indicating that plaque

stability was not affected by SPH administration (Paper 2). Spontaneous plaque

rupture and subsequent formation of thrombosis in murine models have yet to be

shown and even when the fibrous cap is extremely thin, it does not rupture [257].

Although, the apoE-/- mouse model is a suitable and extensively used model for

studying atherosclerosis, it is not appropriate for investigate susceptibility of the

plaque to rupture [258]. However, another explanation could be a simultaneously

decrease in macrophages and plaque area, thus no change in a percentage

measurement. Decreased sinus plaque area combined with unchanged macrophage

number could be related to a smaller lipid-rich core, hence a reduction in macrophage

size. Another likely explanation may be related to reduced proliferation of VSMC, as

the majority of foam cells could be derived from VSMC [259].

6.1.7 SPH decreased systemic inflammation

Key markers of inflammation and early stage of atherosclerosis are IL-1β, TNF-α and

IL-6, which all were reduced after SPH treatment, in addition to significantly lower

levels of IL-10 and GM-CSF, suggesting that systemic inflammation was reduced

(Paper 2). Previous studies have shown reduced colon cytokine levels in a rat model

Page 64: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

64

of colitis [260], and lower levels of interferon gamma (IFN-γ), in addition to

increased anti-inflammatory index in an inflammatory mouse model [229], after

intervention with the same SPH fraction, further supporting the anti-inflammatory

potential of SPH.

It is generally assumed that high circulating levels of cholesterol and TAG increases

the risk of atherosclerosis, and fish protein display a cholesterol-lowering effect

[233]. In line with our previously results (Paper 1) and by others [229, 260], we

observed no change in plasma cholesterol (Paper 2), however as opposed to our

forgoing findings (Paper 1), plasma TAG levels were unchanged (Paper 2). Activity

of enzymes involved in peroxisomal and mitochondrial fatty acid oxidation, acyl-

coenzyme A oxidase (ACOX)-1, palmitate-CoA production, CPT2, and cholesteryl

ester synthesis, were unchanged, accompanied by unchanged gene expression of the

corresponding genes (Paper 2), thus fatty acid oxidation was not affected by SPH

administration, in agreement with the unchanged plasma TAG level. This could be

explained by the low dose of SPH, as studies showing reduced cholesterol and TAG

levels, have utilized higher doses [76, 215, 233]. However, in our previous study in

wild type mice (Paper 1), reduced liver and plasma levels of TAG were reported.

Thus, persistent lipoprotein disturbance due to the apoE-knockout could be adequate

to interfere, or inhibit a potential effect of SPH on lipogenesis compared to wild type

mice. Reduced hepatic gene expression of Acaca and Scd1, key enzymes in fatty acid

synthesis, was inadequate to significantly reduce plasma and liver TAG levels of SPH

treated mice. SPH administration displayed no effect on the antioxidant defence

system in the heart of apoE-/- mice, as gene expression of the antioxidants superoxide

dismutase (Sod1), Catalase and Nos2 were unaffected (Paper 2). In combination

with the reduced plasma levels of inflammatory markers, these results indicate that

SPH protects against atherosclerotic development by inhibiting inflammation,

independent of plasma lipid changes.

Page 65: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

65

6.2 CP as a protein source

Protein from animal sources such as milk and meat from cattle is considered less

health-promoting than leaner meat from i.e. poultry, due to high content of fat.

Increased protein intake is recommended to keep satiety for longer period in an

attempt to reduce energy intake, as protein being the macronutrient requiring most

comprehensive processes in digestion and absorption.

Proteins from plant origin like soy and animal origin like casein, is widely studied,

surpassing others like chicken. Chicken is an abundant supply all over the world and

is considered a well-balanced protein source, rich in essential amino acids. The large

consumption of chicken combined with its nutritional value, makes it interesting to

elucidate potential mechanisms CP may possess regarding improvement of human

health. Previously, CP was shown to have antioxidant [261] and antihypertensive

[262, 263] capacities. Therefore, in Paper 3 we wanted to determine a potential effect

of CP on lipid metabolism, as proteins have been suggested to influence parameters

involved in inflammation.

6.2.1 CP reduced body weight and lipogenesis in rats

After 4 weeks of dietary treatment with a water-soluble extract of CP at three

different doses, we found lower body weight, despite a higher feed intake, in rats fed

CP compared to protein from casein (Paper 3). This result was most prominent in the

group receiving the highest dose of CP, the 20% CP group (Paper 3). There are

several possible explanations for this finding, one of them being poorer digestibility

of the CP. Dietary proteins have been suggested to interrupt with digestion forming

complexes difficult to absorb and proposal that peptides have a higher affinity

towards bile acids lowering micelle solubility [239]. This could decrease fat

absorption, hence reduce energy uptake contributing to lower weight gain. Peptides

are known to affect lipid metabolism, probably through PPAR [171], and an up-

regulation of Ppara gene expression can explain the increase in mitochondrial fatty

acid oxidation we observed, accompanied by reduced plasma and liver TAG content

(Paper 3). The increase in fatty acid oxidation can also partly account for the weight

Page 66: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

66

gain, as well as the reduced plasma and liver levels of TAG. Also, activity of the

glycerolipid synthesizing enzyme GPAT was decreased further contributing to lower

TAG levels (Paper 3). Enzymes involved in fatty acid synthesis like the rate-limiting

enzyme ACC and FAS decreased at all three doses, but only significant in the 20%

group (Paper 3). Gene expressions of the corresponding genes, Acaca and Fasn,

were also decreased, indicating a reduced fatty acid synthesis, which also could have

contributed to the lower levels of plasma and liver TAG (Paper 3). Gene expression

of the transcription factor Srebf1, involved in sterol biosynthesis, was significantly

lower in the 20% CP fed group probably contributing to the lipogenic-suppressing

effect of CP. However, gene expression of Srepf2, Hmgcs1 and Hmgcr, all involved

in cholesterol synthesis, was unaltered, thus the lower plasma and liver cholesterol

levels could be due to post-transcriptional mechanisms (Paper 3).

6.2.2 Elevated plasma level of bile acids after CP intervention

Plasma bile acids and gene expression of the bile transporting Abcb4 was

significantly increased, which may indicate increase transportation of bile,

accompanied by the possibility of cholesterol loss through increased bile excretion

(Paper 3). The present amount of the amino acids taurine and glycine were

substantial in the CP diet, and previously findings with taurine and glycine rich diets

fed to rats, have shown increased level of plasma bile acids and enhanced excretion

of bile acids through faeces [215].

There was a change in liver fatty acid composition, and increase in α-linoleic acid and

docosapentaenoic acid (DPA) levels were observed, whereas eicosapentaenoic acid

(EPA) decreased (Paper 3). However, DPA has shown bioactivity by influencing

endothelial cell migration, hence better wound-healing [264] and reducing FAS

activity [265]. We observed an increased level of LCFAs compared to SCFAs, which

can be a consequence of an unchanged peroxisomal oxidation of LCFA and a

preferential increased mitochondrial oxidation of SCFAs and MCFAs (Paper 3). The

tendency towards decreased content of MUFAs and decreased C16:1n7/16:0 ratio

could be related to decreased in gene expression of Scd1 (Paper 3).

Page 67: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

67

6.2.3 Cellular toxicity could be reduced

Carnitine is directly involved in fatty acid metabolism by transporting fatty acid

across the two mitochondrial membranes, thus free carnitine, its precursor and

metabolites were measured. The three CP diets demonstrated a clear dose-response

effect of CP on all carnitine parameters measured (Paper 3). Free carnitine increased

significantly at all three doses. The precursor of carnitine, γ-butyrobetaine, was also

increased, as were short-, medium- and long-chained acylcarnitines suggesting

reduced cellular toxicity as acyl-CoA derivatives causes mitochondrial swelling and

cellular toxicity [266], thus increased carnitine levels could improve mitochondrial

capacity as potentially toxic fatty acyl-derivatives may be removed.

The increased β-oxidation and bile formation, and decreased lipogenesis could be

related to the amino acid composition of CP compared to casein. The 20% CP diet

was characterized by low ratios of methionine/glycine and lysine/arginine which have

been suggested to exert cholesterol-reducing [95, 104, 267] and TAG -lowering

effects [268]. Lower ratios of both methionine/glycine and lysine/arginine were also

detected in plasma of the 20% CP-fed rats (Paper 3). Although the content of some

amino acids in the CP diet were lower compared to the casein diet, we did not suspect

amino acid deficiency in CP-treated rats as amino acid deprived rats would have

shown hypercholesterolemia [269], fatty liver and hair loss [270]. In addition, alanine

transaminase (ALT) and aspartate aminotransferase (AST) was unchanged further

supporting a healthy liver. The mechanisms of action behind the possible capacity of

amino acids to influence cholesterol and TAG levels, is unknown and should be

further investigated in future studies.

6.2.4 CP showed no anti-atherosclerotic effect in apoE-/- mice

As elevated plasma levels of cholesterol and TAG are strongly linked to CVD-risk

factors and a water-soluble extract of chicken CP was shown to reduce plasma and

liver lipids, it was of particular interest to investigate the CP extract in the

atheroprone mice model, apoE-/- mice to see if CP could affect lipid metabolism

(Paper 4). In addition, we wanted to see if CP could attenuate atherosclerotic

Page 68: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

68

development in accordance with the SPH-diet (Paper 2). Similar to our previous

study (Paper 3), CP exerted plasma TAG-lowering effect; however, the reduction

was relatively small, accompanied by a presumably increase in mitochondrial β-

oxidation as enzyme activity and gene expression of CPT2 was elevated (Paper 4).

The indication of increased mitochondrial fatty acid oxidation could be associated

with our observation of decreased RER after monitoring the mice in metabolic cages.

Also, the lower plasma TAG level could be related to RER as a decrease in RER is

linked to a higher energy contribution from fat. This is also in line with the lower

body weight despite a higher feed intake in CP-fed mice. Noteworthy, apoE-/- mice

are not suitable for studying body weight due to the knockout of a crucial gene

involved in TAG transportation. SPH treatment in apoE-/- mice displayed reduced

atherosclerotic plaque combined with a decreased systemic inflammatory state

(Paper 2). Unlike our previous results, we observed no effect on atherosclerotic

plaque development after CP-intervention in apoE-/- mice (Paper 4). This was further

supported by unaffected heart mRNA levels of the adhesion molecules Icam1 and

Vcam1, the antioxidant Nos2 and the chemoattractant protein, Mcp1, in addition to

unaltered plasma concentrations of proinflammatory cytokines. In our previous study

with CP, we detected plasma and liver cholesterol-reducing effects (Paper 3),

whereas CP administration had no effect on the cholesterol level in apoE-/- mice.

Although rats were fed a low-fat diet compared to high-fat diet-feeding in apoE-/-

mice, could have influenced our results, it is more likely the different rodent models.

Knockout of apoE results in accumulation of TAG-rich VLDL in plasma, however,

due to the extended residence time and a co-occurrence of lipolysis, plasma VLDLs

in apoE-/- could be TAG-deprived and result in a compensating increase of

cholesterol. In contrast to CP-fed Wistar rats, lipogenesis was unaffected in apoE-/-

mice after CP intervention. Further, there was no effect on plasma TAG levels after

SPH treatment in apoE-/- mice (Paper 2), although there was TAG-reducing effect of

SPH in wild type mice (Paper 1). Thus, it seem neither SPH nor CP were able to

influence plasma TAG levels in apoE-/- mice, probably due to the persistent disturbed

lipid metabolism in this mouse model.

Page 69: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

69

Our studies suggest that CP have minor potential to offset the negative effect of apoE

knockout and does not have the ability to affect plaque development or reduce

circulating inflammatory markers compared to SPH in this mouse model.

Noteworthy, the hydrolyzing process of SPH was much more comprehensive than the

one producing the water-soluble protein extract of chicken. Protein hydrolysates are

assumed to have more potent health-promoting effects, which is attributed bioactive

peptides hidden within the protein structure. The hydrolyzing process makes these

peptides more accessible, increasing their bioavailability. Thus, it could be that CP

possesses lipid-lowering and anti-atherosclerotic effects if the extract were to be

hydrolyzed. Also, it could be that the knockout of apoE suppresses the effect of CP.

The small reduction in plasma TAG levels and unchanged cholesterol levels was not

sufficient to influence lesion development. Although, the TAG reduction was small,

this may further supporting our speculations from our previous study in apoE-/- mice;

that plaque development is independent of changes in plasma lipids and could be

more closely related to inflammation.

Page 70: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

70

7. CONCLUSIONS

This thesis supports the assumption that various protein sources possess different

bioactive potential. In accordance with the aims of the thesis, the following

conclusions can be drawn:

Hydrolyzing methods using different enzymes influence the potential bioactivity of a

salmon protein source. Three different pre-digested fractions of SPH displayed

different effects on lipogenesis as well as liver and plasma TAG levels in male

C57BL/6 mice. The findings show that SPH can possess bioactive peptides with

diverse capability of affecting gene expression and activity of enzymes regulating

fatty acid metabolism, thus affecting plasma and liver lipid levels.

In female apoE-/- mice SPH displayed an ability to attenuate atherosclerosis by

reducing lesion area in the aortic sinus and arch. Systemic inflammation was reduced,

while the effects on fatty acid oxidation and plasma lipid levels were minor. This

indicates that systemic anti-inflammatory effects are crucial in attenuating

atherosclerosis.

CP revealed distinct dose-dependent effects on energy metabolism in male Wistar

rats by increased fatty acid catabolism, lower cholesterol levels and possible

increased excretion of bile. Although effects on weight gain complicate the

interpretation of these results, they could be attributed the distinct amino acid profile

of the CP.

CP showed a small effect on fatty acid oxidation in apoE-/- mice, but did not influence

systemic or local inflammation or plaque area. Thus, despite a lipid-lowering effect in

rats, CP seems to be less potent in counteracting disrupted lipid metabolism in apoE-/-

mice, and had no anti-inflammatory effect. This could explain why CP was not able

to affect atherosclerotic development.

Page 71: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

71

Notably, the substantially less hydrolysis degree of the CP compared to SPH could

explain the different capacity of these protein sources and indicates the potential in

hydrolyzing proteins to liberate bioactive peptides.

In addition, it appears that plaque development is more related to inflammatory state

than changes in plasma lipid levels in the apoE-/- mouse model.

Page 72: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

72

8. FUTURE PERSPECTIVES

These studies have provided insight into protein and protein hydrolysate effect on

lipid metabolism and inflammation. The difference in anti-atherogenic potential

between the 70% hydrolyzed SPH, in contrast to 30% hydrolysis in the CP, indicates

that a more extensive hydrolyzation of the CP should be considered in future

experiments.

Another atherosclerotic mice model for studying the effect of CP should be

considered.

Mitochondrial oxidative stress is strongly linked to atherosclerosis and based on our

studies it appears that inflammation has a prominent role on plaque development

compared to high plasma lipid levels. Thus markers and mediators of inflammation

both locally and in circulation should be further investigated. Oxidative stress in

mitochondria and inflammation in adipose tissue could be of interest.

After protein hydrolyzation, the peptide amino acid sequences should be determined,

to get a better survey of potential bioactive peptides. However, this is an expensive

process and should only be considered when identification and purification of

bioactive peptides is relevant.

Page 73: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

73

9. REFERENCES 1. World Health Organization - Obesity and Overweight. 2013 [cited 2014 15th of March]; Available

from: http://www.who.int/mediacentre/factsheets/fs311/en/. 2. Grundy, S.M., Brewer, H.B., Jr., Cleeman, J.I., Smith, S.C., Jr., and Lenfant, C., Definition of

metabolic syndrome: report of the National Heart, Lung, and Blood Institute/American Heart Association conference on scientific issues related to definition. Arteriosclerosis, Thrombosis and Vascular Biology, 2004. 24(2): p. e13-18.

3. Blaha, M. and Elasy, T.A., Clinical Use of the Metabolic Syndrome: Why the Confusion? Clinical Diabetes, 2006. 24: p. 125-131.

4. Kostoglou-Athanassiou and P, A., Metabolic syndrome and sleep apnea. Hippokratia, 2008. 12: p. 81-86.

5. Kahn, R., Buse, J., Ferrannini, E., and Stern, M., The metabolic syndrome: time for a critical appraisal: joint statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care, 2005. 28(9): p. 2289-2304.

6. Carr, M.C. and Brunzell, J.D., Abdominal obesity and dyslipidemia in the metabolic syndrome: importance of type 2 diabetes and familial combined hyperlipidemia in coronary artery disease risk. Journal of Clinical Endocrinology and Metabolism, 2004. 89(6): p. 2601-2607.

7. Garcia-Lara, J.M., Aguilar-Navarro, S., Gutierrez-Robledo, L.M., and Avila-Funes, J.A., The metabolic syndrome, diabetes, and Alzheimer's disease. Revista de Investigación Clínica, 2010. 62(4): p. 343-349.

8. Razay, G., Vreugdenhil, A., and Wilcock, G., The metabolic syndrome and Alzheimer disease. Archives of Neurology, 2007. 64(1): p. 93-96.

9. Castro, J.P., El-Atat, F.A., McFarlane, S.I., Aneja, A., and Sowers, J.R., Cardiometabolic syndrome: pathophysiology and treatment. Current hypertension reports, 2003. 5(5): p. 393-401.

10. Vamecq, J., Dessein, A.F., Fontaine, M., Briand, G., Porchet, N., Latruffe, N., Andreolotti, P., and Cherkaoui-Malki, M., Mitochondrial dysfunction and lipid homeostasis. Current Drug Metabolism, 2012. 13(10): p. 1388-1400.

11. Johannsen, D.L. and Ravussin, E., The role of mitochondria in health and disease. Current Opinion in Pharmacology, 2009. 9(6): p. 780-786.

12. Bratic, I. and Trifunovic, A., Mitochondrial energy metabolism and ageing. Biochimica et Biophysica Acta, 2010. 1797(6-7): p. 961-967.

13. Liesa, M. and Shirihai, O.S., Mitochondrial dynamics in the regulation of nutrient utilization and energy expenditure. Cell Metabolism, 2013. 17(4): p. 491-506.

14. Kim, J.A., Wei, Y., and Sowers, J.R., Role of mitochondrial dysfunction in insulin resistance. Circulation Research, 2008. 102(4): p. 401-414.

15. Gutierrez, J., Ballinger, S.W., Darley-Usmar, V.M., and Landar, A., Free radicals, mitochondria, and oxidized lipids: the emerging role in signal transduction in vascular cells. Circulation Research, 2006. 99(9): p. 924-932.

16. Hojlund, K., Mogensen, M., Sahlin, K., and Beck-Nielsen, H., Mitochondrial dysfunction in type 2 diabetes and obesity. Endocrinology Metabolism Clinics of North America, 2008. 37(3): p. 713-731.

17. Madamanchi, N.R. and Runge, M.S., Mitochondrial dysfunction in atherosclerosis. Circulation Research, 2007. 100(4): p. 460-473.

18. Wilms, L., Larsen, J., Pedersen, P.L., and Kvetny, J., Evidence of mitochondrial dysfunction in obese adolescents. Acta Paediatrica, 2010. 99(6): p. 906-911.

19. Yu, E., Mercer, J., and Bennett, M., Mitochondria in vascular disease. Cardiovascular Research, 2012. 95(2): p. 173-182.

20. Valcárcel-Ares, M.N., Vaamonde-García, C., Riveiro-Naveira, R.R., Lema, B., Blanco, F.J., and López-Armada, M.J., A novel role for mitochondrial dysfunction in the inflammatory response of rheumatoid arthritis. Annals of the Rheumatic Disease, 2010. 69: p. A56.

21. Mercer, J.R., Cheng, K.-K., Figg, N., Gorenne, I., Mahmoudi, M., Griffin, J., Vidal-Pui, A., Logan, A., and P, M., DNA damage links mitochondrial dysfunction to atherosclerosis and the metabolic syndrome. Circulation Research, 2010. 107(8): p. 1021-1031.

22. Escames, G., Lopez, L.C., Garcia, J.A., Garcia-Corzo, L., Ortiz, F., and Acuna-Castroviejo, D., Mitochondrial DNA and inflammatory diseases. Human Genetics, 2012. 131(2): p. 161-173.

23. Racette, S.B., Deusinger, S.S., and Deusinger, R.H., Obesity: overview of prevalence, etiology, and treatment. Physical Therapy, 2003. 83(3): p. 276-288.

Page 74: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

74

24. Trayhurn, P., Bing, C., and Wood, I.S., Adipose tissue and adipokines - energy regulation from the human perspective. Journal of Nutrition, 2006. 136(7 Suppl): p. 1935S-1939S.

25. Wisse, B.E., The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. Journal of the American Society of Nephrology, 2004. 15(11): p. 2792-2800.

26. Harwood, H.J., Jr., The adipocyte as an endocrine organ in the regulation of metabolic homeostasis. Neuropharmacology, 2012. 63(1): p. 57-75.

27. Hajer, G.R., van Haeften, T.W., and Visseren, F.L., Adipose tissue dysfunction in obesity, diabetes, and vascular diseases. European Heart Journal, 2008. 29(24): p. 2959-2971.

28. Kalupahana, N.S., Claycombe, K.J., and Moustaid-Moussa, N., (n-3) Fatty acids alleviate adipose tissue inflammation and insulin resistance: mechanistic insights. Advances in Nutrition, 2011. 2(4): p. 304-316.

29. Mowers, J., Uhm, M., Reilly, S.M., Simon, J., Leto, D., Chiang, S.H., Chang, L., and Saltiel, A.R., Inflammation produces catecholamine resistance in obesity via activation of PDE3B by the protein kinases IKK{varepsilon} and TBK1. Elife, 2013. 2: p. e01119.

30. Aguirre, V., Uchida, T., Yenush, L., Davis, R., and White, M.F., The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser(307). Journal of Biological Chemistry, 2000. 275(12): p. 9047-9054.

31. Engelman, J.A., Berg, A.H., Lewis, R.Y., Lisanti, M.P., and Scherer, P.E., Tumor necrosis factor alpha-mediated insulin resistance, but not dedifferentiation, is abrogated by MEK1/2 inhibitors in 3T3-L1 adipocytes. Molecular Endocrinology, 2000. 14(10): p. 1557-1569.

32. Liu, Y.F., Paz, K., Herschkovitz, A., Alt, A., Tennenbaum, T., Sampson, S.R., Ohba, M., Kuroki, T., LeRoith, D., and Zick, Y., Insulin stimulates PKCzeta -mediated phosphorylation of insulin receptor substrate-1 (IRS-1). A self-attenuated mechanism to negatively regulate the function of IRS proteins. Journal of Biological Chemistry, 2001. 276(17): p. 14459-14465.

33. Haruta, T., Uno, T., Kawahara, J., Takano, A., Egawa, K., Sharma, P.M., Olefsky, J.M., and Kobayashi, M., A rapamycin-sensitive pathway down-regulates insulin signaling via phosphorylation and proteasomal degradation of insulin receptor substrate-1. Molecular Endocrinology, 2000. 14(6): p. 783-794.

34. Aguirre, V., Werner, E.D., Giraud, J., Lee, Y.H., Shoelson, S.E., and White, M.F., Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action. Journal of Biological Chemistry, 2002. 277(2): p. 1531-1537.

35. De Fea, K. and Roth, R.A., Modulation of insulin receptor substrate-1 tyrosine phosphorylation and function by mitogen-activated protein kinase. Journal of Biological Chemistry, 1997. 272(50): p. 31400-31406.

36. Gao, Z., Hwang, D., Bataille, F., Lefevre, M., York, D., Quon, M.J., and Ye, J., Serine phosphorylation of insulin receptor substrate 1 by inhibitor kappa B kinase complex. Journal of Biological Chemistry, 2002. 277(50): p. 48115-48121.

37. Libby, P., Inflammation and cardiovascular disease mechanisms. American Journal of Clinical Nutrition, 2006. 83(2): p. 456S-460S.

38. Pearson, T.A., Mensah, G.A., Alexander, R.W., Anderson, J.L., III, R.O.C., Criqui, M., Fadl, Y.Y., Fortmann, S.P., Hong, Y., Myers, G.L., Rifai, N., Jr, S.C.S., Taubert, K., Tracy, R.P., and Vinicor, F., Markers of Inflammation and Cardiovascular Disease. Circulation, 2003. 107: p. 499-511.

39. Willerson, J.T. and Ridker, P.M., Inflammation as a Cardiovascular Risk Factor. Circulation, 2004. 109: p. II-2-II-10.

40. Endemann, D.H. and Schiffrin, E.L., Endothelial dysfunction. Journal of the American Society of Nephrology, 2004. 15(8): p. 1983-1992.

41. Abraham, D. and Distler, O., How does endothelial cell injury start? The role of endothelin in systemic sclerosis. Arthritits Research & Therapy, 2007. 9: p. S2.

42. Albelda, S., Smith, C., and Ward, P., Adhesion molecules and inflammatory injury. Journal of the Federation of American Societies for Experimental Biology, 1994. 8: p. 504-512.

43. Alexander, R.W., Theodore Cooper Memorial Lecture. Hypertension and the pathogenesis of atherosclerosis. Oxidative stress and the mediation of arterial inflammatory response: a new perspective. Hypertension, 1995. 25(2): p. 155-161.

44. McLenachan, J.M., Vita, J., Fish, D.R., Treasure, C.B., Cox, D.A., Ganz, P., and Selwyn, A.P., Early evidence of endothelial vasodilator dysfunction at coronary branch points. Circulation, 1990. 82(4): p. 1169-1173.

45. Petecchia, L., Sabatini, F., Usai, C., Caci, E., Varesio, L., and Rossi, G.A., Cytokines induce tight junction disassembly in airway cells via an EGFR-dependent MAPK/ERK1/2-pathway. Laboratory Investigation, 2012. 92: p. 1140–1148.

Page 75: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

75

46. Dhaliwal, B.S. and Steinbrecher, U.P., Scavenger receptors and oxidized low density lipoproteins. Clinica Chimica Acta, 1999. 286(1-2): p. 191-205.

47. Trpkovic, A., Resanovic, I., Stanimirovic, J., Radak, D., Mousa, S.A., Cenic-Milosevic, D., Jevremovic, D., and Isenovic, E.R., Oxidized low-density lipoprotein as a biomarker of cardiovascular diseases. Critical reviews in clinical laboratory sciences, 2014: p. 1-16.

48. Johnson, J.L., Emerging regulators of vascular smooth muscle cell function in the development and progression of atherosclerosis. Cardiovasc Res, 2014. 103(4): p. 452-460.

49. Viles-Gonzalez, J.F., Fuster, V., and Badimon, J.J., Atherothrombosis: a widespread disease with unpredictable and life-threatening consequences. European Heart Journal, 2004. 25(14): p. 1197-1207.

50. Wildgruber, M., Swirski, F.K., and Zernecke, A., Molecular Imaging of Inflammation in Atherosclerosis. Theranostics, 2013. 3(11): p. 865-884.

51. van der Wal, A.C. and Becker, A.E., Atherosclerotic plaque rupture--pathologic basis of plaque stability and instability. Cardiovascular Research, 1999. 41(2): p. 334-344.

52. Schoenhagen, P., Ziada, K.M., Vince, D.G., Nissen, S.E., and Tuzcu, E.M., Arterial remodeling and coronary artery disease: the concept of “dilated” versus “obstructive” coronary atherosclerosi. Journal of the American College of Cardiology, 2001. 38(2).

53. Libby, P., Multiple mechanisms of thrombosis complicating atherosclerotic plaques. Clinical Cardiology, 2000. 23 Suppl 6: p. VI-3-7.

54. Rentrop, K.P., Thrombi in acute coronary syndromes : revisited and revised. Circulation, 2000. 101(13): p. 1619-1626.

55. Nishi, T., Hara, H., Asano, K., and Tomita, F., The soybean beta-conglycinin beta 51-63 fragment suppresses appetite by stimulating cholecystokinin release in rats. Journal of Nutrition, 2003. 133(8): p. 2537-2542.

56. Uhe, A.M., Collier, G.R., and O'Dea, K., A comparison of the effects of beef, chicken and fish protein on satiety and amino acid profiles in lean male subjects. Journal of Nutrition, 1992. 122(3): p. 467-472.

57. Anderson, G.H., Metabolic regulation of food intake. In: (Shils, M. E. & Young, V. R., eds.), ed. 7th. 1989, Philadelphia, PA.: Lea and Febiger.

58. Anderson, G.H. and Li, E.T., Protein and amino acids in the regulation of quantitative and qualitative aspects of food intake. International journal of obesity, 1987. 11 Suppl 3: p. 97-108.

59. Femstrom, J.D., Wurtman, R.J., Hammarstrom-Wiklund, B., Rand, W.M., Munro, N., and Davidson, C.S., Diurnal variations in plasma concentrations of tryptophan, tyrosine, and other neutral amino acids: effect of dietary protein intake. American Journal of Clinical Nutrition, 1979. 32: p. 1912-1922.

60. Gendall, K.A. and Joyce, P.R., Meal-induced changes in tryptophan:LNAA ratio: effects on craving and binge eating. Eating behaviors, 2000. 1(1): p. 53-62.

61. Jump, D.B., Fatty acid regulation of gene transcription. Critical reviews in clinical laboratory sciences, 2004. 41(1): p. 41-78.

62. Hulbert, A.J., Turner, N., Storlien, L.H., and Else, P.L., Dietary fats and membrane function: implications for metabolism and disease. Biological Reviews of the Cambridge Philosophical Society, 2005. 80(1): p. 155-169.

63. Jensen, M.D., Diet effects on fatty acid metabolism in lean and obese humans. American Journal of Clinical Nutrition, 1998. 67(3 Suppl): p. 531S-534S.

64. Jurgonski, A., Juskiewicz, J., and Zdunczyk, Z., A high-fat diet differentially affects the gut metabolism and blood lipids of rats depending on the type of dietary fat and carbohydrate. Nutrients, 2014. 6(2): p. 616-626.

65. Schwarz, J., Tome, D., Baars, A., Hooiveld, G.J., and Muller, M., Dietary protein affects gene expression and prevents lipid accumulation in the liver in mice. PLoS One, 2012. 7(10): p. e47303.

66. Tovar, A.R., Murguia, F., Cruz, C., Hernandez-Pando, R., Aguilar-Salinas, C.A., Pedraza-Chaverri, J., Correa-Rotter, R., and Torres, N., A soy protein diet alters hepatic lipid metabolism gene expression and reduces serum lipids and renal fibrogenic cytokines in rats with chronic nephrotic syndrome. Journal of Nutrition, 2002. 132(9): p. 2562-2569.

67. Bortolotti, M., Kreis, R., Debard, C., Cariou, B., Faeh, D., Chetiveaux, M., Ith, M., Vermathen, P., Stefanoni, N., Le, K.A., Schneiter, P., Krempf, M., Vidal, H., Boesch, C., and Tappy, L., High protein intake reduces intrahepatocellular lipid deposition in humans. American Journal of Clinical Nutrition, 2009. 90(4): p. 1002-1010.

68. Wiseman, H., O'Reilly, J.D., Adlercreutz, H., Mallet, A.I., Bowey, E.A., Rowland, I.R., and Sanders, T.A., Isoflavone phytoestrogens consumed in soy decrease F(2)-isoprostane concentrations and increase resistance of low-density lipoprotein to oxidation in humans. American Journal of Clinical Nutrition, 2000. 72(2): p. 395-400.

Page 76: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

76

69. John W. Erdman Jr, Soy Protein and Cardiovascular Disease. Circulation, 2000. 102: p. 2555-2559. 70. Um, M.Y., Ahn, J., Jung, C.H., and Ha, T.Y., Cholesterol-lowering Effect of Rice Protein by

Enhancing Fecal Excretion of Lipids in Rats. Preventive Nutrition and Food Science, 2013. 18(3): p. 210-213.

71. Hunt, M.C., Tillander, V., and Alexson, S.E., Regulation of peroxisomal lipid metabolism: The role of acyl-CoA and coenzyme A metabolizing enzymes. Biochimie, 2014. 98C: p. 45-55.

72. Nagata, Y., Ishiwaki, N., and Sugano, M., Studies on the mechanism of the antihypercholesterolemic action of soy protein and soy protein-type amino acid mixtures in relation to their casein counterparts in rats. Journal of Nutrition, 1982. 112: p. 1614-1625.

73. Potter, S.M., Overview of proposed mechanisms for the hypocholesterolemic effect of soy. Journal of Nutrition, 1995. 125(3 Suppl): p. 606S-611S.

74. Bakhit, R.M., Klein, B.P., Essex-Sorlie, D., Ham, J.O., Erdman, J.W., Jr., and Potter, S.M., Intake of 25 g of soybean protein with or without soybean fiber alters plasma lipids in men with elevated cholesterol concentrations. Journal of Nutrition, 1994. 124(2): p. 213-222.

75. Fatma, Z.N., Bety Wahyu, H., and Waluyo, The effect of peptide (Asp – Glu) synthetic base on sterilized fermented soymilk on lipid profile of Sprague Dawley rats. International Food Research Journal, 2013. 20(6): p. 3047-3052.

76. Wergedahl, H., Liaset, B., Gudbrandsen, O.A., Lied, E., Espe, M., Muna, Z., Mork, S., and Berge, R.K., Fish protein hydrolysate reduces plasma total cholesterol, increases the proportion of HDL cholesterol, and lowers acyl-CoA:cholesterol acyltransferase activity in liver of Zucker rats. Journal of Nutrition, 2004. 134(6): p. 1320-1327.

77. Hosomi, R., Fukunaga, K., Arai, H., Kanda, S., Nishiyama, T., and Yoshida, M., Effect of Simultaneous Intake of Fish Protein and Fish Oil on Cholesterol Metabolism in Rats Fed High-Cholesterol Diets. Open Nutraceuticals Journal, 2011. 4: p. 12-19.

78. Kim, E.K., Lee, S.J., Jeon, B.T., Moon, S.H., Kim, B., Park, T.K., Han, J.S., and Park, P.J., Purification and characterisation of antioxidative peptides from enzymatic hydrolysates of venison protein. Food Chemistry, 2009. 114: p. 1365–1370.

79. Sakanaka, S. and Tachibana, Y., Active scavenging activity of egg-yolk protein hydrolysates and their effects on lipid oxidation in beef and tuna homogenates. Food Chemistry, 2006. 95: p. 243–249.

80. Cinq-Mars, C.D., Hu, C., Kitts, D.D., and Li-Chan, E.C.Y., Investigations into inhibitor type and mode, simulated gastrointestinal digestion, and cell transport of the angiotensin I-Converting enzyme-inhibitory peptides in pacific hake (Merluccius productus) fillet hydrolysate. Journal of Agricultural and Food Chemistry, 2008. 56: p. 410–419.

81. Raghavan, S. and Kristinsson, H.G., Antioxidant efficacy of alkali-treated tilapia protein hydrolysates: A comparison study of five enzymes. Journal of Agricultural and Food Chemistry, 2008. 56: p. 1434–1441.

82. Ahhmed, A.M. and Muguruma, M., A review of meat protein hydrolysates and hypertension. Meat science, 2010. 86(1): p. 110-118.

83. Shahidi, F. and Amarowicz, R., Antioxidant activity of protein hydrolysates from aquatic spe- cies. Journal of American Oil Chemists´ Society. Journal of the American Oil Chemists’ Society, 1996. 73: p. 1197–1199.

84. Manson, J.E., Gaziano, J.M., Jonas, M.A., and Hennekens, C.H., Antioxidants and cardiovascular disease: a review. Journal of the American College of Nutrition, 1993. 12(4): p. 426-432.

85. Lip, G.Y.H., Felmeden, D.C., Li-Saw-Hee, F.L., and Beevers, D.G., Hypertensive heart disease - A complex syndrome or a hypertensive ‘cardiomyopathy’? European Heart Journal, 2000. 21: p. 1653–1665.

86. Vercruysse, L., Van Camp, J., and Smagghe, G., ACE inhibitory peptides derived from enzymatic hydrolysates of animal muscle protein: a review. Journal of Agricultural and Food Chemistry, 2005. 53(21): p. 8106-8115.

87. Saiga, A., Iwai, K., Hayakawa, T., Takahata, Y., Kitamura, S., Nishimura, T., and Morimatsu, F., Angiotensin I-converting enzyme-inhibitory peptides obtained from chicken collagen hydrolysate. Journal of Agricultural and Food Chemistry, 2008. 56(20): p. 9586-9591.

88. Theodore, A.E. and Kristinsson, H.G., Angiotensin converting enzyme inhibition of fish protein hydrolysates prepared from alkaline-aided channel catfish protein isolate. Journal of the Science of Food and Agriculture, 2007. 87: p. 2353-2357.

89. Salze, G.P. and Davis, D.A., Taurine: a critical nutrient for future fish feeds. Aquaculture, 2015. 437: p. 215-229.

90. Rosa, F.T., Freitas, E.C., Deminice, R., Jordao, A.A., and Marchini, J.S., Oxidative stress and inflammation in obesity after taurine supplementation: a double-blind, placebo-controlled study. European journal of nutrition, 2014. 53(3): p. 823-830.

Page 77: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

77

91. Merheb, M., Daher, R.T., Nasrallah, M., Sabra, R., Ziyadeh, F.N., and Barada, K., Taurine intestinal absorption and renal excretion test in diabetic patients: a pilot study. Diabetes Care, 2007. 30(10): p. 2652-2654.

92. Hansen, S.H., Andersen, M.L., Cornett, C., Gradinaru, R., and Grunnet, N., A role for taurine in mitochondrial function. Journal of biomedical science, 2010. 17 Suppl 1: p. S23.

93. Chen, W., Guo, J.X., and Chang, P., The effect of taurine on cholesterol metabolism. Molecular Nutrition & Food Research, 2012. 56(5): p. 681-690.

94. Murakami, S., Kondo, Y., Toda, Y., Kitajima, H., Kameo, K., Sakono, M., and Fukuda, N., Effect of taurine on cholesterol metabolism in hamsters: up-regulation of low density lipoprotein (LDL) receptor by taurine. Life sciences, 2002. 70(20): p. 2355-2366.

95. Gudbrandsen, O.A., Wergedahl, H., Liaset, B., Espe, M., and Berge, R.K., Dietary proteins with high isoflavone content or low methionine-glycine and lysine-arginine ratios are hypocholesterolaemic and lower the plasma homocysteine level in male Zucker fa/fa rats. British Journal of Nutrition, 2005. 94(3): p. 321-330.

96. Tome, D. and Bos, C., Lysine requirement through the human life cycle. Journal of Nutrition, 2007. 137(6 Suppl 2): p. 1642S-1645S.

97. Weigensberg, B.I., Stary, H.C., and McMillan, G.C., Effect of lysine deficiency on cholesterol atherosclerosis in rabbits. Experimental and Molecular Pathology, 1964. 90: p. 444-454.

98. Davies, K.J. and Delsignore, M.E., Protein damage and degradation by oxygen radicals. III. Modification of secondary and tertiary structure. Journal of Biological Chemistry, 1987. 262(20): p. 9908-9913.

99. Bach, A., Carnitine biosynthesis in mammals. Reproduction, Nutrition, Development, 1982. 22(4): p. 583-596.

100. Siasos, G., Tousoulis, D., Antoniades, C., Stefanadi, E., and Stefanadis, C., L-Arginine, the substrate for NO synthesis: an alternative treatment for premature atherosclerosis? International Journal of Cardiology, 2007. 116(3): p. 300-308.

101. Oda, H., Fukui, H., Hitomi, Y., and Yoshida, A., Alteration of Serum Lipoprotein Metabolism by Polychlorinated Biphenyls and Methionine in Rats Fed a Soybean Protein Diet. Journal of Nutrition, 1991. 121: p. 925-933.

102. Hadi, H.A., Carr, C.S., and Al Suwaidi, J., Endothelial dysfunction: cardiovascular risk factors, therapy, and outcome. Journal of Vascular Health and Risk Managment, 2005. 1(3): p. 183-198.

103. Zulli, A., Hare, D.L., Buxton, B.F., and Black, M.J., High dietary methionine plus cholesterol exacerbates atherosclerosis formation in the left main coronary artery of rabbits. Atherosclerosis, 2004. 176(1): p. 83-89.

104. Morita, T., Oh-hashi, A., Takei, K., Ikai, M., Kasaoka, S., and Kiriyama, S., Cholesterol-lowering effects of soybean, potato and rice proteins depend on their low methionine contents in rats fed a cholesterol-free purified diet. Journal of Nutrition, 1997. 127(3): p. 470-477.

105. Sugiyama K, Ohkawa S, and K., M., Relationship between amino acid composition of diet and plasma cholesterol level in growing rats fed a high cholesterol diet. Journal of Nutritional Science and Vitaminology (Tokyo), 1986. 32(4): p. 413-423.

106. Fukagawa, N.K., Protein and amino acid supplementation in older humans. Amino Acids, 2013. 44(6): p. 1493-1509.

107. Newton, A.C., Protein Kinase C: Structure, Function, and Regulation. Journal of Biological Chemistry, 1995. 270: p. 28495-28498.

108. Dircks, L. and Sul, H.S., Acyltransferases of de novo glycerophospholipid biosynthesis. Progress in Lipid Research, 1999. 38(5-6): p. 461-479.

109. Lehner, R. and Kuksis, A., Biosynthesis of triacylglycerols. Progress in Lipid Research, 1996. 35(2): p. 169-201.

110. Coleman, R.A. and Lee, D.P., Enzymes of triacylglycerol synthesis and their regulation. Progress in Lipid Research, 2004. 43(2): p. 134-176.

111. Gonzalez-Baró, M.R., Lewin, T.M., and Coleman, R.A., Regulation of Triglyceride Metabolism II. Function of mitochondrial GPAT1 in the regulation of triacylglycerol biosynthesis and insulin action. American Journal of Physiology - Gastrointestinal and Liver Physiology, 2007. 292(5): p. G1195-G1199.

112. Houten, S.M., Watanabe, M., and Auwerx, J., Endocrine functions of bile acids. EMBO Journal, 2006. 25(7): p. 1419-1425.

113. Kowal, R.C., Herz, J., Goldstein, J.L., Esser, V., and Brown, M.S., Low density lipoprotein receptor-related protein mediates uptake of cholesteryl esters derived from apoprotein E-enriched lipoproteins. Proceedings of the National Academy of Sciences of the United States of America, 1989. 86: p. 5810-5814.

Page 78: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

78

114. Goldberg, I.J., Scheraldi, C.A., Yacoub, L.K., Saxena, U., and Bisgaier, C.L., Lipoprotein ApoC-II activation of lipoprotein lipase. Modulation by apolipoprotein A-IV. Journal of Biological Chemistry, 1990. 265(8): p. 4266-4272.

115. Cooper, A.D., Hepatic uptake of chylomicron remnants. Journal of Lipid Research, 1997. 38: p. 2173-2192.

116. Ginsberg, H.N., New Perspectives on Atherogenesis: Role of Abnormal Triglyceride-Rich Lipoprotein Metabolism. Circulation, 2002. 106: p. 2137-2142.

117. Kinnunen, P.K., Jackson, R.L., Smith, L.C., Gotto, A.M., Jr., and Sparrow, J.T., Activation of lipoprotein lipase by native and synthetic fragments of human plasma apolipoprotein C-II. Proceedings of the National Academy of Sciences of the United States of America, 1977. 74(11): p. 4848-4851.

118. Have, R.J., The formation of LDL: mechanisms and regulation. Journal of Lipid Research. 25: p. 1570-1576.

119. Segrest, J.P., Jones, M.K., De Loof, H., and Dashti, N., Structure of apolipoprotein B-100 in low density lipoproteins. Journal of Lipid Research, 2001. 42(9): p. 1346-1367.

120. Rader, D.J., Molecular regulation of HDL metabolism and function: implications for novel therapies. Journal of Clinical Investigation, 2006. 116(12): p. 3090-3100.

121. Fielding, C.J. and Fielding, P.E., Molecular physiology of reverse cholesterol transport. Journal of Lipid Research, 1995. 36: p. 211-228.

122. Holleboom, A.G., Kuivenhoven, J.A., Vergeer, M., Hovingh, G.K., Miert, J.N.v., Wareham, N.J., Kastelein, J.J.P., Khaw, K.-T., and Boekholdt, S.M., Plasma levels of lecithin:cholesterol acyltransferase and risk of future coronary artery disease in apparently healthy men and women: a prospective case-control analysis nested in the EPIC-Norfolk population study. Journal of Lipid Research, 2010. 51(2): p. 416-421.

123. Panda, T., Basak, T., Saraswathi, G., and Théodore, T., Kinetic Mechanisms of Cholesterol Synthesis: A Review. Industrial and Engineering Chemistry Research, 2011. 50: p. 12847-12864.

124. Liscum, L., Biochemsitry of Lipids, Lipoprotein and Membranes (4th edition). Cholesterol biosynthesis, ed. D.E. Vance and J.E. Vance. 2002: Elsevier Science B.V.

125. Olivier, L.M. and Krisans, S.K., Peroxisomal protein targeting and identification of peroxisomal targeting signals in cholesterol biosynthetic enzymes. Biochimica et Biophysica Acta, 2000. 1529(1-3): p. 89-102.

126. Crockett, E.L., Cholesterol Function in Plasma Membranes from Ectotherms: Membrane-Specific Roles in Adaptation to Temperature. Integrative & Comparative Biology, 1998. 38(2): p. 291-304.

127. Hu, J., Zhang, Z., Shen, W.J., and Azhar, S., Cellular cholesterol delivery, intracellular processing and utilization for biosynthesis of steroid hormones. Nutrition & Metabolism (Lond), 2010. 7: p. 47.

128. Mast, N., Graham, S.E., Andersson, U., Bjorkhem, I., Hill, C., Peterson, J., and Pikuleva, I.A., Cholesterol binding to cytochrome P450 7A1, a key enzyme in bile acid biosynthesis. Biochemistry, 2005. 44(9): p. 3259-3271.

129. Russell, D.W., The enzymes, regulation, and genetics of bile acid synthesis. Annual Review of Biochemistry, 2003. 72: p. 137-174.

130. Bellentani, S., Pecorari, M., Cordoma, P., Marchegiano, P., Manenti, F., Bosisio, E., Fabiani, E.D., and Galli, G., Taurine increases bile acid pool size and reduces bile saturation index in the hamster. Journal of Lipid Research, 1987. 28: p. 1021-1027.

131. Mathur, F.J.F.a.S.N., Regulation of acyl CoA:cholesterol acyltransferase by 25-hydroxycholesteroI in rabbit intestinal microsomes and absorptive cells. Journal of Lipid Research, 1983. 24: p. 1049-1059.

132. Zerbin, J. Metabolic cholestatic pruritus. 2013 [cited 2014 5th of April]. 133. Jonckheere, A.I., Smeitink, J.A.M., and Rodenburg, R.J.T., Mitochondrial ATP synthase: architecture,

function and pathology. Journal of Inherited Metabolic Disease, 2012. 35(2): p. 211-225. 134. Wanders, R.J.A., Ruiter, J.P.N., IJlst, L., Waterham, H.R., and Houten, S.M., The enzymology of

mitochondrial fatty acid beta-oxidation and its application to follow-up analysis of positive neonatal screening results. Journal of Inherited Metabolic Disease 2010. 33(5): p. 479-494.

135. Flanagan, J.L., Simmons, P.A., Vehige, J., Willcox, M.D.P., and Garrett, Q., Role of carnitine in disease. Nutrition & Metabolism, 2010. 7: p. 30.

136. Houten, S.M. and Wanders, R.J.A., A general introduction to the biochemistry of mitochondrial fatty acid β-oxidation. Journal of Inherited Metabolic Disease, 2010. 33(5): p. 469-477.

137. Nelson, D.L. and Cox, M.M., Lehninger Priciples of biochemistry 4th edition. 2005: W.H. Freeman and Company, New York.

138. BioCarta. Comparison of Beta oxidation in mitochondria and peroxisomes and glyoxysomes. 2001 [cited 2014 10th of March]; Available from: http://www.biocarta.com/pathfiles/boxnpPathway.asp.

Page 79: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

79

139. Manninen, A.H., Metabolic Effects of the Very-Low-Carbohydrate Diets: Misunderstood "Villains" of Human Metabolism. Journal of the International Society of Sports Nutrition, 2004. 1: p. 7-11.

140. Reddy, J.K. and Hashimoto, T., Peroxisomal beta-oxidation and peroxisome proliferator-activated receptor alpha: an adaptive metabolic system. Annual Review of Nutrition, 2001. 21: p. 193-230.

141. Reddy, J.K. and Hashimoto, T., Peroxisomal β-oxidation and peroxisiome proliferator-activated receptor α: An Adaptive Metabolic System. Annual Review of Nutrition. 21: p. 193-230.

142. Antonenkov, V.D. and Hiltunen, J.K., Transfer of metabolites across the peroxisomal membrane. Biochimica et Biophysica Acta, 2012. 1822(9): p. 1374-1386.

143. Poirier, Y., Antonenkov, V.D., Glumoff, T., and Hiltunen, J.K., Peroxisomal beta-oxidation - a metabolic pathway with multiple functions. Biochimica et Biophysica Acta, 2006. 1763(12): p. 1413-1426.

144. Vlaemincka, B., Fieveza, V., Cabritab, A.R.J., Fonsecac, A.J.M., and Dewhurstd, R.J., Factors affecting odd- and branched-chain fatty acids in milk: A review. Animal Feed Science and Technology, 2006. 131(3-4): p. 389-417.

145. Fatty acid synthesis. [cited 2014 7th of May]; Available from: http://ansci.illinois.edu/static/ansc438/Milkcompsynth/milksynth_fatfasynthesis.html.

146. Cinti, D.L., Cook, L., Nagi, M.N., and Suneja, S.K., The fatty acid chain elongation system of mammalian endoplasmic reticulum. Progress in Lipid Research, 1992. 31(1): p. 1-51.

147. Seubert, W. and Podack, E.R., Mechanisms and physiological roles of fatty acid chain elongation in microsomes and mitochondria. Molecular and Cellular Biochemistry, 1973. 1(1): p. 29-40.

148. Liu, X., Strable, M.S., and Ntambi, J.M., Stearoyl CoA desaturase 1: role in cellular inflammation and stress. Advances in Nutrition, 2011. 2(1): p. 15-22.

149. Nakamura, M.T. and Nara, T.Y., Structure, function, and dietary regulation of delta6, delta5, and delta9 desaturases. Annual Review of Nutrition, 2004. 24: p. 345-376.

150. Miyazaki, M., Kim, H.-J., Man, W.C., and Ntambi, J.M., Oleoyl-CoA Is the Major de Novo Product of Stearoyl-CoA Desaturase 1 Gene Isoform and Substrate for the Biosynthesis of the Harderian Gland 1-Alkyl-2,3-diacylglycerol. Journal of Biological Chemistry, 2001. 276: p. 39455-39461.

151. Maceyka, M. and Machamer, C.E., Ceramide accumulation uncovers a cycling pathway for the cis-Golgi network marker, infectious bronchitis virus M protein. The Journal of cell biology, 1997. 139(6): p. 1411-1418.

152. Cowart, L.A., Sphingolipids and Metabolic Disease. Vol. 721. New York, NY: Springer Science+Business Media , LLC, Landes Bioscience.

153. Johnson A, A.B. and Lewis, J. Molecular Biology of the cell. 4th edition. 2002 [cited 2015 12th of April]; Available from: http://www.ncbi.nlm.nih.gov/books/NBK26830/.

154. Wernerman, J., von der Decken, A., and Vinnars, E., Protein synthesis in skeletal muscle in relation to nitrogen balance after abdominal surgery: the effect of total parenteral nutrition. Journal of Parenteral Enteral Nutrition, 1986. 10(6): p. 578-582.

155. Cahill, G.F., Jr. and Owen, O.E., Starvation and survival. Transactions of the American Clinical and Climatological Association, 1968. 79: p. 13-20.

156. Rittig, N., Bach, E., Thomsen, H.H., Johannsen, M., Jørgensen, J.O., Richelsen, B., Jessen, N., and Møller, N., Amino acid supplementation is anabolic during the acute phase of endotoxin-induced inflammation: A human randomized crossover trial. Clinical Nutrition, 2015. In Press, Corrected Proof.

157. Butterworth, R.F., Pathophysiology of brain dysfunction in hyperammonemic syndromes: The many faces of glutamine. Molecular Genetics and Metabolism, 2014. 113(1-2): p. 113-117.

158. Silk, D.B.A., Grimble, G.K., and Rees, R.G., Protein digestion and amino acid and peptide absorption Proceedings of the Nutrition Society, 1985. 44: p. 63-72.

159. Lyons, P.J. and Fricker, L.D., Carboxypeptidase O Is a Glycosylphosphatidylinositol-anchored Intestinal Peptidase with Acidic Amino Acid Specificity. Journal of Biological Chemistry, 2011. 286(45): p. 39023–39032.

160. Adibi, S.A. and Morse, E.L., Intestinal transport of dipeptides in man: relative importance of hydrolysis and intact absorption. Journal of Clinical Investigation, 1971. 50(11): p. 2266–2275.

161. Shanbhag, V.M. and Martell, A.E., Oxidative deamination of amino acids by molecular oxygen with pyridoxal derivatives and metal ions as catalysts. Journal of the American Chemical Society, 1991. 113(17): p. 6479–6487.

162. Owen, O.E., Ketone Bodies as a Fuel for the Brain during Starvation. The International Union of Biochemistry and Molecular Biology, 2005. 33: p. 246-251.

163. Sierra-Santoyo, A., López, M.L., Henrnández, T., and Mendoza-Figueroa, T., Urea production in long-term cultures of adult rat hepatocytes. Toxicology in Vitro, 1994. 8(2): p. 293-299.

Page 80: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

80

164. T., S.R., Adaptive characteristics of urea cycle enzymes in the rat. Journal of Biological Chemistry, 1962. 237: p. 459-468.

165. Kim, E.K., Lee, S.J., Jeon, B.T., Moon, S.H., Kim, B., Park, T.K., Han, J.S., and Park, P.J., Purification and characterisation of antioxidative peptides from enzymatic hydrolysates of venison protein. Food Chemistry, 2009. 114: p. 1365–1370.

166. Matsui, T., Matsumoto, K., Mahmud, T.H.K., and Arjumand, A., Antihypertensive peptides from natural resources Advances in Phytomedicine. Vol. 2. 2006, UK: Elsevier; Oxford,.

167. Ryan, J.T., Ross, P.R., Bolton, D., Fitzgerald, G.F., and Stanton, C., Bioactive Peptides from Muscle Sources: Meat and Fish. Nutrients, 2011. 3(9): p. 765-791.

168. S. Ono, M. Hosokawa, K. Miyashita, and Takahashi, K., Isolation of Peptides with Angiotensin I–converting Enzyme Inhibitory Effect Derived from Hydrolysate of Upstream Chum Salmon Muscle. Journal of food science, 2003. 68(5): p. 1611-1614.

169. Samaranayaka, A.G., Kitts, D.D., and Li-Chan, E.C., Antioxidative and angiotensin-I-converting enzyme inhibitory potential of a Pacific Hake ( Merluccius productus ) fish protein hydrolysate subjected to simulated gastrointestinal digestion and Caco-2 cell permeation. Journal of Agricultural and Food Chemistry, 2010. 58(3): p. 1535-1542.

170. Valenzuela, A., Sanhueza, J., and Nieto, S., Natural antioxidants in functional foods: From food safety to health benefits. . Grasas Aceites, 2003. 54: p. 295-303.

171. Contreras, A., Rangel, C., Ortiz, V., Aleman, G., Palacios-Gonzalez, B., Tejero, E., Torres, N., and Tovar, A., Dietary protein regulates gene expression of amino acid-degrading enzymes through PPARα. The Journal of the Federation of American Societies for Experimental Biology, 2013. 27: p. 631.

172. Sheikh, K., Camejo, G., Lanne, B., Halvarsso, T., Landergren, M.R., and Oakes, N.D., Beyond lipids, pharmacological PPARα activation has important effects on amino acid metabolism as studied in the rat. American Journal of Physiology - Endocrinology and Metabolism 2007. 292(4): p. E1157-E1165

173. Kersten, S., Mandard, S., Escher, P., Gonzalez, F.J., Tafuri, S., Desvergne, B., and Wahli, W., The peroxisome proliferator-activated receptor alpha regulates amino acid metabolism. Federation of American Societies for Experimental Biology Journal, 2001. 15(11): p. 1971-1978.

174. Krey, G., Braissant, O., L'Horset, F., Kalkhoven, E., Perroud, M., Parker, M.G., and Wahli, W., Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay. Molecular Endocrinology, 1997. 11(6): p. 779-791.

175. Matsuda, S. and Kitagishi, Y., Peroxisome proliferator-activated receptor and vitamin d receptor signaling pathways in cancer cells. Cancers (Basel), 2013. 5(4): p. 1261-1270.

176. Temple, K.A., Cohen, R.N., Wondisford, S.R., Yu, C., Deplewski, D., and Wondisford, F.E., An intact DNA-binding domain is not required for peroxisome proliferator-activated receptor gamma (PPARgamma) binding and activation on some PPAR response elements. Journal of Biological Chemistry, 2005. 280(5): p. 3529-3540.

177. Pruimboom-Brees, I., Haghpassand, M., Royer, L., Brees, D., Aldinger, C., Reagan, W., Singh, J., Kerlin, R., Kane, C., Bagley, S., Hayward, C., Loy, J., O'Brien, P., and Francone, O.L., A critical role for peroxisomal proliferator-activated receptor-alpha nuclear receptors in the development of cardiomyocyte degeneration and necrosis. American Journal of Pathology, 2006. 169(3): p. 750-760.

178. Plutzky, J., The PPAR-RXR transcriptional complex in the vasculature: energy in the balance. Circulation Research, 2011. 108(8): p. 1002-1016.

179. Larsen, T.M., Toubro, S., and Astrup, A., PPARgamma agonists in the treatment of type II diabetes: is increased fatness commensurate with long-term efficacy? International Journal of Obesity Related Metabolic Disorders, 2003. 27(2): p. 147-161.

180. Auboeuf, D., Rieusset, J., Fajas, L., Vallier, P., Frering, V., Riou, J.P., Staels, B., Auwerx, J., Laville, M., and Vidal, H., Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-alpha in humans: no alteration in adipose tissue of obese and NIDDM patients. Diabetes, 1997. 46(8): p. 1319-1327.

181. Staels, B. and Fruchart, J.-C., Therapeutic Roles of Peroxisome Proliferator–Activated Receptor Agonists. American Diabetes Association, 2005. 51: p. 2460-2470.

182. Vosper, H., Patel, L., Graham, T.L., Khoudoli, G.A., Hill, A., Macphee, C.H., Pinto, I., Smith, S.A., Suckling, K.E., Wolf, C.R., and Palmer, C.N.A., The Peroxisome Proliferator-activated Receptor δ Promotes Lipid Accumulation in Human Macrophages. Journal of Biological Chemistry, 2001. 276: p. 44258-44265.

183. Lalloyer, F. and Staels, B., Fibrates, glitazones, and peroxisome proliferator-activated receptors. Arteriosclerosis, Thrombosis and Vascular Biology, 2010. 30(5): p. 894-899.

Page 81: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

81

184. Chan, D.C., Watts, G.F., Ooi, E.M., Ji, J., Johnson, A.G., and Barrett, P.H., Atorvastatin and fenofibrate have comparable effects on VLDL-apolipoprotein C-III kinetics in men with the metabolic syndrome. Arteriosclerosis, Thrombosis and Vascular Biology, 2008. 28(10): p. 1831-1837.

185. Berthou, L., Duverger, N., Emmanuel, F., Langouet, S., Auwerx, J., Guillouzo, A., Fruchart, J.C., Rubin, E., Denefle, P., Staels, B., and Branellec, D., Opposite regulation of human versus mouse apolipoprotein A-I by fibrates in human apolipoprotein A-I transgenic mice. Journal of Clinical Investigation, 1996. 97(11): p. 2408-2416.

186. Hossain, M.A., Tsujita, M., Gonzalez, F.J., and Yokoyama, S., Effects of fibrate drugs on expression of ABCA1 and HDL biogenesis in hepatocytes. Journal of Cardiovascular Pharmacology, 2008. 51(3): p. 258-266.

187. Finck, B.N., The PPAR regulatory system in cardiac physiology and disease. Cardiovascular Research, 2006. 73: p. 269-277.

188. Lee, M.Y., Choi, R., Kim, H.M., Cho, E.J., Kim, B.H., Choi, Y.S., Naowaboot, J., Lee, E.Y., Yang, Y.C., Shin, J.Y., Shin, Y.G., and Chung, C.H., Peroxisome proliferator-activated receptor delta agonist attenuates hepatic steatosis by anti-inflammatory mechanism. Experimental & Molecular Medicine, 2012. 44(10): p. 578-585.

189. Alemán, G., Ortiz, V., Contreras, A.V., Quiroz, G., Ordaz-Nava, G., Langley, E., Torres, N., and Tovar, A.R., Hepatic amino acid-degrading enzyme expression is downregulated by natural and synthetic ligands of PPARα in rats. Journal of Nutrition, 2013. 143(8): p. 1211-1218.

190. Amemiya-Kudo, M., Shimano, H., Hasty, A.H., Yahagi, N., Yoshikawa, T., Matsuzaka, T., Okazaki, H., Tamura, Y., Iizuka, Y., Ohashi, K., Osuga, J., Harada, K., Gotoda, T., Sato, R., Kimura, S., Ishibashi, S., and Yamada, N., Transcriptional activities of nuclear SREBP-1a, -1c, and -2 to different target promoters of lipogenic and cholesterogenic genes. Journal of Lipid Research, 2002. 43(8): p. 1220-1235.

191. Horton, J.D., Goldstein, J.L., and Brown, M.S., SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. Journal of Clinical Investigation, 2002. 109(9): p. 1125-1131.

192. Sakai, J., Nohturfft, A., Cheng, D., Ho, Y.K., Brown, M.S., and Goldstein, J.L., Identification of complexes between the COOH-terminal domains of sterol regulatory element-binding proteins (SREBPs) and SREBP cleavage-activating protein. Journal of Biological Chemistry, 1997. 272(32): p. 20213-20221.

193. Eberle, D., Hegarty, B., Bossard, P., Ferre, P., and Foufelle, F., SREBP transcription factors: master regulators of lipid homeostasis. Biochimie, 2004. 86(11): p. 839-848.

194. Sakai, J., Duncan, E.A., Rawson, R.B., Hua, X., Brown, M.S., and Goldstein, J.L., Sterol-regulated release of SREBP-2 from cell membranes requires two sequential cleavages, one within a transmembrane segment. Cell, 1996. 85(7): p. 1037-1046.

195. Wang, X., Sato, R., Brown, M.S., Hua, X., and Goldstein, J.L., SREBP-1, a membrane-bound transcription factor released by sterol-regulated proteolysis. Cell, 1994. 77(1): p. 53-62.

196. Shimano, H., Sterol regulatory element-binding proteins (SREBPs): transcriptional regulators of lipid synthetic genes. Progress in Lipid Research, 2001. 40(6): p. 439-452.

197. Foufelle, F. and Ferre, P., New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-1c. The Biochemical journal, 2002. 366(Pt 2): p. 377-391.

198. Ascencio, C., Torres, N., Isoard-Acosta, F., Gomez-Perez, F.J., Hernandez-Pando, R., and Tovar, A.R., Soy protein affects serum insulin and hepatic SREBP-1 mRNA and reduces fatty liver in rats. Journal of Nutrition, 2004. 134(3): p. 522-529.

199. Tovar, A.R., Ascencio, C., and Torres, N., Soy protein, casein, and zein regulate histidase gene expression by modulating serum glucagon. American journal of physiology. Endocrinology and metabolism, 2002. 283(5): p. E1016-1022.

200. Torres, N., Torre-Villalvazo, I., and Tovar, A.R., Regulation of lipid metabolism by soy protein and its implication in diseases mediated by lipid disorders. The Journal of nutritional biochemistry, 2006. 17(6): p. 365-373.

201. Carroll, K.K., Review of clinical studies on cholesterol-lowering response to soy protein. Journal of the American Dietetic Association, 1991. 91(7): p. 820-827.

202. Lin, Y., Meijer, G.W., Vermeer, M.A., and Trautwein, E.A., Soy protein enhances the cholesterol-lowering effect of plant sterol esters in cholesterol-fed hamsters. Journal of Nutrition, 2004. 134(1): p. 143-148.

203. Moriyama, T., Kishimoto, K., Nagai, K., Urade, R., Ogawa, T., Utsumi, S., Maruyama, N., and Maebuchi, M., Soybean beta-conglycinin diet suppresses serum triglyceride levels in normal and genetically obese mice by induction of beta-oxidation, downregulation of fatty acid synthase, and

Page 82: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

82

inhibition of triglyceride absorption. Bioscience, biotechnology, and biochemistry, 2004. 68(2): p. 352-359.

204. Wang, Y., Jones, P.J., Ausman, L.M., and Lichtenstein, A.H., Soy protein reduces triglyceride levels and triglyceride fatty acid fractional synthesis rate in hypercholesterolemic subjects. Atherosclerosis, 2004. 173(2): p. 269-275.

205. Lovati, M.R., Manzoni, C., Gianazza, E., Arnoldi, A., Kurowska, E., Carroll, K.K., and Sirtori, C.R., Soy protein peptides regulate cholesterol homeostasis in Hep G2 cells. Journal of Nutrition, 2000. 130(10): p. 2543-2549.

206. Johnson, M., Laboratory mice and rats. The world of laboratory, 2012. 2: p. 113. 207. Clause, B.T., The Wistar rat as a right choice: Establishing Mammalian Standards and the Ideal of a

Standardized Mammal. Journal of the History of Biology, 1993. 26: p. 329-349. 208. Mekada, K., Abe, K., Murakami, A., Nakamura, S., Nakata, H., Moriwaki, K., Obata, Y., and Yoshiki,

A., Genetic differences among C57BL/6 substrains. Experimental Animals, 2009. 58(2): p. 141-149. 209. Engber, D. The Trouble with black-6. 2011 [cited 2014 10th of April]; Available from:

http://www.slate.com/articles/health_and_science/the_mouse_trap/2011/11/black_6_lab_mice_and_the_history_of_biomedical_research.html.

210. Jawien, J. Mouse Models of Experimental Atherosclerosis as a Tool for Checking a Putative Anti-Atherogenic Action of Drugs. 2012 [cited 2014 10th of April]; Available from: http://cdn.intechopen.com/pdfs-wm/25897.pdf.

211. Laboratory, T.J. B6.129P2-Apoetm1Unc/J. 2006 [cited 2014 10th of April]; Available from: http://jaxmice.jax.org/strain/002052.html.

212. Taconic. Apoe (ApoE) - Model APOE Constitutive Knock Out. [cited 2014 10th of April]; Available from: http://www.taconic.com/apoe.

213. Veniant, M.M., Withycombe, S., and Young, S.G., Lipoprotein size and atherosclerosis susceptibility in Apoe(-/-) and Ldlr(-/-) mice. Arteriosclerosis, Thrombosis and Vascular Biology, 2001. 21(10): p. 1567-1570.

214. Halpin, H.A., Morales-Suárez-Varela, M.M., and Martin-Moreno, J.M., Chronic Disease Prevention and the New Public Health. Public Health Reviews, 2010. 32: p. 120-154.

215. Liaset, B., Madsen, L., Hao, Q., Criales, G., Mellgren, G., Marschall, H.U., Hallenborg, P., Espe, M., Froyland, L., and Kristiansen, K., Fish protein hydrolysate elevates plasma bile acids and reduces visceral adipose tissue mass in rats. Biochimica et Biophysica Acta, 2009. 1791(4): p. 254-262.

216. Zhu, C.F., Li, G.Z., Peng, H.B., Zhang, F., Chen, Y., and Li, Y., Treatment with marine collagen peptides modulates glucose and lipid metabolism in Chinese patients with type 2 diabetes mellitus. Applied Physiology, Nutrition and Metabolism, 2010. 35(6): p. 797-804.

217. Klompong, V., Benjakul, S., Kantachote, D., and Shahidi, F., Antioxidative activity and functional properties of protein hydrolysate of yellow stripe trevally (Selaroides leptolepis) as influenced by the degree of hydrolysis and enzyme type. Food Chemistry, 2007. 102(1317-1327).

218. Cho, J. and Kim, Y., Sharks: a potential source of antiangiogenic factors and tumor treatments. Marine Biotechnology (NY), 2002. 4(6): p. 521-525.

219. Kristinsson, H.G. and Rasco, B.A., Fish protein hydrolysates: production, biochemical, and functional properties. Critical Reviews in Food Science and Nutrition, 2000. 40(1): p. 43-81.

220. Mullally, M.M., O’Callaghan, D.M., FitzGerald, R.J., Donnelly, W.J., and Dalton, J.P., Proteolytic and peptidolytic activities in commercial pancreatin protease preparations and their relationship to some whey protein hydrolysate characteristics. Journal of Agricultural and Food Chemistry, 1994. 42: p. 2973.

221. O'Dea, K., Marked improvement in carbohydrate and lipid metabolism in diabetic Australian aborigines after temporary reversion to traditional lifestyle. Diabetes, 1984. 33(6): p. 596-603.

222. Wolfe, B.M. and Giovannetti, P.M., Short-term effects of substituting protein for carbohydrate in the diets of moderately hypercholesterolemic human subjects. Metabolism, 1991. 40(4): p. 338-343.

223. Wolfe, B.M. and Piche, L.A., Replacement of carbohydrate by protein in a conventional-fat diet reduces cholesterol and triglyceride concentrations in healthy normolipidemic subjects. Clinical & Investigative Medicine, 1999. 22(4): p. 140-148.

224. Crovetti, R., Porrini, M., Santangelo, A., and Testolin, G., The influence of thermic effect of food on satiety. European Journal of Clinical Nutrition, 1998. 52(7): p. 482-488.

225. Johnston, C.S., Day, C.S., and Swan, P.D., Postprandial thermogenesis is increased 100% on a high-protein, low-fat diet versus a high-carbohydrate, low-fat diet in healthy, young women. Journal of the American College of Nutrition, 2002. 21(1): p. 55-61.

226. Stubbs, R.J., Nutrition Society Medal Lecture. Appetite, feeding behaviour and energy balance in human subjects. Proceedings of the Nutrition Society, 1998. 57(3): p. 341-356.

Page 83: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

83

227. Cordain, L., Eaton, S.B., Miller, J.B., Mann, N., and Hill, K., The paradoxical nature of hunter-gatherer diets: meat-based, yet non-atherogenic. European Journal of Clinical Nutrition, 2002. 56 Suppl 1: p. S42-52.

228. Skov, A.R., Toubro, S., Ronn, B., Holm, L., and Astrup, A., Randomized trial on protein vs carbohydrate in ad libitum fat reduced diet for the treatment of obesity. International Journal of Obesity Relatated Metabolic Disorders, 1999. 23(5): p. 528-536.

229. Bjørndal, B., Berge, C., Ramsvik, M.S., Svardal, A., Bohov, P., Skorve, J., and Berge, R.K., A fish protein hydrolysate alters fatty acid composition in liver and adipose tissue and increases plasma carnitine levels in a mouse model of chronic inflammation. Lipids in Health and Disease, 2013. 12: p. 143.

230. Harris, W.S., n-3 fatty acids and lipoproteins: comparison of results from human and animal studies. Lipids, 1996. 3: p. 243-252.

231. Shukla, A., Bettzieche, A., Hirche, F., Brandsch, C., Stangl, G.I., and Eder, K., Dietary fish protein alters blood lipid concentrations and hepatic genes involved in cholesterol homeostasis in the rat model. British Journal of Nutrition, 2006. 96: p. 674- 682.

232. Hosomi, R., Fukunaga, K., Arai, H., Nishiyama, T., and Yoshida, M., Effects of dietary fish protein on serum and liver lipid concentrations in rats and the expression of hepatic genes involved in lipid metabolism. Journal of Agricultural and Food Chemistry, 2009. 57: p. 9256-9262.

233. Zhang, X. and Beynen, A.C., Influence of dietary fish proteins on plasma and liver cholesterol concentrations in rats. British Journal of Nutrition, 1993. 69: p. 767-777.

234. Bergeron, N. and Jacques, H., Influence of fish protein as compared to casein and soy protein on serum and liver lipids, and serum lipoprotein cholesterol levels in the rabbit. Atherosclerosis, 1989. 78(2-3): p. 113-121.

235. Wakutsu, M., Tsunoda, N., Shiba, S., Muraki, E., and Kasono, K., Peroxisome proliferator-activated receptors (PPARs)-independent functions of fish oil on glucose and lipid metabolism in diet-induced obese mice. Lipids in Health and Disease, 2010. 9: p. 101.

236. Kim, H.J., Takahashi, M., and Ezaki, O., Fish oil feeding decreases mature sterol regulatory element-binding protein 1 (SREBP-1) by down-regulation of SREBP-1c mRNA in mouse liver. A possible mechanism for down-regulation of lipogenic enzyme mRNAs. Journal of Biological Chemistry, 1999. 274(36): p. 25892-25898.

237. Clarke, S.D. and Jump, D.B., Dietary polyunsaturated fatty acid regulation of gene transcription. Annual Review of Nutrition, 1994. 14: p. 83-98.

238. Pégorier, J.P., Regulation of gene expression by fatty acids. Current Opinion in Clinical Nutrition and Metababolic Care, 1998. 1: p. 329-334.

239. Hosomi, R., Fukunaga, K., Arai, H., Kanda, S., Nishiyama, T., and Yoshida, M., Fish protein decreases serum cholesterol in rats by inhibition of cholesterol and bile acid absorption. Journal of Food Science, 2011. 76(4): p. H116-121.

240. Pilon, G., Ruzzin, J., Rioux, L.E., Lavigne, C., White, P.J., Froyland, L., Jacques, H., Bryl, P., Beaulieu, L., and Marette, A., Differential effects of various fish proteins in altering body weight, adiposity, inflammatory status, and insulin sensitivity in high-fat-fed rats. Metabolism, 2011. 60(8): p. 1122-1130.

241. Peluffo, R.O., Nervi, A.M., Gonzalez, M.S., and Brenner, R.R., Effect of different amino acid diets on delta 5, delta 6 and delta 9 desaturases. Lipids, 1984. 19(2): p. 154-157.

242. Madeira, M.S., Pires, V.M., Alfaia, C.M., Luxton, R., Doran, O., Bessa, R.J., and Prates, J.A., Combined effects of dietary arginine, leucine and protein levels on fatty acid composition and gene expression in the muscle and subcutaneous adipose tissue of crossbred pigs. British Journal of Nutrition, 2014. 111(9): p. 1521-1535.

243. Sjogren, P., Sierra-Johnson, J., Gertow, K., Rosell, M., Vessby, B., de Faire, U., Hamsten, A., Hellenius, M.L., and Fisher, R.M., Fatty acid desaturases in human adipose tissue: Relationships between gene expression, desaturation indexes and insulin resistance. Diabetologia, 2008. 51(2): p. 328–335.

244. Kolak, M., Westerbacka, J., Velagapudi, V.R., Wagsater, D., Yetukuri, L., Makkonen, J., Rissanen, A., Hakkinen, A.M., Lindell, M., Bergholm, R., Hamsten, A., Eriksson, P., Fisher, R.M., Oresic, M., and Yki-Jarvinen, H., Adipose tissue inflammation and increased ceramide content characterize subjects with high liver fat content independent of obesity. Diabetes, 2007. 56(8): p. 1960-1968.

245. Bikman, B.T. and Summers, S.A., Ceramides as modulators of cellular and whole-body metabolism. Journal of Clinical Investigation, 2011. 121(11): p. 4222-4230.

246. Kotronen, A., Seppanen-Laakso, T., Westerbacka, J., Kiviluoto, T., Arola, J., Ruskeepaa, A.L., Oresic, M., and Yki-Jarvinen, H., Hepatic stearoyl-CoA desaturase (SCD)-1 activity and diacylglycerol but

Page 84: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

84

not ceramide concentrations are increased in the nonalcoholic human fatty liver. Diabetes, 2009. 58(1): p. 203-208.

247. Nagaoka, S., Futamura, Y., Miwa, K., Awano, T., Yamauchi, K., Kanamaru, Y., Tadashi, K., and Kuwata, T., Identification of novel hypocholesterolemic peptides derived from bovine milk beta-lactoglobulin. Biochemical and Biophysical Research Communications, 2001. 281(1): p. 11-17.

248. Brandsch, C., Shukla, A., Hirche, F., Stangl, G.I., and Eder, K., Effect of proteins from beef, pork, and turkey meat on plasma and liver lipids of rats compared with casein and soy protein. Nutrition, 2006. 22: p. 1162-1170.

249. Morimatsu, F., Ito, M., Budijanto, S., Watanabe, I., Furukawa, Y., and Kimura, S., Plasma cholesterol-suppressing effect of papain-hydrolyzed pork meat in rats fed hypercholesterolemic diet. Journal of Nutritional Science and Vitaminology (Tokyo), 1996. 42(2): p. 145-153.

250. Gabriel, S.E., Cardiovascular morbidity and mortality in rheumatoid arthritis. American Journal of Medicine, 2008. 121(10 Suppl 1): p. S9-14.

251. Kardys, I., de Maat, M.P., Uitterlinden, A.G., Hofman, A., and Witteman, J.C., C- reactive protein gene haplotypes and risk of coronary heart disease: the Rotterdam Study. European Heart Journal, 2006. 27: p. 1331-1337.

252. Prodanovich, S., Kirsner, R.S., Kravetz, J.D., Ma, F., Martinez, L., and Federman, D.G., Association of psoriasis with coronary artery, cerebrovascular, and peripheral vascular diseases and mortality. Archive of Dermatology, 2009. 145: p. 700-703.

253. Zacho, J., Tybjaerg-Hansen, A., Jensen, J.S., Grande, P., Sillesen, H., and Nordestgaard, B.G., Genetically elevated C-reactive protein and ischemic vascular disease. New England Journal of Medicine, 2008. 359(18): p. 1897-1908.

254. Dembinska-Kiec, A. and Gryglewski, R.J., Contribution of arachidonic acid metabolites to atherosclerosis. Wiener Klinische Wochenschrift, 1986. 98(7): p. 198-206.

255. Riccioni, G., Zanasi, A., Vitulano, N., Mancini, B., and D'Orazio, N., Leukotrienes in atherosclerosis: new target insights and future therapy perspectives. Mediators of Inflammation, 2009. 2009: p. 737282.

256. Høstmark, A.T. and Haug, A., Percentages of oleic acid and arachidonic acid are inversely related in phospholipids of human sera. Lipids in Health and Disease, 2013. 12: p. 106.

257. Schwart, S.M., Galis, Z.S., Rosenfeld, M.E., and Falk, E., Plaque rupture in humans and mice. Arteriosclerosis, Thrombosis and Vascular Biology, 2007. 27: p. 705-713.

258. Rioua, L.M., Broisata, A., Ghezzia, C., Finetb, G., Rioufolb, G., Gharibc, A.M., Pettigrewc, R.I., and Ohayond, J., Effects of mechanical properties and atherosclerotic artery size on biomechanical plaque disruption – Mouse vs. human. Journal of Biomechanics 2014. 47(4): p. 765-772.

259. Björkbacka, L., Atherosclerosis: cell biology and lipoproteins. Current Opinion in Lipidology, 2015. 26(1): p. 67-69.

260. Grimstad, T., Bjørndal, B., Cacabelos, D., Aasprong, O.G., Omdal, R., Svardal, A., Bohov, P., Pamplona, R., Portero-Otin, M., Berge, R.K., and Hausken, T., A salmon peptide diet alleviates experimental colitis as compared with fish oil. Journal of Nutritional Science, 2012. 2: p. 1-8.

261. Sun, Y., Pan, D., Guo, Y., and Li, J., Purification of chicken breast protein hydrolysate and analysis of its antioxidant activity. Food and Chemical Toxicology, 2012. 50(10): p. 3397-3404.

262. Zhang, Y., Kouguchi, T., Shimizu, M., Ohmori, T., Takahata, Y., and Morimatsu, F., Chicken collagen hydrolysate protects rats from hypertension and cardiovascular damage. Journal of Medicinal Food, 2010. 13(2): p. 399-405.

263. Saiga-Egusa, A., Iwai, K., Hayakawa, T., Takahata, Y., and Morimatsu, F., Antihypertensive effects and endothelial progenitor cell activation by intake of chicken collagen hydrolysate in pre- and mild-hypertension. Bioscience, biotechnology, and biochemistry, 2009. 73(2): p. 422-424.

264. Kaur, G., Cameron-Smith, D., Garg, M., and Sinclair, A.J., Docosapentaenoic acid (22:5n-3): a review of its biological effects. Prog Lipid Res, 2011. 50(1): p. 28-34.

265. Kaur, G., Sinclair, A.J., Cameron-Smith, D., Barr, D.P., Molero-Navajas, J.C., and Konstantopoulos, N., Docosapentaenoic acid (22:5n-3) down-regulates the expression of genes involved in fat synthesis in liver cells. Prostaglandins, leukotrienes, and essential fatty acids, 2011. 85(3-4): p. 155-161.

266. Singh, A.K., Yoshida, Y., Garvin, A.J., and Singh, I., Effect of fatty acids and their derivatives on mitochondrial structures. International Journal of Experimental Pathology, 1989. 4(1): p. 9-15.

267. Xu, Y.J., Arneja, A.S., Tappia, P.S., and Dhalla, N.S., The potential health benefits of taurine in cardiovascular disease. Experimental & Clinical Cardiology, 2008. 13(2): p. 57-65.

268. Kritchevsky, D., Tepper, S.A., Czarnecki, S.K., and Klurfeld, D.M., Atherogenicity of animal and vegetable protein. Influence of the lysine to arginine ratio. Atherosclerosis, 1982. 41(2-3): p. 429-431.

Page 85: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

85

269. Moundras, C., Remesy, C., Levrat, M.A., and Demigne, C., Methionine deficiency in rats fed soy protein induces hypercholesterolemia and potentiates lipoprotein susceptibility to peroxidation. Metabolism, 1995. 44(9): p. 1146-1152.

270. Barakat, H.A. and Hamza, A.H., Glycine alleviates liver injury induced by deficiency in methionine and or choline in rats. European Review for Medical and Pharmacology Sciences, 2012. 16(6): p. 728-736.

Page 86: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were
Page 87: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

I

Page 88: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were
Page 89: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Three differently generated salmon protein hydrolysates reveal oppositeeffects on hepatic lipid metabolism in mice fed a high-fat diet

Rita Vik a, Veronika Tillander b, Jon Skorve a, Terhi Vihervaara c, Kim Ekroos c, Stefan E.H. Alexson b,Rolf K. Berge a,d, Bodil Bjørndal a,⇑aDepartment of Clinical Science, University of Bergen, N-5020 Bergen, NorwaybDepartment of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institutet, C1-74, Karolinska University Hospital, SE-141 86 Stockholm, Swedenc Zora Biosciences Oy, Biologinkuja 1, FI-02150 Espoo, FinlanddDepartment of Heart Disease, Haukeland University Hospital, N-5021 Bergen, Norway

a r t i c l e i n f o

Article history:Received 17 October 2014Received in revised form 12 February 2015Accepted 3 March 2015Available online 17 March 2015

Keywords:Marine bioactive peptidesFatty acid compositionDe novo lipogenesisFatty acid desaturaseBeta-oxidationWeight gain

a b s t r a c t

This study investigates the effects of salmon peptide fractions, generated using different enzymatichydrolyzation methods, on hepatic lipid metabolism. Four groups of mice were fed a high-fat diet with20% casein (control group) or 15% casein and 5% of peptide fractions (treatment groups E1, E2 and E4)for 6 weeks. Weight gain was reduced in mice fed E1 and E4-diets compared to control, despite a similarfeed intake. Reduced plasma and liver triacylglycerol levels in E1 and E4-mice were linked to reducedfatty acid synthase (FAS) activity and hepatic expression of lipogenic genes. By contrast, plasma and liverlipids increased in the E2 group, concomitant with increased hepatic FAS activity and D9 desaturase geneexpression. Shotgun lipidomics showed that MUFAs were significantly reduced in the E1 and E4 groups,whereas PUFAs were increased, and the opposite was observed in the E2 group. In conclusion, bioactivepeptides with distinctive properties could potentially be isolated from salmon hydrolysates.� 2015 The Authors. Published by Elsevier Ltd. This is anopenaccess article under the CCBY-NC-ND license

(http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction

The role of dietary protein is to provide the body with essentialamino acids for protein synthesis and energy. Beyond this nutri-tional role, ingested proteins have a wide range of biological func-tions affecting protein, glucose and lipid metabolism,transportation, immune function, blood pressure and hormonalfunctions (Chou, Affolter, & Kussmann, 2012). It has been increas-ingly clear that the dietary source of protein can affect cellularenergy metabolism, and that hydrolyzed peptides can have potentand specific bioactive potential (Erdmann, Cheung, & Schroder,2008). Health benefits from fish consumption have been attributedto the n-3 polyunsaturated fatty acids, in particular eicosapen-taenoic acid (EPA) and docosahexaenoic acid (DHA). However,recent studies have drawn attention towards proteins from marinesources, which are considered valuable bioactive components astheir amino acid composition and protein profile differ from terres-trial sources (Kim, Ngo, & Vo, 2012; Larsen, Eilertsen, & Elvevoll,2011). According to Kelleher, 7 million tons of fish byproductswere discarded as processing waste in 2005 (Kelleher, 2005),

which constituted 50% of the total catch being used for human con-sumption (Rustad, 2003). Fish byproducts can be hydrolyzedenzymatically, using various techniques, liberating potentiallybioactive peptides incorporated in the parental molecule, accord-ing to molecule size, stability in water, refined protein and proteinmix. Rodent studies on fish protein hydrolysates have shown thatmarine proteins exhibit cholesterol-lowering (Shukla et al., 2006),antihypertensive (Je, Park, Kwon, & Kim, 2004; Kim & Mendis,2006), immunomodulating and antioxidant effects (Ahn, Cho, &Je, 2015), in addition to reparative properties in the intestine(Fitzgerald et al., 2005), and increased insulin sensitivity (Pilonet al., 2011). How fish protein hydrolysate may affect lipid metabo-lism in liver is less clear. The liver is the major site of lipid metabo-lism and both fatty acid oxidation and liponeogenesis are carriedout here. These processes are dependent on rate-limiting enzymes,e.g., carnitine palmitoyltransferase (CPT)-1 and 2, acetyl-CoA car-boxylase (ACC), and fatty acid synthase (FAS). Several desaturasesare involved in de novo fatty acid synthesis. The D9 desaturase,encoded by the gene Scd1, generates monounsaturated fatty acids.Along with elongases, the D5 and D6 fatty acid desaturases,encoded by the genes Fads1 and 2, respectively, are important inthe biosynthesis of essential polyunsaturated fatty acids (PUFAs).Regulation of these enzymes will influence fatty acid composition

http://dx.doi.org/10.1016/j.foodchem.2015.03.0110308-8146/� 2015 The Authors. Published by Elsevier Ltd.This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

⇑ Corresponding author. Tel.: +47 55975846; fax: +47 55975890.E-mail address: [email protected] (B. Bjørndal).

Food Chemistry 183 (2015) 101–110

Contents lists available at ScienceDirect

Food Chemistry

journal homepage: www.elsevier .com/locate / foodchem

Page 90: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

and thus important cellular functions, including cell signaling(Nakamura & Nara, 2004).

In this study, we investigated the effect of three different sal-mon peptide fractions (designated E1, E2, and E4), generated usingdifferent enzymatic hydrolyzation and microfiltration methods, onhepatic lipid metabolism in male C57BL/6J mice. We show that dif-ferent peptide compositions, generated from the same species offish, varied in their beneficial effects on body weight and lipidmetabolism.

2. Materials and methods

2.1. Animals and diets

The animal experiments were carried out with ethical permis-sion obtained from the Norway State Board for BiologicalExperiments and followed the Norwegian Research Councils ethi-cal guidelines. Nine to ten weeks old male C57BL/6J mice werehoused, 3 per cage, at constant temperature (22 ± 2 �C) and humid-ity (55 ± 5%), and exposed to a 12 h light–dark cycle with unrest-ricted access to food and tap water. After 1 week ofacclimatization to these conditions, they were divided into 4groups and fed either a high-fat (HF) diet containing 24% (w/w)fat (21.3% lard and 2.7% soy oil) and 20% casein (control group,n = 9), or the HF diet supplemented with protein hydrolysate E1,E2 or E4 from salmon byproducts (a generous gift from MarineBioproducts, Storebø, Norway) (15% casein and 5% peptide, groupE1, E2 and E4, n = 6) ad libitum for 6 weeks. The different salmonpeptide fractions were produced as follows: for fractions E1 andE2, fish material (spine) was treated enzymatically with alkalineprotease and a neutral protease and the resulting protein hydroly-sate was subjected to a second enzymatic treatment. Fraction E1was treated with an Acid Protease A, while fraction E2 was treatedwith the proteolytic enzyme Umamizyme from Aspergillus oryzae.The final hydrolysate was then filtered, using micro- and ultra-fil-tration, and the size distribution of the peptides analyzed. For bothfractions, more than 50% of the final preparation consisted of pep-tides in the range 200–1200 Da and approximately 25% of the pre-paration consisted of peptides below 200 Da. Salmon backbones,including heads, were hydrolyzed with proteolytic Alcalase 2.4 L(Novozymes, Denmark) and subjected to micro- and ultra-filtra-tion and constituted peptide fraction E4. Nearly 60% of the finalpreparation consisted of peptides in the range below 1200 Da.

Amino acid composition of the control and peptide diets isgiven in Supplementary Table 1. Diets were packed airtight andstored at �20 �C until used to prevent lipid oxidation. Mice werehoused in groups of three per cage at a constant temperature of22 ± 2 �C and a dark/light cycle of 12/12 h. Body weights of themice were measured approximately every seventh day and foodintake was measured 3 times during the study. At sacrifice, animalswere fasted overnight, anesthetized with 2% isoflurane (Schering-Plough, Kent, UK) and blood was collected by heart puncture. Theblood was centrifuged, EDTA–plasma separated and frozen priorto further analysis. Livers were collected and immediately frozenin liquid nitrogen and stored at �80 �C prior to further analysis.

2.2. Lipid and fatty acid analysis

Liver lipids were extracted according to Bligh and Dyer (1959),solvents were evaporated under nitrogen and samples re-dissolvedin isopropanol before analysis. Lipids from liver extracts or plasmawere then measured enzymatically on a Hitachi 917 system (RocheDiagnostics, Mannheim, Germany), using kits for analyzing totalTAG (GPO–PAP kit, Roche Diagnostics), cholesterol (CHOD–PAPkit, Roche Diagnostics), and total phospholipids (Diagnostic

Systems GmbH, Holzheim, Germany). Fatty acid composition wasanalyzed in extracted liver lipid, using gas chromatography, asdescribed previously by Grimstad et al. (2012).

2.3. Lipidomic analysis

Liver samples were stored at�80 �C prior to analysis and lipido-mics analysis was performed on 50–100 mg of liver tissue fromeach mouse from two groups, E1 and E2, in addition to the controlgroup. The tissue samples were pulverized with a CP02 CryoPrepDry Pulverization System (Covaris), and resuspended in ice-coldmethanol containing 0.1% butyl-hydroxytoluene (BHT) at a con-centration of 100 mg/mL.

For lipidomics analysis, lipids were extracted from liver homo-genates, using a modified Folch lipid extraction procedure (Ekroos,Chernushevich, Simons, & Shevchenko, 2002). Samples werespiked with known amounts of deuterium-labeled or heptade-canoyl-based synthetic internal standards, serving for quan-tification of the endogenous lipid species, as previously described(Bergan et al., 2013). The samples were stored at �80 �C prior tomass spectrometry analysis.

Molecular glycerophospholipids, glycerolipids, cholesterylesters sphingomyelins and triacylglycerols (TAGs) were analyzedby shotgun analysis on a hybrid triple quadrupole/linear ion trapmass spectrometer (QTRAP 5500, ABSCIEX) equipped with arobotic nanoflow ion source (NanoMate HD, Advion Biosciences)(Stahlman et al., 2009). In shotgun lipidomics a 5 lL volume wasinfused at a concentration of 0.5 lg liver/lL. For TAG analysis,the concentration was diluted to 0.05 lg/lL. The analyses wereperformed in both positive and negative ion modes, using multipleprecursor ion scanning (MPIS) and neutral loss (NL)-based meth-ods (Ekroos et al., 2002, 2003). Sphingolipids were analyzed byreverse phase ultra-high pressure liquid chromatography (RheosAllegro UHPLC, Flux Instruments AG), using an Acquity BEH C18,2.1 � 50 mm column with a particle size of 1.7 lm (Waters,Milford, Massachusetts, USA) coupled to a hybrid triplequadrupole/linear ion trap mass spectrometer (QTRAP 5500,ABSCIEX). A 25 min gradient, using 10 mM ammonium acetate inwater with 0.1% formic acid (mobile phase A) and 10 mM ammo-nium acetate in acetonitrile:2-propanol (4:3, v/v), containing0.1% formic acid (mobile phase B), was used. The column tempera-ture was set to 60 �C and the flow rate to 500 lL/min. 10 lL sam-ples were injected. Sphingolipids were monitored, using multiplereactions monitoring (MRM) as described by Merrill, Sullards,Allegood, Kelly, and Wang (2005).

The MS data files were processed, using Lipid Profiler™ andMultiQuant™ software for producing a list of lipid names and peakareas. The individual lipids measured can be found inSupplementary Table 2. Masses and counts of detected peaks wereconverted into a list of corresponding lipid names. Lipids were nor-malized to their respective internal standard (Bergan et al., 2013)and tissue weight to retrieve their concentrations. Data were ana-lyzed, using the software Tableau Desktop 7.0 and the percentagedifferences between the groups (E1 vs. controls and E2 vs. controls)were estimated in pairwise comparisons, using a Hodges–Lehmannestimator, and the significances were calculated using Wilcoxonrank-sum t-test.

2.4. Hepatic enzyme activities

Liver samples were homogenized and a post-nuclear fractionwas prepared as previously described (Berge, Flatmark, &Osmundsen, 1984). The activity of CPT-1 was measured in the pres-ence and absence ofmalonyl-CoA, as previously described (Vik et al.,2014). The assay conditions for CPT-2 were identical to CPT-1, apartfrom some changes in the reaction mix; BSA and KCN were

102 R. Vik et al. / Food Chemistry 183 (2015) 101–110

Page 91: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

exchanged with 0.01% Triton X-100, and 35 lg of total protein wereused for the assays. Acyl-CoA oxidase (ACOX)-1 activity was mea-sured, using 20 lg protein, by a coupled assay, described by Small,Burdett, and Connock (1985), with some modifications (Madsenet al., 1999). Glycerol-3-phosphate acyltransferase (GPAT) activitywas measured, as described by Skorve et al. (1993).

2.5. RNA isolation, cDNA synthesis, and real-time PCR

Total RNA from liver tissue was purified, using theMagMax totalRNA isolation system (Applied Biosystems, Carlsbad, CA, USA) aftertissue homogenization. The quantity of the RNA was measuredspectrophotometrically, using a NanoDrop 1000 (NanoDropProducts,Wilmington, DE, USA) and the quality of the RNAwas ana-lyzed, using the Experion Automated Electrophoresis System (Bio-Rad Laboratories, Hercules, CA, USA). Quality limit for further useof RNA was set to a R/Q value of P7 (out of 10). cDNA was synthe-sized with 500 ng of RNA per reaction, using High Capacity RNA tocDNAMastermix (Applied Biosystems). Genes of interest were ana-lyzed in individual samples from liver, using SYBR Green geneexpression assays, and using primers for acetyl-CoA carboxylasealpha (Acaca, 50acctgtacaagcagtgtgggct, 30cacatggcctggcttggaggg),acyl-CoA thioesterase (Acot1, 50ctggcgcatgcaggatc, 30ggcacttttcttg-gatagctcc), fatty acid desaturase 1 (Fads1, 50ggctcccgggtcatcag,30acccttgttgatgtggaatgc), (Fads2, 50ggacataaagagcctgcatgtg, 30gggcaggtatttcagcttcttc), (Fas, 50ggcatcattgggcactcctt, 30gctgcaag-cacagcctctct), 3-hydroxy-3-methylglutaryl-coenzyme A reductase(Hmgcr, 50ccggcaacaacaagatctgtg, 30atgtacaggatggcgatgca), fattyacid binding protein, liver (L-fabp, 50ccatgactggggaaaaagtc, 30gc-ctttgaaagttgtcaccat), stearoyl-CoA desaturase (Scd1, 50acgggctccggaaccgaagt, 30ctggagatctcttggagcatgtggg). SYBRGreen primerswereused at concentrations ranging from100 to 200 nMand runwith thePower SYBR GreenMasterMix (Applied Biosystems) in an ABI Prism7500 sequence detection system. Gene expression was estimated,using the average threshold (Ct) value, in triplicate calculated, usingthe 2�DDCtmethod, according to Livak and Schmittgen (2001), usinghypoxanthine phosphoribosyltransferase 1 (Hprt1, 50ggtgaaaaggac-ctctcgaagtg, 30atagctaagggcatatccaacaac) as reference gene and oneindividual sample in the high-fat group as calibrator.

2.6. Statistics

All values are presented as means ± standard deviation (SD).One-way ANOVA was used for analysis of differences betweengroups, followed by Dunnett’s multiple comparison test betweenall groups vs. control when data followed Gaussian distributionand the Kruskal–Wallis test, with Dunn’s multiple comparison test,was used when data were not normally distributed. Significancewas set to (P < 0.05). All statistics were calculated usingGraphPad Prism 6 for Mac OS X with the exception of the lipido-mics data where software Tableau Desktop 7.0 was used.

3. Results

3.1. Body weight was influenced by dietary intake of salmon peptidefractions

C57BL/6J mice, fed high-fat diets with 15% casein and 5% (w/w)of salmon peptide fractions E1 or E4, demonstrated lower bodyweight gain throughout the 6 weeks feeding period (Fig. 1A), aswell as a significantly lower total weight gain than did control micefed 20% (w/w) casein as protein source (Fig. 1B). By contrast, micefed peptide E2 had a body weight curve and total weight gain moresimilar to the control group. Feed intake was measured 3 timesduring the experiment, and the average feed intakes were similar

in control, E2 and E4-fed mice (Fig. 1C), while it was higher inpeptide E1-fed mice, despite the low weight gain in this group. Asmall, but significant reduction was observed in the liver index(% liver weight/body weight) of E4-fed mice compared to controls(Fig. 1D).

3.2. The salmon peptide fractions differentially affected plasma lipidsbut not bile acid levels

Plasma lipids were measured in samples from fasted animals.While mice fed peptides E1 and E4 demonstrated significantlylower plasma TAG levels than did the control, the TAG level in micefed peptide E2 was elevated (Fig. 2A). The peptide E2 group alsodemonstrated elevated levels of total cholesterol, phospholipidsand HDL-cholesterol compared to the control (Fig. 2B–D). Nochange was observed in LDL-cholesterol or the HDL/LDL ratio inany of the feeding groups (Fig. 2E and F), but non-esterified fattyacids showed a tendency to increase in the E4 group (Fig. 2G, con-trol vs. E4, Mann–Whitney, P-value = 0.044). There was a smallincrease in plasma bile acids in the E4 group, when measured inplasma samples pooled from 2 to 4 animals (Fig. 2H). However,statistical significance could not be analyzed (n = 2–3).

3.3. The effect of salmon peptide fractions on hepatic enzymes

As the studied salmon peptide fractions affected weight gainand lipid levels differently in C57BL/6J mice, we analyzed the activ-ity of hepatic enzymes involved in lipid catabolism and synthesis.CPT-1 is the rate-limiting enzyme in fatty acid import intomitochondria for b-oxidation. In mice fed E1, no change wasobserved in the activity of CPT-1 (Fig. 3A), or its % inhibition bymalonyl-CoA (Fig. 3B). CPT-2 activity was also unchanged, furthersupporting no influence on mitochondrial b-oxidation in this group(Fig. 3C). The activity of GPAT, involved in the synthesis of TAG,was unchanged (data not shown). However, de novo lipogenesiscould be affected, as FAS activity tended to be reduced in E1-fedmice compared to control-mice (Fig. 3D). In E4 mice, a potentialconcomitant increase in b-oxidation and reduction in lipogenesiswas observed; CPT-1 and -2 activities demonstrated small, but sig-nificantly higher levels than the control, suggesting more CPT-mediated mitochondrial import of fatty acids in these mice. Also,FAS activity was significantly lower in E4-fed mice than in controlmice. By contrast, E2 mice demonstrated a significantly higher FASactivity than did control mice, suggesting increased de novolipogenesis in this group, while CPT-1 and -2 activity wereunchanged.

We further analyzed hepatic expression of genes involved infatty acid metabolism. The liver fatty acid binding protein (L-fabp)was not affected by the diets (Fig. 4A). Acot1, which regulates thecytosolic levels of acyl-CoA, CoASH and free fatty acids, was signifi-cantly increased in the E2 and E4 groups compared to control mice(Fig. 4B). In agreement with the enzyme activity data, Fas geneexpression was significantly reduced in the E1 and E4 groups com-pared to control (Fig. 4C). The expression of another importantgene in fatty acid synthesis, Acaca, was significantly decreased inthe E1 group compared to the control (Fig. 4D). The D9 desat-urase (Scd1) mRNA level tended to be increased in livers fromE2-fed mice, supporting an increase in de novo lipogenesis in thisgroup (Fig. 4E). There was also indication of a differential effecton D5 and D6 desaturation of fatty acids, as Fads1was significantlylower expressed in the E4 group vs. the control group (Fig. 4F)and Fads1 and Fads2 tended to be lower in the E1 group comparedto the control (Fig. 4G). HMG-CoA reductase (Hmgcr), the rate-limiting enzyme in cholesterol synthesis, was significantly reducedin the E1 and E4 groups compared to the control (Fig. 4H).

R. Vik et al. / Food Chemistry 183 (2015) 101–110 103

Page 92: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

3.4. A differential effect of salmon peptide fractions on hepatic fattyacid composition

To determine if the differently processed peptide fractionscould influence fatty acid composition, total fatty acids in liverwere analyzed. Saturated fatty acids (SFAs) were unaffected in allpeptide diet groups compared to the control. Largely similar effectson monounsaturated fatty acid (MUFA) and polyunsaturated fattyacid (PUFA) composition were observed in the E1 and E4 groups:While MUFAs tended to be reduced in the E1 group, and were sig-nificantly reduced in the E4 group (Table 1), n-3 PUFAs were sig-nificantly increased in the E4 group, and n-6 PUFAs increased inthe E1 group compared to the control (Table 1). The opposite effectwas observed in the liver of E2 mice, where MUFAs increasedstrongly, while n-3 and n-6 PUFAs were significantly reduced com-pared to control mice. In line with these results, the D9 desaturaseindex, calculated by the ratio C16:1n-7/C16:0 or the ratio C18:1n-9/C18:0, was increased in the E2 group compared to the control.The D5 desaturase (n-6) index, calculated by the ratio C20:4n-6/C20:3n-6, increased in the E1 and E4 group, while the D6 desat-urase (n-3) index, calculated by the ratio C20:5n-3/C20:4n-3, wassignificantly increased only in E4 (Table 1). In addition, the D5desaturase (n-3) index was significantly increased in the E4 groupcompared to the control, while no change was seen in the D6desaturase (n-6) index (data not shown).

3.5. Comparison of the effect of salmon peptide fractions on hepaticlipids

In the enzymatic measurements of lipids from liver extracts, wefound that TAG tended to be reduced by peptides E1 and E4 (E1 vs.

control, student’s t-test, P-value = 0.042) (Supplementary Table 3).Surprisingly, cholesterol showed a small but significant increasein E4 vs. control, while phospholipids were increased by both E1and E4. However, the total hepatic lipid levels (TAG, cholesteroland PL) were unchanged in E1 and E4 compared to the control. Inthe livers of E2 mice, both TAG and total hepatic lipids were signifi-cantly increased (Supplementary Table 3), in line with plasma lipidresults.

Since the E1 and E2 salmon peptide fractions were generatedfrom the same raw-material, but showedopposite effects on hepaticlipid metabolism, liver samples frommice fed these diets were fur-ther compared to controls in three separate analytical runs in shot-gun, sphingolipid and TAG lipidomics. Fig. 5A shows a summary ofthe effects of these two salmon protein hydrolysate fractions atthe level of the major lipid classes. In line with the enzymatic mea-surement, the relative difference (%) of TAG was significantly ele-vated in the E2 group vs. control, and tended to decrease in the E1group vs. control. Striking differences were also seen with the cera-mides (Cer) and glycerophosphoethanolamines (PE), whichincreased significantly in E1 vs. control, diacylglycerols (DAG),which increased in E2 vs. control, and glycosyl/galactosylceramide(Glc/GalCer), which were specifically reduced in E1 vs. control.The difference in TAG in the E1 group compared to the controlwas seen as a reduction in all species of TAG, whereas the E2-dietprimarily increased the TAG species containing short, saturated ormonounsaturated fatty acids compared to the control (Fig. 5B).The peptide fractions had a prominent different effect on hepaticceramide levels. The relative differences of all d18:1 ceramideswereincreasedwith the E1 diet compared to the control,while largely theopposite effect was observed in the E2 group, apart from a sta-tistically significant increase in Cer (d18:1/18:0) (Fig. 5C).

A

C

B

Feed intake

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

2

4

6

g/da

y/m

ouse

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

2

4

6

liver

/bod

y w

eigh

t(%

)

Liver index

**

Weight gain

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

5

10

15

gram ** ***

Body weight

0 8 14 22 27 3625

30

35

40

gram

ControlPeptide E1Peptide E2Peptide E4

D

Fig. 1. (A) Body weight curve, (B) weight gain, (C) feed intake, and (D) hepatic index in mice fed a high-fat casein diet (control, n = 9) or a diet where 5% of the protein sourcewas replaced with 5% of three different salmon protein hydrolysates E1, E2 or E4 (n = 6). Data are shown as means with SD and dissimilar letters indicate significantlydifferent values (P < 0.05). Statistical significance could not be calculated in (C) since data from 3 mice were pooled.

104 R. Vik et al. / Food Chemistry 183 (2015) 101–110

Page 93: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

TAG

mm

ol/L

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0.0

0.5

1.0

1.5

2.0

**

**

**

Phospholipids

mm

ol/L

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

1

2

3

4

5 **

LDL

mm

ol/L

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0.0

0.1

0.2

0.3

0.4

NEFA

mm

ol/L

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0.0

0.1

0.2

0.3

0.4

Cholesterol

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

1

2

3

4

5

mm

ol/L

***

HDL

mm

ol/L

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

1

2

3

4 *

HDL/LDL

Rat

io

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

5

10

15

20

25

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0.0

0.5

1.0

1.5

2.0

μmol

/L

Bile acids

BA

DC

FE

HG

Fig. 2. Plasma lipids in mice fed a high-fat casein diet (control, n = 8) or a diet where 5% of the protein source was replaced with 5% of three different salmon proteinhydrolysates, E1, E2, and E4, respectively (n = 6). (A) Triacylglycerol (TAG), (B) cholesterol, (C) phospholipids, (D) high-density lipoprotein (HDL), (E) low-density lipoprotein(LDL), (F) HDL/LDL ratio, and (G) non-esterified fatty acids (NEFA). Bile acids in pooled plasma samples from 2 to 4 mice (n = 2–3) are shown in (G). Data are shown as meanswith SD and values significantly different from control were determined by one-way ANOVA (A–C and F) or Kruskal–Wallis test (D, E and G), ⁄P < 0.05, ⁄⁄P < 0.01, ⁄⁄⁄P < 0.001.Statistics could not be performed in (H).

R. Vik et al. / Food Chemistry 183 (2015) 101–110 105

Page 94: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

4. Discussion

Based on recent studies, fish proteins and peptides have diversebioactive properties (Kim & Mendis, 2006). The purpose of ourstudy was to evaluate the effects of three different fractions of sal-mon protein hydrolysate on body weight development and plasmalipids, as well as hepatic lipid metabolism, as the liver is the mainorgan maintaining lipid homeostasis. The novel findings of ourstudy were the divergent effects on body weight, plasma and hep-atic TAG levels, hepatic lipogenesis and b-oxidation, and hepaticfatty acid composition by peptide fractions generated using differ-ent enzymatic methods.

The groups fed peptide fractions E1 and E4 exhibited a lowerweight gain throughout the study, despite the same or higher feedintake than control. Furthermore, both E1 and E4-fed groups hadreduced plasma TAG levels and a tendency to reduced liver TAG.In E1 mice, this was linked to decreased expression of the Fasand Acaca genes, as well as a tendency to decrease FAS activity.The reduction in these rate-limiting steps in fatty acid synthesisindicates a repression of de novo lipogenesis by the E1 peptide.Similar to E1-fed mice, key enzymes in lipogenesis were signifi-cantly reduced by E4, and in addition, an increased CPT-1 and -2activity suggested that increased fatty acid oxidation could havecontributed to the lower body weight and plasma TAG observedin this group.

The generation of the E2 peptide fraction differed from that ofthe E1 peptide fraction in the secondary enzymatic treatment,and opposite effects were observed in these treatment groups.E2-fed mice had a body weight gain curve similar to that of

control-fed mice, and importantly, displayed increased hepaticTAG, as well as plasma TAG, cholesterol, and phospholipid levels.FAS activity was significantly increased by E2, and thus the differ-ences in plasma and liver TAG could partly be explained by anopposite regulation of fatty acid synthesis in mice fed the E2 vs.the E1 and E4 peptide fractions. In support of this, the increasedD9 index, and the tendency to increase in hepatic gene expressionof Scd1, the enzyme performing the crucial D9 unsaturation stepduring fatty acid synthesis, indicates stimulated fatty acid synthe-sis in E2-fed mice. The GPAT activity was not altered in the inter-vention groups; thus TAG synthesis was probably not affected bythe diets.

A number of studies, using high doses of fish protein or proteinhydrolysate from Atlantic salmon (15–20% of diet), have demon-strated reduced plasma cholesterol levels in Wistar rats (Hosomiet al., 2011; Liaset et al., 2009). This has been linked to increasedplasma bile acid levels, and cholesterol clearance through the bile.However, in a number of comparable studies in rabbits and mice,no influence on plasma cholesterol levels has been shown(Bergeron & Jacques, 1989; Bjørndal et al., 2013). In line with this,no plasma cholesterol-reducing effects or increases in plasma bileacids were observed in the treatment groups compared to control,despite a reduction in the gene expression of Hmgcr, involved incholesterol biosynthesis, in the E1 and E4 groups. This could eitherbe due to the animal model or the lower dose of salmon proteinhydrolysate used compared to previous studies.

Different sources of protein are assumed to differ in digestibil-ity, thus contributing unequally to energy supply; other proteinsor peptides are believed to suppress appetite (Nishi, Hara, Asano,

CPT-1 activity

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

1

2

3

4nm

ol/m

g/m

in

FAS activity

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0.0

0.2

0.4

0.6

nmo

l/mg/

min

*

BA

DC CPT-2 activity

Control

Peptid

e E1

Peptid

e E2

Peptid

e E40

1

2

3

4

nmol

/mg

/min

**

% inhibition

Control

Peptid

e E1

Peptid

e E2

Peptid

e E4

0

10

20

30

40

50

% in

hibi

tion

by

Mal

onyl

-Co

A

Fig. 3. Hepatic enzyme activities in mice fed a high-fat casein diet (control) or a diet where 5% of the protein source was replaced with 5% of three different salmon proteinhydrolysates, E1, E2, and E4, respectively. (A) Carnitine palmitoyltransferase-1 (CPT-1) activity, (B) % inhibition of CPT-1 activity by malonyl-CoA, (C) carnitinepalmitoyltransferase-2 (CPT-2) activity and (D) fatty acid synthase (FAS) activity. Data are shown as means with SD (n = 6) and values significantly different from control weredetermined by one-way ANOVA (⁄P < 0.05, ⁄⁄P < 0.01).

106 R. Vik et al. / Food Chemistry 183 (2015) 101–110

Page 95: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Fig. 4. Hepatic gene expressions in mice fed a high-fat casein diet (control) or a diet where 5% of the protein source was replaced with 5% of three different salmon proteinhydrolysates, E1, E2, and E4, respectively. (A) Fatty acid binding protein, liver (L-fabp), (B) acyl-coenzyme A thioesterase 1 (Acot1), (C) fatty acid synthase (Fas), (D) acetyl-coenzyme A carboxylase alpha (Acaca), (E) stearyl-coenzyme A desaturase (Scd1), (F) fatty acid desaturase 1 (Fads1), (G) fatty acid desaturase 2 (Fads2), and (H) 3-hydroxy-3-methylglutaryl CoA reductase (Hmgcr). Data are shown as means with SD (n = 6) and dissimilar letters indicate significantly different values.

R. Vik et al. / Food Chemistry 183 (2015) 101–110 107

Page 96: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

& Tomita, 2003), while some are suggested to increase satiety. Inaddition, a previous study showed that salmon protein uniquelyprotects against body weight gain, independently of feed intake(Pilon et al., 2011). In support of this, feed intake in the four groupswas unable to explain the divergent effect on body weight. Thus, inthe present study, the distinct peptide or amino acid compositionof each salmon protein hydrolysate-fraction may explain the varia-tion in body weight gain between the treatment groups(Supplementary Table 1). The degree of hydrolysis of corn glutenmeal has been reported to affect bioavailability and weight gainin rats (Jin et al., 2014); however, in our study, the three differenthydrolysates had similar compositions with regard to peptidesequence size, indicating similar degrees of hydrolysis.

Dietary proteins may play a role in mechanisms affecting fattyacid composition (Bjørndal et al., 2013; Leveille, Tillotson, &Sauberlich, 1963). The amino acid composition of the diet has beenshown to influence lipogenesis and desaturation, as tyrosinedown-regulates hepatic D6 activity (Peluffo, Nervi, Gonzalez, &

Brenner, 1984), and arginine and leucine can regulate the geneexpression of Scd1 and Fas in muscle and adipose tissue (Madeiraet al., 2014). Here, the amino acid composition did not differ to alarge extent since they were all dominated by amino acids fromcasein. However, tyrosine and leucine were higher in the E2 dietcompared to the control, E1 and E4, while arginine was reducedin the E1 and E4 diets compared to the control and E2. In line withthis, the E1 and E4 groups displayed similar patterns in fatty acidcomposition, including an increase in the fatty acid ratios 18:3n-6/18:2n-6 and 20:4n-6/20:3n-6 compared to the control. Theseratios are commonly used as an index for the activities of D6-and D5 desaturase, respectively. As also reported by others(Sjogren et al., 2008), the D5 and D6 desaturation indices are influ-enced by a number of pathways, and failed to reflect the expressionof Fads1 and Fads2. In contrast, Scd1 expression correlated with theD9 desaturase index in liver. Thus, concomitant with increasedScd1 expression in E2-mice, an increase in monounsaturated fattyacids and the D9 desaturase index was observed. Interestingly,while hepatic MUFA increased, n-3 PUFA and n-6 PUFAs decreasedin the E2 group. The main product of D9 desaturase, oleoyl-CoA(C18:1), is mainly used to generate TAG (Mauvoisin & Mounier,2011), and this was reflected in the predominant increase in TAGspecies containing short MUFAs. Studies investigating the effectof MUFA supplementation on plasma lipid levels in humans areinconclusive; however, in rats, a high MUFA amount in a diet con-taining 40% energy from fat was shown to increase plasma totalcholesterol, TAG and phospholipid levels as compared to a lowdietary MUFA amount (Chang & Huang, 1998). The current studysuggests that an increased hepatic expression of Scd1, and acorresponding increase in MUFAs incorporated into TAG could leadto increased VLDL release.

The E1 and E4 diets resulted in increased hepatic PUFA levels,indicating increased biosynthesis of PUFAs at the expense ofMUFAs. Dietary and endogenous fatty acids, in particular PUFAs,are involved in metabolic regulation of lipid and glucose metabo-lism. Thus, the differential peptide-effects on lipid levels couldhave been reinforced by their opposite regulation of fatty acidsimportant in cell signaling.

Table 1Hepatic fatty acid composition mice fed a casein control diet or diets with 5% of threedifferent salmon protein hydrolysates for 4 weeksa.

Fatty acids Control E1 E2 E4

SFAs 34.8 ± 1.4 36.3 ± 0.9 34.2 ± 0.7 35.3 ± 1.3MUFAs 23.8 ± 6.4 17.8 ± 1.8 31.1 ± 4.1⁄⁄ 18.1 ± 4.4n-3 PUFAs 9.8 ± 1.4 10.7 ± 0.9 7.7 ± 1.1⁄⁄⁄ 14.1 ± 1.4⁄⁄⁄

n-6 PUFAs 31.4 ± 3.8 34.9 ± 0.7 26.8 ± 2.5⁄⁄⁄ 32.4 ± 1.8bD9-desat. (C16:0) 0.06 ± 0.03 0.05 ± 0.01 0.10 ± 0.02⁄⁄ 0.05 ± 0.02bD9-desat. (C18:0) 2.1 ± 1.3 1.2 ± 0.1 3.1 ± 0.9⁄⁄ 1.3 ± 0.5cD5-desat. 9.5 ± 2.1 13.5 ± 1.2⁄ 9.3 ± 1.3 17.6 ± 4.6⁄⁄⁄dD6-desat. 0.08 ± 0.02 0.10 ± 0.02 0.11 ± 0.02 0.12 ± 0.03⁄

Data are shown as means ± SD (n = 6–9).Values significantly different from control were determined by one-way ANOVA(⁄P < 0.05, ⁄⁄P < 0.01, ⁄⁄⁄P < 0.001).Abbreviations: SFAs, saturated fatty acids; MUFAs, monounsaturated fatty acids,PUFAs, polyunsaturated fatty acids.

a wt%.b Delta9 desaturase index (C16:1n-7/C16:0 and C18:1n-9/C18:0).c Delta5 desaturase index (C20:4n-6/C20:3n-6).d Delta6 desaturase index (C20:5n-3/C20:4n-3).

Fig. 5. Summary of lipid classes (A), TAG species (B), and ceramide species (C) in liver, presented as relative molar percentage differences between treatment groups andcontrol, in mice fed a high-fat casein diet (control) or the replacement with 5% of the protein source with 5% of peptide fraction E1 or E2 (n = 6). Significance is indicated as⁄P < 0.05 and ⁄⁄P < 0.01.

108 R. Vik et al. / Food Chemistry 183 (2015) 101–110

Page 97: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Assessment of liver lipids, using lipidomics, also revealed a sig-nificantly elevated level of ceramides in the E1 group. These waxylipid molecules can induce both inflammation and insulin resis-tance (Summers, 2006). Increased ceramide levels in adipose tissuehave been linked to higher levels of liver fat (Kolak et al., 2012);however, in our study, TAG decreased while ceramides increasedin the liver of E1-fed mice. A reduction in diacylglycerols (DAG)in the E1 group could be of significance in view of its physiologicalimportance in activating protein kinase C (PKC). PKC phos-phorylates the hydroxyl-groups of serine and threonine amino acidresidues on several important target proteins and modulatesphysiological processes in all cells. In our study, DAG and TAG dis-played a simultaneous decrease in the E1 group and increase in theE2 group, relative to control. The significant increase of DAG in thepeptide E2 group, combined with increased gene expression ofScd1, but not in ceramide levels, is similar to findings in the liverof patients with non-alcoholic fatty liver disease (Kotronen et al.,2009). In further studies, it will be interesting to evaluate the effectof the E1 and E4 fish protein hydrolysate fractions on animal mod-els, on NAFLD, as well as obesity and insulin resistance.

5. Conclusions

The three different peptide fractions from salmon proteinhydrolysate show diverse effects on weight gain, plasma and liverlipids and lipid synthesis, suggesting that protein products that areenzymatically hydrolyzed exhibit distinct and, in some cases,opposite effects. Thus, prior to their potential use as dietary sup-plements, it is of importance to analyze protein hydrolysates thor-oughly to exclude possible negative metabolic effects. Pre-digestion of proteins gives the opportunity to optimize nutritionalvalue and bioavailability of the peptides, thus obtaining a morespecific product regarding its use, either as a balanced food supple-ment, food aid or simply healthy food.

Acknowledgments

The authors wish to thank Svein Krüger, Randi Sandvik, LivKristine Øysæd, Kari Williams and Kari Mortensen for valuabletechnical assistance. We also wish to thank Eline Milde Nævdaland the staff at the animal facility. We also thank MarineBioproducts, Storebø, Norway for providing the salmon proteinhydrolysates. This work was supported by NordForsk under theNordic Centers of Excellence program in Food, Nutrition, andHealth; Project (070010) ‘‘MitoHealth’’.

Appendix A. Supplementary data

Supplementary data associated with this article can be found, inthe online version, at http://dx.doi.org/10.1016/j.foodchem.2015.03.011.

References

Ahn, C. B., Cho, Y. S., & Je, J. Y. (2015). Purification and anti-inflammatory action oftripeptide from salmon pectoral fin byproduct protein hydrolysate. FoodChemistry, 168, 151–156.

Bergan, J., Skotland, T., Sylvanne, T., Simolin, H., Ekroos, K., & Sandvig, K. (2013). Theether lipid precursor hexadecylglycerol causes major changes in the lipidome ofHEp-2 cells. PLoS One, 8(9), e75904.

Berge, R. K., Flatmark, T., & Osmundsen, H. (1984). Enhancement of long-chain acyl-CoA hydrolase activity in peroxisomes and mitochondria of rat liver byperoxisomal proliferators. European Journal of Biochemistry, 141(3), 637–644.

Bergeron, N., & Jacques, H. (1989). Influence of fish protein as compared to caseinand soy protein on serum and liver lipids, and serum lipoprotein cholesterollevels in the rabbit. Atherosclerosis, 78(2–3), 113–121.

Bjørndal, B., Berge, C., Ramsvik, M., Svardal, A., Bohov, P., Skorve, J., et al. (2013). Afish protein hydrolysate alters fatty acid composition in liver and adipose tissue

and increases plasma carnitine levels in a mouse model of chronicinflammation. Lipids Health Disease, 12, 143.

Bligh, E. G., & Dyer, W. J. (1959). A rapid method of total lipid extraction andpurification. Canadian Journal of Biochemistry an Physiology, 37(8), 911–917.

Chang, N. W., & Huang, P. C. (1998). Effects of the ratio of polyunsaturated andmonounsaturated fatty acid to saturated fatty acid on rat plasma and liver lipidconcentrations. Lipids, 33(5), 481–487.

Chou, C. J., Affolter, M., & Kussmann, M. (2012). A nutrigenomics view of proteinintake: Macronutrient, bioactive peptides, and protein turnover. Progress inMolecular Biology and Translational Science, 108, 51–74.

Ekroos, K., Chernushevich, I. V., Simons, K., & Shevchenko, A. (2002). Quantitativeprofiling of phospholipids by multiple precursor ion scanning on a hybridquadrupole time-of-flight mass spectrometer. Analytical Chemistry, 74(5),941–949.

Ekroos, K., Ejsing, C. S., Bahr, U., Karas, M., Simons, K., & Shevchenko, A. (2003).Charting molecular composition of phosphatidylcholines by fatty acid scanningand ion trap MS3 fragmentation. Journal of Lipid Research, 44(11),2181–2192.

Erdmann, K., Cheung, B. W., & Schroder, H. (2008). The possible roles of food-derived bioactive peptides in reducing the risk of cardiovascular disease. Journalof Nutritional Biochemistry, 19(10), 643–654.

Fitzgerald, A., Rai, P., Marchbank, T., Taylor, G., Ghosh, S., Ritz, B., et al. (2005).Reparative properties of a commercial fish protein hydrolysate preparation. Gut,54, 775–781.

Grimstad, T., Bjorndal, B., Cacabelos, D., Aasprong, O. G., Janssen, E. A., Omdal, R.,et al. (2012). Dietary supplementation of krill oil attenuates inflammation andoxidative stress in experimental ulcerative colitis in rats. Scandinavian Journal ofGastroenterology, 47(1), 49–58.

Hosomi, R., Fukunaga, K., Arai, H., Kanda, S., Nishiyama, T., & Yoshida, M. (2011).Fish protein decreases serum cholesterol in rats by inhibition of cholesterol andbile acid absorption. Journal of Food Science, 76(4), H116–H121.

Je, J. Y., Park, P. J., Kwon, J. Y., & Kim, S. K. (2004). A novel angiotensin I convertingenzyme inhibitory peptide from Alaska pollack (Theragra chalcogramma) frameprotein hydrolysate. Journal of Agricultural and Food Chemistry, 52(26),7842–7845.

Jin, J., Ma, H., Zhou, C., Luo, M., Liu, W., Qu, W., et al. (2014). Effect of degree ofhydrolysis on the bioavailability of corn gluten meal hydrolysates. Journal of theScience of Food and Agriculture.

Kelleher, K. (2005). Discards in the world’s marine fisheries: An update. Rome, Italy:Food and Agriculture Organization of the United Nations (p. 131).

Kim, S.-K., & Mendis, E. (2006). Bioactive compounds from marine processingbyproducts – A review. Food Research International, 39, 383–393.

Kim, S. K., Ngo, D. H., & Vo, T. S. (2012). Marine fish-derived bioactive peptides aspotential antihypertensive agents. Advances in Food and Nutrition Research, 65,249–260.

Kolak, M., Gertow, J., Westerbacka, J., Summers, S. A., Liska, J., Franco-Cereceda, A.,et al. (2012). Expression of ceramide-metabolising enzymes in subcutaneousand intra-abdominal human adipose tissue. Lipids in Health and Disease,11(115).

Kotronen, A., Seppanen-Laakso, T., Westerbacka, J., Kiviluoto, T., Arola, J., Ruskeepaa,A. L., et al. (2009). Hepatic stearoyl-CoA desaturase (SCD)-1 activity anddiacylglycerol but not ceramide concentrations are increased in thenonalcoholic human fatty liver. Diabetes, 58(1), 203–208.

Larsen, R., Eilertsen, K. E., & Elvevoll, E. O. (2011). Health benefits of marine foodsand ingredients. Biotechnology Advances, 29(5), 508–518.

Leveille, G. A., Tillotson, J. A., & Sauberlich, H. E. (1963). Fatty acid composition ofplasma and liver lipid components as influenced by diet in the growing chick.Journal of Nutrition, 81, 357–362.

Liaset, B., Madsen, L., Hao, Q., Criales, G., Mellgren, G., Marschall, H. U., et al. (2009).Fish protein hydrolysate elevates plasma bile acids and reduces visceral adiposetissue mass in rats. Biochimica et Biophysica Acta, 1791(4), 254–262.

Livak, K. J., & Schmittgen, T. D. (2001). Analysis of relative gene expression datausing real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods,25(4), 402–408.

Madeira, M. S., Pires, V. M., Alfaia, C. M., Luxton, R., Doran, O., Bessa, R. J., et al.(2014). Combined effects of dietary arginine, leucine and protein levels on fattyacid composition and gene expression in the muscle and subcutaneous adiposetissue of crossbred pigs. British Journal of Nutrition, 111(9), 1521–1535.

Madsen, L., Rustan, A. C., Vaagenes, H., Berge, K., Dyroy, E., & Berge, R. K. (1999).Eicosapentaenoic and docosahexaenoic acid affect mitochondrial andperoxisomal fatty acid oxidation in relation to substrate preference. Lipids,34(9), 951–963.

Mauvoisin, D., & Mounier, C. (2011). Hormonal and nutritional regulation of SCD1gene expression. Biochimie, 93(1), 78–86.

Merrill, A. H., Jr., Sullards, M. C., Allegood, J. C., Kelly, S., & Wang, E. (2005).Sphingolipidomics: High-throughput, structure-specific, and quantitativeanalysis of sphingolipids by liquid chromatography tandem massspectrometry. Methods, 36(2), 207–224.

Nakamura, M. T., & Nara, T. Y. (2004). Structure, function, and dietary regulation ofdelta6, delta5, and delta9 desaturases. Annual Review of Nutrition, 24, 345–376.

Nishi, T., Hara, H., Asano, K., & Tomita, F. (2003). The soybean beta-conglycinin beta51–63 fragment suppresses appetite by stimulating cholecystokinin release inrats. Journal of Nutrition, 133(8), 2537–2542.

Peluffo, R. O., Nervi, A. M., Gonzalez, M. S., & Brenner, R. R. (1984). Effect of differentamino acid diets on delta 5, delta 6 and delta 9 desaturases. Lipids, 19(2),154–157.

R. Vik et al. / Food Chemistry 183 (2015) 101–110 109

Page 98: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Pilon, G., Ruzzin, J., Rioux, L. E., Lavigne, C., White, P. J., Froyland, L., et al. (2011).Differential effects of various fish proteins in altering body weight, adiposity,inflammatory status, and insulin sensitivity in high-fat-fed rats. Metabolism,60(8), 1122–1130.

Rustad, T. (2003). Utilisation of marine by-products. Electronic Journal ofEnvironmental, Agricultural and Food Chemistry, 2, 458–463.

Shukla, A., Bettzieche, A., Hirche, F., Brandsch, C., Stangl, G. I., & Eder, K. (2006).Dietary fish protein alters blood lipid concentrations and hepatic genesinvolved in cholesterol homeostasis in the rat model. British Journal ofNutrition, 96(4), 674–682.

Sjogren, P., Sierra-Johnson, J., Gertow, K., Rosell, M., Vessby, B., de Faire, U., et al.(2008). Fatty acid desaturases in human adipose tissue: Relationships betweengene expression, desaturation indexes and insulin resistance. Diabetologia,51(2), 328–335.

Skorve, J., al-Shurbaji, A., Asiedu, D., Bjorkhem, I., Berglund, L., & Berge, R. K. (1993).On the mechanism of the hypolipidemic effect of sulfur-substituted

hexadecanedioic acid (3-thiadicarboxylic acid) in normolipidemic rats. Journalof Lipid Research, 34(7), 1177–1185.

Small, G. M., Burdett, K., & Connock, M. J. (1985). A sensitive spectrophotometricassay for peroxisomal acyl-CoA oxidase. The Biochemical Journal, 227(1),205–210.

Stahlman, M., Ejsing, C. S., Tarasov, K., Perman, J., Boren, J., & Ekroos, K. (2009). High-throughput shotgun lipidomics by quadrupole time-of-flight massspectrometry. Journal of Chromatography. B, Analytical Technologies in theBiomedical and Life Science, 877(26), 2664–2672.

Summers, S. A. (2006). Ceramides in insulin resistance and lipotoxicity. Progress inLipid Research, 45(1), 42–72.

Vik, R., Bjorndal, B., Bohov, P., Brattelid, T., Svardal, A., Nygard, O. K., et al. (2014).Hypolipidemic effect of dietary water-soluble protein extract from chicken:Impact on genes regulating hepatic lipid and bile acid metabolism. EuropeanJournal of Nutrition.

110 R. Vik et al. / Food Chemistry 183 (2015) 101–110

Page 99: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

II

Page 100: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were
Page 101: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

A Salmon Protein Hydrolysate Exerts Lipid-IndependentAnti-Atherosclerotic Activity in ApoE-Deficient MiceCinzia Parolini1*., Rita Vik2*., Marco Busnelli1, Bodil Bjørndal2, Sverre Holm3, Trond Brattelid4,

Stefano Manzini1, Giulia S. Ganzetti1, Federica Dellera1, Bente Halvorsen3, Pal Aukrust3,

Cesare R. Sirtori1, Jan E. Nordrehaug5, Jon Skorve2, Rolf K. Berge2,5, Giulia Chiesa1

1Department of Pharmacological and Biomolecular Sciences, Universita degli Studi di Milano, Milan, Italy, 2Department of Clinical Science, University of Bergen, Bergen,

Norway, 3 Research Institute of Internal Medicine, Rikshospitalet University Hospital, Oslo, Norway, 4National Institute of Nutrition and Seafood Research, NIFES, Bergen,

Norway, 5Department of Heart Disease, Haukeland University Hospital, Bergen, Norway

Abstract

Fish consumption is considered health beneficial as it decreases cardiovascular disease (CVD)-risk through effects on plasmalipids and inflammation. We investigated a salmon protein hydrolysate (SPH) that is hypothesized to influence lipidmetabolism and to have anti-atherosclerotic and anti-inflammatory properties. 24 female apolipoprotein (apo) E2/2 micewere divided into two groups and fed a high-fat diet with or without 5% (w/w) SPH for 12 weeks. The atherosclerotic plaquearea in aortic sinus and arch, plasma lipid profile, fatty acid composition, hepatic enzyme activities and gene expressionwere determined. A significantly reduced atherosclerotic plaque area in the aortic arch and aortic sinus was found in the 12apoE2/2 mice fed 5% SPH for 12 weeks compared to the 12 casein-fed control mice. Immunohistochemical characterizationof atherosclerotic lesions in aortic sinus displayed no differences in plaque composition between mice fed SPH compared tocontrols. However, reduced mRNA level of Icam1 in the aortic arch was found. The plasma content of arachidonic acid(C20:4n-6) and oleic acid (C18:1n-9) were increased and decreased, respectively. SPH-feeding decreased the plasmaconcentration of IL-1b, IL-6, TNF-a and GM-CSF, whereas plasma cholesterol and triacylglycerols (TAG) were unchanged,accompanied by unchanged mitochondrial fatty acid oxidation and acyl-CoA:cholesterol acyltransferase (ACAT)-activity.These data show that a 5% (w/w) SPH diet reduces atherosclerosis in apoE2/2 mice and attenuate risk factors related toatherosclerotic disorders by acting both at vascular and systemic levels, and not directly related to changes in plasma lipidsor fatty acids.

Citation: Parolini C, Vik R, Busnelli M, Bjørndal B, Holm S, et al. (2014) A Salmon Protein Hydrolysate Exerts Lipid-Independent Anti-Atherosclerotic Activity inApoE-Deficient Mice. PLoS ONE 9(5): e97598. doi:10.1371/journal.pone.0097598

Editor: Andrea Cignarella, University of Padova, Italy

Received November 20, 2013; Accepted April 22, 2014; Published May 19, 2014

Copyright: � 2014 Parolini et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permitsunrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Funding: This work was supported by grants from NordForsk, grant no. 070010, MitoHealth; the Research Council of Norway, grant no. 190287/110; and theEuropean Community’s Seventh Framework Programme (FP7/2007-2013) AtheroRemo, grant no. 201668. The funders had no role in study design, data collectionand analysis, decision to publish, or preparation of the manuscript.

Competing Interests: The authors have declared that no competing interests exist.

* E-mail: [email protected] (CP); [email protected] (RV)

. These authors contributed equally to this work.

Introduction

Cardiovascular disease (CVD) is responsible for approximately

16–17 million deaths annually, making it the leading cause of

mortality in Western countries [1,2]. The disease encompasses

conditions such as coronary artery disease, carotid and cerebral

atherosclerotic disease and peripheral artery atherosclerosis

resulting in chronic and acute ischemia in affected organs. The

underlying pathological process is lipid accumulation leading to

atherosclerosis, a slowly progressing chronic disorder of large and

medium-sized arteries that can lead to intravascular thrombosis

with subsequent development of complications like myocardial

infarction (MI), stroke and acute ischemia of the limb [3]. In the

last years, inflammation has emerged as an additional key factor in

the development of atherosclerosis and seems to be involved in all

stages, from the small inflammatory infiltrate in the early lesions,

to the inflammatory phenotype characterizing an unstable and

rupture-prone atherosclerotic lesion [4]. In fact, today atheroscle-

rosis is regarded as a disorder characterized by a status of non-

resolved inflammation, with bidirectional interaction between

lipids and inflammation as a major phenotype. Inflammation in

atherosclerosis leads to activation of endothelial cells, enhanced

expression of adhesion molecules, inflammatory cytokines and

macrophage accumulation.

Liver is the main organ regulating lipid metabolism, affecting

blood lipids, especially plasma triacylglycerols (TAG) [5]. Recent-

ly, investigators have suggested that the liver plays a key role in the

inflammatory state of an individual [6,7], and that dietary

cholesterol absorbed by the liver contributes to inflammation

[8]. Research into atherosclerosis has led to many compelling

discoveries about the mechanisms of the disease and together with

lipid abnormalities and chronic inflammation, oxidative stress has

a crucial involvement in the initiation and progression of

atherosclerosis [9].

Improvement of life style and dietary habits can reduce some

risk factors such as high levels of low density lipoprotein (LDL)-

cholesterol, TAG and inflammatory molecules [10]. Fish con-

sumption is consider health beneficial as it lowers plasma lipids

PLOS ONE | www.plosone.org 1 May 2014 | Volume 9 | Issue 5 | e97598

Page 102: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

and attenuates inflammation [11]. This is linked to the long-

chained n-3 polyunsaturated fatty acids (PUFA) content, in

particular eicosapentaenoic acid (EPA) and docosahexaenoic acid

(DHA). However, fish protein is a rich source of bioactive peptides

with valuable nutraceutical and pharmaceutical potentials beyond

that of n-3 PUFAs [11]. Fish protein hydrolysates are generated by

enzymatic conversion of fish proteins into smaller peptides, which

normally contain 2–20 amino acids. In recent years, fish protein

hydrolysates have attracted much attention from food scientists

due to a highly balanced amino acid composition, as well as the

presence of bioactive peptides [12]. The organic acid taurine is

mainly found in marine proteins, and is suggested to induce

cholesterol-lowering effect by increasing excretion through bile,

thus potentially exerting an anti-atherosclerotic effect [13]. Recent

studies show TAG-lowering effects [14,15], antioxidant capacity

[12], antihypertensive [11] and cholesterol-lowering effects

[16,17], and potential to reduce markers of reactive oxygen

species [18] from fish protein. Therefore, fish protein hydrolysates

have been implicated in several processes with potential anti-

atherogenic effects. In this study, we examined the anti-athero-

sclerotic potential of a salmon protein hydrolysate (SPH) on

atherosclerotic development in apolipoprotein E-knockout

(apoE2/2) mice.

Materials and Methods

Experimental DesignThe study was conducted according to national (D.L. 116, G.U.

Suppl. 40, February 18, 1992, Circolare No. 8, G.U July 1994)

and international laws and policies (EEC Council Directive 2010/

63, September 22, 2010: Guide for the Care and Use of

Laboratory Animals, United States National Research Council,

2011). The Italian Ministry of Health approved the protocol (nu04/2012).

24 female apoE2/2 mice from the breeding strain C57BL/6, 8

weeks old, were purchased from Charles River Laboratories

(Calco, Italy), and kept under standard laboratory conditions (12

hours light cycle, temperature 2261uC, humidity 5565%), with

free access to standard chow and tap water. After 1 week of

acclimatization under these conditions, mice were randomly

divided into two groups of 12 mice. Although apoE2/2 mice

spontaneously develop atherosclerosis, both groups were fed a

high-fat diet (23.7% w/w) consisting of 21,3% lard (Ten Kate

Vetten BV, Musselkanaal, Netherlands) and 2.4% soy oil (Dyets.

Inc., Betlehem, PA, USA) to accelerate the atherosclerotic

formation. The control diet contained 21% w/w casein as protein

source, whereas 5% casein was replaced with an equal amount of

salmon protein hydrolysate (SPH) (Marine Bioproducts, Storebø,

Norway) in the intervention diet. The SPH was produced by

enzymatic hydrolysis from salmon by-products (spine) using

controlled autolysis with an alkaline protease and a neutral

protease, and the resulting protein hydrolysate was then subjected

to a second enzymatic treatment with an acid protease A. The

final hydrolysate was fractionated using micro- and ultra- filtration

and the size distribution of the peptides was analysed. The final

preparation consisted of peptides ,1200 Da and 25% of the

peptides were below 200 Da. The diets were isocaloric containing

21% protein, 24% fat, 42% carbohydrates and 6% micronutrients,

and administered for 12 weeks. Other diet ingredients were from

Dyets. Inc., and the full composition of the diets, as well as amino

acid composition, is given in Table S1.

Harvesting of TissueDuring the treatment period, blood samples were collected at

day 1 and after 77 days from the retro-orbital plexus into tubes

containing 0.1% (w/v) EDTA after an overnight fast. Blood

samples were chilled on ice for at least 15 minutes and stored at 280uC until analyses.

After 12 weeks of treatment, mice were sacrificed under general

anaesthesia with 2% isoflurane (Forane, from Abbot Laboratories

Ltd, Illinois, USA) and blood was removed by perfusion with

phosphate-buffered saline (PBS). Aorta was rapidly dissected from

the aortic root to the iliac bifurcation, periadventitial fat and

connective tissue was removed as much as possible. Aorta was

longitudinally opened pinned flat on a black wax surface in ice-

cold PBS, photographed unstained [19] for subsequent plaque

quantification (see En face analysis), and then immediately put in a

tissue-freezing medium, snap-frozen in liquid nitrogen and stored

at 280uC. For histological/immunohistochemical analysis, six

hearts from each group were removed, fixed in 10% formalin for

30 min and transferred into PBS containing 20% sucrose (w/v)

overnight at 4uC before being embedded in OCT compound

(Sakura Finetek Euope B.V., Alphen aan den Rijn, The Nether-

lands) and stored at280uC. An equal subset of hearts and all livers

were immediately snap-frozen in liquid nitrogen for subsequent

analyses.

En Face AnalysisAorta images were recorded with a stereomicroscope-dedicated

camera (IC80 HD camera, MZ6 microscope, Leica Microsystems,

Germany) and analysed using ImageJ image processing program

(http://rsb.info.nih.gov/ij/). An operator blinded to dietary

treatment quantified the atherosclerotic plaques.

Aortic Sinus Histology/immunohistochemistrySerial cryosections (7 mm thick) of the aortic sinus were cut.

Approximately 25 slides with 3 cryosections/slide were obtained,

spanning the three cusps of the aortic valves. Every fifth slide was

fixed and stained with hematoxylin and eosin (Bio-Optica, Milano,

Italy) to detect plaque area. Plaque area was calculated as the

mean area of those sections showing the three cusps of the aortic

valves. Adjacent slides were stained to characterize plaque

composition. Specifically, Masson’s Trichrome (04-010802, Bio-

Optica, Milano, Italy) was used to detect extracellular matrix

deposition and Oil red O staining (Sigma-Aldrich, St. Louis, MO,

USA) was used to detect intraplaque neutral lipids.

Macrophages and T-lymphocytes were detected using an anti-

F4/80 antibody (ab6640, Abcam, Cambridge, UK), and an anti-

CD3 antibody (ab16669, Abcam, Cambridge, UK), respectively.

A biotinylated secondary antibody was used for streptavidine-

biotin-complex peroxidase staining (Vectastain Abc Kit, Vector

Laboratories, Peterborough, UK). 3,39-Diaminobenzidine was

used as chromogen (Sigma-Aldrich, St. Louis, MO, USA), and

sections were counterstained with hematoxylin (Gill’s Hematox-

ylin, Bio-Optica, Milano, Italy). To acquire and process digital

images an Aperio ScanScope GL Slide Scanner (Aperio Technol-

ogies, Vista, CA, USA), equipped with a Nikon 206/0.75 Plan

Apochromat objective producing a 0.25 mm/pixel scanning

resolution with a 406magnification and the Aperio ImageScope

software (version 8.2.5.1263) was used. A blinded operator to the

study quantified plaque area, extracellular matrix and lipid

deposition, as well as inflammatory cell infiltrate. The amount of

extracellular matrix, lipids, macrophages and T-lymphocytes was

expressed as percent of the stained area over the total plaque area.

Salmon Protein Hydrolysate Reduces Atherosclerosis

PLOS ONE | www.plosone.org 2 May 2014 | Volume 9 | Issue 5 | e97598

Page 103: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Plasma Lipid and Fatty Acid Composition MeasurementsEnzymatically measurements of plasma lipids were performed

with an automated method for direct measurement of lipids on a

Hitachi 917 system (Roche Diagnostics GmbH, Mannheim,

Germany) using triacylglycerol (GPO-PAP), total- and free

cholesterol kits (CHOD-PAP) from Roche Diagnostics, and

phospholipids FS kit and a non-esterified fatty acids (NEFA) kit

from DiaSys (Diagnostic Systems GmbH, Holzheim, Germany).

Total plasma fatty acid composition was analyzed as previously

described [20].

Gene Expression in Liver, Heart and AortaTotal cellular RNA was purified from 20 mg liver, total

homogenized heart and pooled aorta samples from six mice using

the RNeasy kit and the protocol for purification of total RNA from

animal cells and fibrous tissue (Qiagen GmbH, Hilden, Germany),

as described by Vigerust et al. and Strand et al., respectively

[21,22]. cDNA was obtained as described by Strand et al. [22].

Real-time PCR was performed on an ABI prism 7900 H sequence

detection system (Applied Biosystems, Foster City, CA, USA) using

384-well multiply PCR plates (Sarstedt Inc., Newton, NC, USA)

and probes and primers from Applied Biosystems, Foster City,

CA, USA as described by Strand et al. [22]. The primers used are

listed in Table S2. Six different reference genes were included for

liver: 18s (Kit-FAM-TAMRA (Reference RT-CKFT-18s)) from

Eurogentec (Seraing, Belgium), ribosomal protein, large, P0

(Rplp0, AX-061958-00-0100), hypoxanthine guanine phosphor-

ibosyltransferase 1 (Hprt1, AX-045271-00), ribosomal protein,

large, 32 (Rpl32, AX-055111-00), polymerase (RNA)II(DNA

directed) polypeptide A, (Polr2a, AX-046005-00) and TATA-box

binding protein (Tbp, AX-041188-00) all five from Thermo Fisher

Scientific Inc. (Waltham, MA, USA). For the heart 18s, Rplp0 and

Hprt1 were used, and for aorta 18s, Rplp0, Rpl32 and Hprt1. The

software GeNorm (http://www.gene-quantification.de/hkg.html)

was used to evaluate the reference genes, and data normalized to

Rplp0 and Rpl32 for liver, Hprt1 for heart and Rplp0 and Hprt1 for

aorta, are presented.

Hepatic Enzyme ActivitiesLivers were homogenized and the post-nuclear fraction isolated

as described earlier [23]. The assay for carnitine palmitoyltrans-

ferase (CPT)-2 was performed according to Bremer [24] and

Skorve et al. [25], but with some modifications: the reaction mix

contained 17.5 mM HEPES pH 7.5, 52.5 mM KCl, 5 mM KCN,

100 mM palmitoyl-CoA and 0.01% Triton X-100. The reaction

was initiated with 100 mM [methyl-14C]-L-carnitine (1100 cpm/

gmol), and 35 mg total protein was used. Palmitoyl-CoA oxidation

was measured in the post-nuclear fraction from liver as acid-

soluble products [26]. The activity of fatty acyl-CoA oxidase

(ACOX)-1 and acyl-CoA: cholesterol transferase (ACAT) were

measured in post-nuclear fractions as described by Madsen et al.

[26] and Field et al. [27], respectively.

Measurements of Plasma Inflammatory MarkersLevels of interleukin (IL)-1b, IL-6, IL-10, tumor necrosis factor

(TNF)-a and granulocyte-macrophage colony-stimulating factor

(GM-CSF) were analyzed on plasma samples collected at day 77 of

treatment by Multiplex suspension technology using a customized

Bio-Plex Pro Mouse assay (Bio-Rad Laboratories, Hercules, CA).

Statistical AnalysisThe results are presented as mean with standard deviation (SD)

for 4–12 mice per group. Normal distribution was assessed by the

Kolmogorov-Smirnov test. Unpaired Student’s t-test was used to

evaluate statistical differences between groups; Mann-Whitney test

was applied when data were not normally distributed. A value of

P,0.05 was considered statistically significant. Statistical analyses

were performed using Prism Software (GraphPad Prism version

5.0; GraphPad Prism, San Diego, CA, USA).

Results

The SPH-diet Decreased Atherosclerotic PlaqueDevelopmentAfter 12 weeks on a high-fat diet, 5% SPH-fed mice displayed a

weight gain similar to the control group. At sacrifice, the average

weight gain was 5.9861.78 g (mean 6 SD) in controls and

5.0460.88 g in SPH mice (P.0.05). A significantly lower plaque

development was observed in the aortic arch in SPH-fed mice

compared to control mice (0.5560.33 vs. 1.6360.996106 mm2;

Fig. 1, corresponding to 0.9160.55 vs. 2.7261.72% of the aortic

surface covered by plaque). There were no differences in thoracic

(1.0860.47 vs. 0.8560.416106 mm2; Fig. 1, corresponding to

1.7160.84 vs. 1.4160.68% of the aortic surface covered by

plaque) or abdominal aorta sections (0.8160.53 vs.

0.7860.536106 mm2; Fig. 1, corresponding to 1.3660.89 vs.

1.2960.88% of the aortic surface covered by plaque).

A significant reduction in lesion area was observed at the aortic

sinus of mice fed SPH compared to controls (1.2760.416105 mm2

vs. 2.0260.316105 mm2; Fig. 2A–C). Plaque stability is an

important factor concerning the severity of atherosclerosis.

However, histological/immunohistochemical characterization of

atherosclerotic lesions displayed no significant difference in plaque

composition between mice fed SPH and controls, showing a

comparable percentage of area occupied by extracellular matrix

(34.5660.56% vs. 30.31618.25%; Fig. 2D–F), lipids

(74.0667.48% vs. 79.6866.45%; Fig. 2G–I), macrophages

(64.4764.47% vs. 60.5763.71%; Fig. 2J–L), and lymphocytes

(27.36611.73% vs. 22.6267.24%; Fig. 2M–O).

Inflammation and oxidative stress are strong contributing

factors in atherosclerosis, thus gene expression of inflammatory

markers and redox regulators in aorta and heart were measured.

Accompanied by decreased plaque area in sinus and aortic arch,

mRNA level of intracellular adhesion molecule (Icam1) was

decreased with 59.54%, in addition to a small decrease in

expression of vascular cell adhesion molecule (Vcam1) and

monocyte chemoattractant protein 1 (Mcp1) in pooled aortic arch

from six mice, whereas mRNA level of inducible nitric oxidase 2

(Nos2) was not modified by the dietary treatment with SPH

(Fig. 3A). In contrast, no changes were found in gene expression

in the heart of Icam1, Vcam1, Mcp1, Nos2 or Tnfa, nor of the

antioxidant markers superoxide dismutase 1, soluble (Sod1),superoxide dismutase 2, mitochondrial (Sod2) or catalase (Cat)(data not shown).

Decreased Plasma Levels of Inflammatory MarkersTo further elucidate the potential anti-inflammatory effects of

SPH in this experimental model of atherosclerosis, we examined

plasma levels of inflammatory mediators. As shown in Fig. 3B–F,levels of IL-1b, IL-6, IL-10, TNF-a and GM-CSF were

significantly lower in SPH-treated mice compared to controls.

SPH-intervention Affected Hepatic mRNA ExpressionInvolved in LipogenesisHyperlipidemia is closely linked to atherosclerotic development.

Liver is the main tissue regulating lipid metabolism, and

mitochondrial b-oxidation is important in regulating plasma

Salmon Protein Hydrolysate Reduces Atherosclerosis

PLOS ONE | www.plosone.org 3 May 2014 | Volume 9 | Issue 5 | e97598

Page 104: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

TAG. Hepatic gene expression showed a significant downregula-

tion in mRNA level of Acaca in SPH-fed mice (Fig. 4A). Moreover,

the mRNA level of Scd1 was significantly downregulated as well

(Fig. 4B).

Noteworthy, SPH administration had no effect on palmitoyl-

CoA oxidation in the presence and absence of malonyl-CoA (Fig.A in Table S1), nor on mitochondrial and peroxisomal fatty acid

oxidation as the enzyme activities of CPT2 and ACOX1,

respectively, were unchanged (Fig. B and C in Figure S1).ACAT activity, involved in cholesteryl ester synthesis, was also

unaltered (Fig. D in Figure S1).

Effects of SPH on Lipid Concentration and Fatty AcidComposition in PlasmaIn order to evaluate the effect of SPH treatment on plasma lipid

concentration, blood was collected for enzymatic measurement of

lipid profile after 77 days of dietary treatment. As shown in

Table 1, plasma total- and free-cholesterol, as well as TAG,

cholesteryl esters and phospholipids concentrations displayed

comparable levels between SPH-group and control group at the

end of treatment period, whereas NEFAs increased in SPH-fed

mice vs. controls (Table 1). Moreover, no difference was observed

between the two groups in the relative amount of saturated fatty

acids (SFA) (Table 2). The relative amount of monounsaturated

fatty acids (MUFA) in SPH-fed mice was slightly lower than

controls at day 77, mainly due to a small decrease in 18:1n-9 (oleic

acid) and 18:1n-7 (vaccenic acid) (Table 2). Total n-6 PUFAs

displayed a higher amount after 77 days of treatment in the SPH-

group, probably due to the increase of C18:2n-6 (linoleic acid) and

C20:4n-6 (arachidonic acid) compared to controls. In contrast, no

differences were detected in the weight % of n-3 PUFAs between

the two groups. As a consequence, a small reduction in n-3/n-6

ratio was observed after 77 days. Overall, the effect of the SPH-

diet on plasma lipids and fatty acids was modest.

Discussion

Fish intake is inversely correlated to CVD-risk factors in both

observational and clinical interventional trials [28]. Particular

attention has been drawn to the cardio-protective effects of fatty

fish species with high levels of omega-3 PUFAs through their lipid-

lowering, anti-inflammatory, antiplatelet and antiarrhythmic

mechanisms [29,30]. Marine organisms are also a rich source of

bioactive proteins and peptides that may induce health benefits

through antihypertensive and antioxidative [28], immunomodu-

lating [31] and lipid-lowering effects [14,17]. Thus, marine

proteins and peptides have been shown to influence the two

major risks for atherosclerotic development, namely hyperlipid-

emia and inflammation. Therefore, it was of interest to investigate

a potential anti-atherosclerotic effect of SPH-diet in apoE2/2 mice

fed a high-fat diet. Although these mice spontaneously develop

atherosclerosis on a standard rodent diet, a high-fat diet regimen,

Figure 1. Atherosclerotic plaque level in apoE2/2 mice fed a high-fat diet (control) or a diet with 5% SPH. After 12 weeks of dietarytreatment, whole aorta was collected and en-face analysis was performed to quantify aortic surface covered by atherosclerotic plaques. Bars representmeans 6 SD of 12 mice for each diet. Unpaired t-test was used to detect statistical significance (*P,0.05).doi:10.1371/journal.pone.0097598.g001

Salmon Protein Hydrolysate Reduces Atherosclerosis

PLOS ONE | www.plosone.org 4 May 2014 | Volume 9 | Issue 5 | e97598

Page 105: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

combined with female mice, was preferred to accelerate the

progression. We showed that apoE2/2 mice fed a high-fat diet

containing 5% (w/w) SPH for 12 weeks developed less athero-

sclerotic plaques compared to controls. In particular, we observed

a significant reduction of plaque area in the aortic arch as well as

in the aortic sinus. The pathophysiological complication of

atherosclerosis is plaque rupture causing heart attack and stroke

in humans. Vulnerability of plaque rupture is an important

element in the fatal outcomes of atherosclerosis, and content and

stability of the plaque is therefore of interest. However, there was

no change in aortic sinus plaque composition of connective tissue,

macrophages or lymphocytes, indicating that SPH had no effect

on plaque stability. Unfortunately, apoE2/2 mice are not

susceptible to the progress of plaque rupture unless treated with

a high-fat diet for over a year, thus studying plaque stability in this

model is limited.

During plaque development, accumulation of adhesion mole-

cules contributes to foam cell formation. In addition to decreased

plaque area in aortic arch, a decrease in expression of the adhesion

molecule Icam1, as well as a small reduction in Vcam1 and the

chemokine Mcp1, was detected in pooled aortic arch of SPH-

treated mice, suggesting a local anti-atherosclerotic effect of the

SPH-diet. The plaque area decreased, but no reduction in number

of macrophages was observed with immunostaining in the aortic

sinus. This could be due to a simultaneous decrease in number of

macrophages and plaque area, which would not be reflected in a

percentage measurement. The mRNA level of inflammatory

markers in heart was unaltered, and could explain the unchanged

levels of macrophages. However, mRNA levels were measured in

total heart that may weaken a potential reduction of these

inflammatory markers. The decrease in sinus plaque area, without

a change of macrophages could also be explained by shrinkage of

the lipid-rich core due to fewer lipids, thus the macrophages

decrease in size.

Liver is the main organ regulating lipoprotein metabolism,

including plasma TAG and cholesterol levels, and a high dietary

cholesterol intake has been reported to elevate liver inflammation

[8]. Noteworthy, the plasma concentrations of cholesterol and

TAG were not affected by SPH-treatment. This was accompanied

by unchanged fatty acid oxidation and ACAT activity. These

results are in contrast with previous reports showing cholesterol-

lowering effects of fish protein hydrolysates in both rats and mice

[14,16]. Although gene expressions of Acaca and the D9-desaturaseScd1 were decreased, it did not affect plasma TAG in apoE2/

2mice. This lack of effect could be explained, at least partially, by

the lower amount of fish protein used in the present study (5%)

compared to previous studies, where 10–25% fish protein

hydrolysate were applied [14,16,17]. In C57BL/6 mice fed 5%

Figure 2. Histological and immunohistochemical characterization of plaques in the aortic sinus in apoE2/2 mice fed a high-fat diet(control) or a diet with 5% SPH for 12 weeks. Representative photomicrographs and quantification of maximum plaque area (panels A–C).Representative photomicrographs and quantification of extracellular matrix deposition (panels D–F), Lipid deposition (panels G–I), Macrophages(panels J–L) and T lymphocytes (panels M–O). The amount of extracellular matrix, lipids, macrophages and T-lymphocytes is expressed as percentageof the stained area over the total plaque area. Bar in panel A = 100 mm. Positive area (%) refers to the percentage of the plaque area occupied byconnective tissue, lipids, macrophages and T lymphocytes, respectively. Data are shown as means 6 SD for 6 mice for each diet and unpaired t-testwas used to detect significance (*P,0.05).doi:10.1371/journal.pone.0097598.g002

Salmon Protein Hydrolysate Reduces Atherosclerosis

PLOS ONE | www.plosone.org 5 May 2014 | Volume 9 | Issue 5 | e97598

Page 106: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

SPH for 6 weeks, a 32% decrease in plasma TAG has been found,

but no change in plasma cholesterol (data to be published). Thus,

in the present study, the disturbed plasma lipid transport in the

apoE2/2 mouse model might have interfered with the potential

TAG-lowering mechanism of SPH, while cholesterol-lowering

effect might not be expected at this dose. A lower cholesterol level

has been observed in animal studies when taurine was added in the

diets [32,33]. However, in our study, the cholesterol level was not

affected after intervention despite the presence of taurine in the

SPH-diet.

The plasma level of NEFAs was unchanged by SPH adminis-

tration and only minor alterations were observed in plasma fatty

acid composition. During the 12 weeks of feeding the plasma level

of MUFAs was slightly lower in the SPH-fed group, but this was

probably not of biological significance. Total n-6 PUFAs in plasma

was higher in SPH-fed mice at the end-point measurement.

Figure 3. Levels of mRNA expression in aorta and inflammatory mediators in plasma in apoE2/2 mice fed a high-fat diet (control) ora diet with 5% SPH for 12 weeks. (A) The gene expressions of the inflammatory markers Icam1, Vcam1, Nos2 and Mcp1 were measured in pooledaortic arch from six mice. Inflammatory markers in blood samples collected at day 77 of treatment were analysed (B) IL-1b, (C) IL-6, (D) IL-10, (E) TNF-a,(F) GM-CSF and bars represent means6 SD of 4 pooled samples of 3 mice for each diet. Unpaired t-test was used to assess statistical significance andresults significantly different from control are indicated (*P,0.05, **P,0.01).doi:10.1371/journal.pone.0097598.g003

Figure 4. Hepatic gene expression in apoE2/2 mice fed a high-fat diet (control) or a diet with 5% SPH for 12 weeks. Hepatic mRNAlevels of (A) Acaca and (B) Scd1. Data for gene expressions are shown as mean values relative to control6 SD for 4 mice for each diet. Mann-Whitneytest was used to assess statistical significance (*P,0.05).doi:10.1371/journal.pone.0097598.g004

Salmon Protein Hydrolysate Reduces Atherosclerosis

PLOS ONE | www.plosone.org 6 May 2014 | Volume 9 | Issue 5 | e97598

Page 107: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Arachidonic acid and oleic acid was increased and decreased in

the SPH group and controls, respectively, after the feeding period.

The increase in arachidonic acid and linoleic acid with a

simultaneously decrease in oleic acid might be due to increased

synthesis of arachidonic acid and linoleic acid from their precursor

oleic acid. Although arachidonic acid is considered pro-inflam-

matory [34], we detected reduction in plaque area in aortic arch

and sinus, suggesting that SPH reduced atherosclerotic activity

independent of the plasma arachidonic acid level. n-3 PUFAs, the

n3/n6 ratio and anti-inflammatory index were not affected by

SPH feeding, which is in contrast to previous findings [35].

However, as stated previously, in the current study we used a

smaller amount of fish protein (5% vs. 15%) and the mouse model

could also influence the effect on fatty acid composition. Knockout

of the apoE gene causes an abnormal plasma lipid composition

and metabolism, which apparently this SPH-diet cannot counter-

act.

Cytokines play a key role in the progression of atherosclerosis

and it was of interest to note that the reduction in plaque area in

the aortic arch was accompanied by a lowering of inflammatory

markers in plasma, as reported in another study using salmon

protein on inflammatory bowel disease in rats [18]. Peroxisome

proliferator-activated receptors (PPAR), which are ligand-depen-

dent transcriptional factors regulating both fatty acid [36] and

amino acid metabolism [37], are shown to exert anti-inflammatory

potential by inhibiting expression of cytokines and other pro-

inflammatory factors [38]. The mechanism is unclear, but Zhu

et al. has recently shown that marine peptides may act as PPAR-

agonists and exert an anti-inflammatory effect [39]. Altogether,

these results suggest that SPH administration might prevent

atherosclerotic development by inhibiting activation of systemic

inflammation.

A small dose of SPH 3.5% in rats has been shown to potentially

exert antioxidant activities by reducing markers for oxidative stress

in colon [18]. In the current study, gene expressions of the

Table 1. Plasma lipids in apoE2/2 mice fed a high-fat casein diet (control) or a high-fat diet with 5% SPH after 77 days of dietarytreatment.

1Lipid class Day 77

Control SPH

Cholesterol 1260.9 1161.0

TAGs 1.460.1 1.360.1

Phospholipids 3.060.1 3.060.1

NEFAs 0.860.2 1.160.1*

Cholesteryl esters 8.160.8 7.960.8

Free Cholesterol 3.760.1 3.460.2

1mmol/L.Data are shown as mean 6 SD (n= 4).Abbreviations: NEFA, non-esterified fatty acid; SPH, salmon protein hydrolysate; TAG, triacylglycerol.*P,0.05 vs. control.doi:10.1371/journal.pone.0097598.t001

Table 2. Plasma fatty acid composition in apoE2/2 mice fed a high-fat casein diet (control) or a high-fat diet with 5% SPH after 77days of dietary treatment.

1Fatty acids Control SPH

gSFAs 3260.5 3460.5

gMUFAs 3160.4 3060.4*

C18:1n-9 (oleic acid) 2560.4 2460.5

C18:1n-7 (vaccenic acid) 1.360.0 1.260.0*

n-6 PUFAs 2860.4 3060.4**

C18:2n-6 (linoleic acid) 1560.1 1660.2***

C20:4n-6 (arachidonic acid) 1260.4 1360.2*

n-3 PUFAs 6.460.3 6.360.3

C20:5n-3 (eicosapentaenoic acid) 0.5360.0 0.460.0

C22:6n-3 (docosahexaenoic acid) 5.060.3 5.060.2

n-3/n-6 0.260.0 0.260.0*

1Fatty acids (% w/w).Data are shown as mean 6 SD (n= 4).Abbreviations: MUFAs, monounsaturated fatty acids; PUFAs, polyunsaturated fatty acids; SFAs, saturated fatty acids; SPH, salmon protein hydrolysate.*P,0.05 vs. control.**P,0.01 vs. control.***P,0.001 vs. control.doi:10.1371/journal.pone.0097598.t002

Salmon Protein Hydrolysate Reduces Atherosclerosis

PLOS ONE | www.plosone.org 7 May 2014 | Volume 9 | Issue 5 | e97598

Page 108: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

antioxidants Sod1, Catalase and Nos2 in the heart were unchanged

by SPH administration, suggesting that SPH did not affect the

antioxidant defence system in the heart of apoE2/2 mice.

Although the present study has some limitations, such as absent

protein data on inflammatory mediators within the aortic lesions,

it gives indication that a salmon protein source may have a

protective role in atherosclerotic development through mecha-

nisms linked to inhibition of inflammation, and not directly related

to plasma lipid changes. Although the apoE2/2 mice model has

been used extensively in experiments studying atherosclerosis as it

gives the opportunity to study genetic influence on atherosclerosis

without using a high-fat diet rich in cholesterol, it is also a

challenging model to use. These mice develop severe atheroscle-

rosis due to accumulation of VLDL in plasma carrying most of the

cholesterol. VLDL, containing apoB-48, is considered more

atherogenic than the apoB-100-containing LDL. High plasma

levels of LDL are also most present in humans with atherosclerosis,

therefore in future studies it would be of interest to test this SPH in

LDLr2/2 mice.

Supporting Information

Figure S1 Hepatic enzyme activities of enzymes involved in

peroxisomal and mitochondrial b-oxidation; (Figure A) Palmi-

toyl-CoA-b-oxidation with and without inhibition with malonyl-

CoA, (Fig. B) CPT2 activity, (Fig. C) ACOX1 activity and (Fig.D) ACAT activity.

(TIFF)

Table S1 Composition and amino acid contents of the diets.

(DOCX)

Table S2 Overview of analysed genes.

(DOCX)

Acknowledgments

We thank Kari Williams, Liv Kristine Øysæd, Randi Sandvik and Svein

Kruger for excellent technical assistance, and Eline Milde Nevdal for

assisting with the animal experiment.

Author Contributions

Conceived and designed the experiments: CP BB TB JS RKB GC.

Performed the experiments: RV MB SH TB SM GSG BH. Analyzed the

data: RV BB FD. Contributed reagents/materials/analysis tools: CRS

JEN. Wrote the paper: CP RV BB PA RKB.

References

1. Lloyd-Jones DM (2010) Cardiovascular risk prediction: basic concepts, current

status, and future directions. Circulation 121: 1768–1777.

2. Woollard KJ (2013) Immunological aspects of atherosclerosis. Clinical Science

125: 221–235.

3. Hansson GK, Robertson A-KL, Soderberg-Naucler C (2006) Inflammation and

atherosclerosis annual review of pathology: Mechanisms of disease. Annual

Reviews 1 297–329.

4. van der Wal AC, Becker AE, van der Loos CM, Das PK (1994) Site of intimal

rupture or erosion of thrombosed coronary atherosclerotic plaques is

characterized by an inflammatory process irrespective of the dominant plaque

morphology. Circulation 89: 36–44.

5. Sabesin SM (1981) Lipid and lipoprotein abnormalities in alcoholic liver disease.

Circulation 64: III 72–84.

6. Kleemann R, Kooistra T (2005) HMG-CoA reductase inhibitors: effects on

chronic subacute inflammation and onset of atherosclerosis induced by dietary

cholesterol. Curr Drug Targets Cardiovasc Haematol Disord 5: 441–453.

7. Rein D, Schijlen E, Kooistra T, Herbers K, Verschuren L, et al. (2006)

Transgenic flavonoid tomato intake reduces C-reactive protein in human C-

reactive protein transgenic mice more than wild-type tomato. J Nutr 136: 2331–

2337.

8. Kleemann R, Verschuren L, van Erk MJ, Nikolsky Y, Cnubben NH, et al.

(2007) Atherosclerosis and liver inflammation induced by increased dietary

cholesterol intake: a combined transcriptomics and metabolomics analysis.

Genome Biol 8 R200.

9. Singh U, Jialal I (2006) Oxidative stress and atherosclerosis. Pathophysiology 13:

129–142.

10. Ridker PM, Rifai N, Cook NR, Bradwin G, Buring JE (2005) Non-HDL

cholesterol, apolipoproteins A-I and B100, standard lipid measures, lipid ratios,

and CRP as risk factors for cardiovascular disease in women. JAMA 294: 326–

333.

11. Harnedy PA, FitzGerald RJ (2012) Bioactive peptides from marine processing

waste and shellfish: A review. Journal of Functional Foods 4: 6–24.

12. Chalamaiah M, Dinesh Kumar B, Hemalatha R, Jyothirmayi T (2012) Fish

protein hydrolysates: proximate composition, amino acid composition, antiox-

idant activities and applications: a review. Food Chem 135: 3020–3038.

13. Elvevoll EO, Eilertsen KE, Brox J, Dragnes BT, Falkenberg P, et al. (2008)

Seafood diets: hypolipidemic and antiatherogenic effects of taurine and n-3 fatty

acids. Atherosclerosis 200: 396–402.

14. Liaset B, Madsen L, Hao Q, Criaeles G, Mellgren G (2009) Fish protein

hydrolysate elevates plasma bile acids and reduces visceral adipose tissue mass in

rats. Biochim Biophys Acta 1791: 254–262.

15. Jacques H, Gascon A, Bergeron N, Lavigne C, Hurley C, et al. (1995) Role of

dietary fish protein in the regulation of plasma lipids. Can J Cardiol 11: Suppl G

63G–71G.

16. Zhang X, Beynen AC (1993) Influence of dietary fish proteins on plasma and

liver cholesterol concentrations in rats. Br J Nutr 69: 767–777.

17. Wergedahl H, Liaset B, Gudbrandsen OA, Lied E, Espe M, et al. (2004) Fish

protein hydrolysate reduces plasma total cholesterol, increases the proportion of

HDL cholesterol, and lowers acyl- CoA:cholesterol acyltransferase activity in

liver of Zucker rats. Journal of Nutrition 134: 1320–1327.

18. Grimstad T, Bjorndal B, Cacabelos D, Aasprong OG, Omdal R, et al. (2012) A

salmon peptide diet alleviates experimental colitis as compared with fish oil.

Journal of Nutritional Science 1: 1–8.

19. Palinski W, Ord VA, Plump AS, Breslow JL, Steinberg D et al. (1994) ApoE-

deficient mice are a model of lipoprotein oxidation in atherogenesis.

Demonstration of oxidation-specific epitopes in lesions and high titers of

autoantibodies to malondialdehyde-lysine in serum. Arterioscler Thromb 14:

605–616.

20. Bjorndal B, Vik R, Brattelid T, Vigerust NF, Burri L, et al. (2012) Krill powder

increases liver lipid catabolism and reduces glucose mobilization in tumor

necrosis factor-alpha transgenic mice fed a high-fat diet. Metabolism 61: 1461–

1472.

21. Vigerust NF, Bjorndal B, Bohov P, Brattelid T, Svardal A et al. (2012) Krill oil

versus fish oil in modulation of inflammation and lipid metabolism in mice

transgenic for TNF-alpha. Eur J Nutr 52: 1315–1325.

22. Strand E, Bjorndal B, Nygard O, Burrin L, Berge C, et al. (2012) Long-term

treatment with the pan-PPAR agonist tetradecylthioacetic acid or fish oil is

associated with increased cardiac content of n-3 fatty acids in rat. Lipids in

Health and Disease 11: 82

23. Berge RK, Flatmark T, Osmundsen H (1984) Enhancement of long-chain acyl-

CoA hydrolase activity in peroxisomes and mitochondria of rat liver by

peroxisomal proliferators. Eur J Biochem 141: 637–644.

24. Bremer J (1981) The effect of fasting on the activity of liver carnitine

palmitoyltransferase and its inhibition by malonyl-CoA. Biochim Biophys Acta

665: 628–631.

25. Skorve J, al-Shurbaji A, Asiedu D, Bjorkhem I, Berglund L, et al. (1993) On the

mechanism of the hypolipidemic effect of sulfur-substituted hexadecanedioic

acid (3-thiadicarboxylic acid) in normolipidemic rats. J Lipid Res 34: 1177–

1185.

26. Madsen L, Rustan AC, Vaagenes H, Berge K, Dyroy E, et al. (1999)

Eicosapentaenoic and docosahexaenoic acid affect mitochondrial and peroxi-

somal fatty acid oxidation in relation to substrate preference. Lipids 34: 951–

963.

27. Field FJ, Albright E, Mathur S (1991) Inhibition of acylcoenzyme A: cholesterol

acyltransferase activity by PD128O42: effect on cholesterol metabolism and

secretion in CaCo-2 cells. Lipids 26: 1–8.

28. Cinq-Mars DC, Hu C, Kitts DD, Li-Chan EC (2008) Investigations into

inhibitor type and mode, simulated gastrointestinal digestion, and cell transport

of the angiotensin i-converting enzyme-inhibitory peptides in pacific hake

(merluccius productus) fillet hydrolysate. J Agric Food Chem 56: 410–419.

29. Raatz SK, Silverstein JT, Jahns L, Picklo MJ (2013) Issues of fish consumption

for cardiovascular disease risk reduction,. Nutrients 5: 1081–1097.

30. Jung UJ, Torrejon C, Tighe AP, Deckelbaum RJ (2008) n-3 Fatty acids and

cardiovascular disease: mechanisms underlying beneficial effects. Am J Clin Nutr

87: 2003S–2009S.

Salmon Protein Hydrolysate Reduces Atherosclerosis

PLOS ONE | www.plosone.org 8 May 2014 | Volume 9 | Issue 5 | e97598

Page 109: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

31. Duarte J, Vinderola G, Ritz B, Perdigon G, Matar C (2006) Immunomodulating

capacity of commercial fish protein hydrolysate for diet supplementation.Immunobiology 211: 341–350.

32. Murakami S, Kondo-Otha Y, Tomisawa (1998) Improvement in cholesterol

metabolism in mice given chronic treatment of taurine and fed a high-fat diet.Life Sciences 64: 83–91.

33. Chen W, Matuda K, Nishimura N, Yokogoshi H (2004) The effect of taurine oncholesterol degradation in mice fed a high-cholesterol diet. Life Sciences. 74:

1889–1898.

34. Dembinska-Kiec A, Gryglewski RJ (1986) Contribution of arachidonic acidmetabolites to atherosclerosis. Wien Klin Wochenschr 98: 198–206.

35. Bjorndal B, Berge C, Ramsvik MS, Svardal A, Bohov P, et al. (2013) A fishprotein hydrolysate alters fatty acid composition in liver and adipose tissue and

increases plasma carnitine levels in a mouse model of chronic inflammation.

Lipids Health Dis 12: 143.36. Muoio DM, Way JM, Tanner CJ, Winegar DA, Kliewer SA, et al. (2002)

Peroxisome proliferator-activated receptor-alpha regulates fatty acid utilization

in primary human skeletal muscle cells. Diabetes 51: 901–909.37. Kersten S, Mandard S, Escher P, Gonzalez FJ, Tafuri S, et al. (2001) The

peroxisome proliferator-activated receptor alpha regulates amino acid metab-olism. FASEB J 15: 1971–1978.

38. Delerive P, Fruchart JC, Staels B (2001) Peroxisome proliferator-activated

receptors in inflammation control. J Endocrinol 169: 453–459.39. Zhu CF, Li GZ, Peng HB, Zhang F, Chen Y et al (2010) Treatment with marine

collagen peptides modulates glucose and lipid metabolism in Chinese patientswith type 2 diabetes mellitus. Appl Physiol Nutr Metab. 36: 797–804.

Salmon Protein Hydrolysate Reduces Atherosclerosis

PLOS ONE | www.plosone.org 9 May 2014 | Volume 9 | Issue 5 | e97598

Page 110: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were
Page 111: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

III

Page 112: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were
Page 113: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

ORIGINAL CONTRIBUTION

Hypolipidemic effect of dietary water-soluble protein extractfrom chicken: impact on genes regulating hepatic lipid and bileacid metabolism

Rita Vik • Bodil Bjørndal • Pavol Bohov • Trond Brattelid •

Asbjørn Svardal • Ottar K. Nygard • Jan E. Nordrehaug •

Jon Skorve • Rolf K. Berge

Received: 11 November 2013 / Accepted: 3 April 2014 / Published online: 23 April 2014

� European Union 2014

Abstract

Background Amount and type of dietary protein have

been shown to influence blood lipids. The present study

aimed to evaluate the effects of a water-soluble fraction of

chicken protein (CP) on plasma and hepatic lipid metabo-

lism in normolipidemic rats.

Methods Male Wistar rats were fed either a control diet

with 20 % w/w casein as the protein source, or an exper-

imental diet where casein was replaced with CP at 6, 14, or

20 % w/w for 4 weeks.

Results Rats fed CP had markedly reduced levels of

triacylglycerols (TAG) and cholesterol in both plasma and

liver, accompanied by stimulated hepatic mitochondrial

fatty acid oxidation and carnitine palmitoyltransferase 2

activity in the 20 % CP group compared to the control

group. In addition, reduced activities and gene expression

of hepatic enzymes involved in lipogenesis were observed.

The gene expression of sterol regulatory element-binding

transcription factor 1 was reduced in the 20 % CP-fed rats,

whereas gene expression of peroxisome proliferator-acti-

vated receptor alpha was increased. Moreover, 6, 14, and

20 % CP-fed rats had significantly increased free carnitine

and acylcarnitine plasma levels compared to control rats.

The plasma methionine/glycine and lysine/arginine ratios

were reduced in 20 % CP-treated rats. The mRNA level of

ATP-binding cassette 4 was increased in the 20 % CP

group, accompanied by the increased level of plasma bile

acids.

Conclusions The present data suggest that the hypotri-

glyceridemic property of a water-soluble fraction of CP is

primarily due to effects on TAG synthesis and mitochon-

drial fatty acid oxidation. The cholesterol-lowering effect

by CP may be linked to increased bile acid formation.

Keywords Chicken protein � Lipogenesis � b-Oxidation

Abbreviations

ABC ATP-binding cassette

Acaca/ACC Acetyl-coenzyme A carboxylase alpha

ACE Angiotensin-converting enzyme

AST Alanine aminotransferase

ALT Aspartate aminotransferase

CP Chicken protein

CPT Carnitine palmitoyltransferase

CV Coefficient of variation

Fasn/FAS Fatty acid synthase

GPAT Glycerol-3-phosphate acyltransferase

HDL High-density lipoprotein

HMG-CoA

reductase

3-hydroxy-3-methylglutaryl-coenzyme

A reductase

HMG-CoA

synthase

3-hydroxy-3-methylglutaryl-coenzyme

A synthase

LDL Low-density lipoprotein

MUFA Monounsaturated fatty acid

NAFLD Nonalcoholic fatty liver disease

NEFA Nonesterified fatty acid

PC Phospholipid phosphatidylcholine

PL Phospholipid

R. Vik (&) � B. Bjørndal � P. Bohov � A. Svardal �O. K. Nygard � J. E. Nordrehaug � J. Skorve � R. K. BergeDepartment of Clinical Science, University of Bergen,

5020 Bergen, Norway

e-mail: [email protected]

T. Brattelid

National Institute of Nutrition and Seafood Research, NIFES,

5817 Bergen, Norway

O. K. Nygard � R. K. BergeDepartment of Heart Disease, Haukeland University Hospital,

5021 Bergen, Norway

123

Eur J Nutr (2015) 54:193–204

DOI 10.1007/s00394-014-0700-5

Page 114: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

PPARa Peroxisome proliferator-activated

receptor alpha

PUFA Polyunsaturated fatty acid

SFA Saturated fatty acid

Srebf/SREBP Sterol regulatory element-binding

transcription factor

TAG Triacylglycerol

T2DM Type 2 diabetes mellitus

VLDL Very-low-density lipoprotein

Background

Diets and lifestyle influence key metabolic pathways in

lipid metabolism implicated in the pathogenesis of meta-

bolic syndrome, including disturbed mitochondrial fatty

acid oxidation [1]. Hypertriglyceridemia and lower plasma

HDL cholesterol are characteristic features in metabolic

syndrome, and an increasing body of evidence has linked

hypertriglyceridemia to decreased mitochondrial fatty acid

oxidation [1, 2].

Peroxisome proliferator-activated receptor alpha

(PPARa) is a nuclear protein that belongs to the super-

family of nuclear hormone receptors [3]. PPARa can be

activated by long-chain fatty acids, and this receptor is

involved in both mitochondrial and peroxisomal fatty acid

oxidation [4]. Interestingly, PPARa also regulates amino

acid metabolism [4]. Sterol regulatory element-binding

proteins (SREBPs) are membrane-bound transcription

factors regulating transcription of genes involved in cho-

lesterol biosynthesis and uptake, fatty acid and lipid pro-

duction, and glucose metabolism [5, 6]. Soy proteins are

known to affect these transcription factors [7–9], and a

number of recent studies have reported hypolipidemic

effects and lowered blood pressure of processed protein

hydrolysates and peptides in animal studies. Proteins from

fish and other marine species and from vegetables such as

soy, pea, and lupin exert both triacylglycerol (TAG)- and

cholesterol-lowering effects in animal as well as in human

studies [10–13]. Moreover, a dietary single-cell protein

reduced fatty liver in obese Zucker rats [14], and recently, a

chicken collagen hydrolysate was shown to exert a strong

angiotensin-converting enzyme (ACE)-inhibitory activity

and antihypertensive effect in spontaneously hypertensive

rats [15, 16]. Thus, the amount and type of dietary proteins

may influence blood lipid concentrations and blood pres-

sure. Specific amino acids of the protein sources are most

likely involved in the hypolipidemic effects of dietary

proteins. From animal experiments, it has been suggested

that low ratios of dietary methionine/glycine and lysine/

arginine contribute to the cholesterol-lowering effect [17].

The aim of the present study was to evaluate the hypo-

lipidemic properties of a water-soluble fraction of chicken

protein (CP) in rats and to investigatewhether aCP diet could

affect mitochondrial fatty acid oxidation and lipogenesis.

Materials and methods

Animals and diets

Twenty-four male Wistar rats (Mollegaard and Blom-

holtgaard, Denmark) 12-week old were housed in Makro-

lon III cages in groups of three, in an open system. They

were kept under standard laboratory conditions with tem-

perature 22 ± 1 �C, dark/light cycles of 12/12 h, relative

humidity 55 ± 5 %, and 20 air changes per. h. The animal

study was conducted according to the Guidelines for the

Care and Use of Experimental Animals, and the Norwegian

State Board of Biological Experiments with Living Ani-

mals approved the protocol.

After 7 days of acclimatization under standard conditions,

rats were divided at random into four groups of six rats. The

control group was fed a 20 % casein diet with 7 % fat (5 %

lard and 2 % soy oil), while the intervention diets were sup-

plemented with 6, 14, or 20 %CP, respectively. The 20 %CP

dose was selected to constitute a full exchange of casein for

CP, while the low (6 %) and intermediate (14 %) doses were

chosen based on comparable studies with fish protein [18, 19].

All groups had free access to tap water and feed during the

28 days of experiment. The composition of the diets is given

in Table 1. The water-soluble fraction of CP (Norilia, Oslo,

Table 1 Composition of the experimental diets

Treatment

Control 6 % CP 14 % CP 20 % CP

Ingredients (g/kg)

Caseina 219.31 163.82 86.87 21.62

Chickenb 0 58.36 139.32 208.05

Soy oil 19.52 19.45 19.35 19.16

Lard 48.81 48.63 48.38 48.16

Corn starch 387.65 386.22 384.23 382.51

Dyetrose 128.85 128.38 127.72 127.15

Sucrose 97.62 97.26 96.76 96.32

Fiber 48.81 48.63 48.63 48.16

AIN-93G mineral mix 34.37 34.15 33.97 33.82

AIN-93 vitamin mix 9.76 9.73 9.68 9.63

L-Cysteine 2.93 2.92 2.90 2.89

Choline–baritrate 2.45 2.44 2.43 2.42

Tert-butylhydroquinone 0.014 0.014 0.014 0.014

The diets were isoenergetic and isonitrogenous, and contained 20 g

protein per 100 g diet. The final diet contained 20 % (w/w) watera Casein consisted of 91.1 % (w/w) protein and 0.2 % fat (w/w)b Water-soluble fraction of chicken protein consisted of 97 % (w/w)

protein and 3 % fat (w/w)

194 Eur J Nutr (2015) 54:193–204

123

Page 115: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Norway)was prepared as follows:Meat fromchicken legs and

wings were milled, and water was added. The mixture was

treated for 20 min in a pressure cooker at a pressure about 3

bar and at a temperature of approximately 136 �C. The

resulting water-soluble fraction of chicken protein was

evaporated and spray dried. The amino acid composition of

the 20 %CPdiet compared to control is shown inTable 2. Rat

feed intake and weight gain were measured twice a week. On

day 28, rats were anesthetized after an overnight fast by

inhalation of 2 % isofluorane (Forane, from Abbot Labora-

tories Ltd, Illinois, USA) in an anesthesia chamber, and tho-

racotomy, cardiac puncture, and exsanguination were

performed. Blood was collected in tubes with K3EDTA,

chilled on ice for approximately 15 min, and stored at-80 �Cuntil analyses. Livers were harvested, freeze-clamped, and

stored at-80 �C.

Plasma and liver lipids and fatty acid composition

Liver lipids were extracted according to Bligh and Dyer

[20], evaporated under nitrogen, and redissolved in

isopropanol before analysis. Lipids from liver extracts or

plasma were then measured enzymatically on a Hitachi 917

system (Roche Diagnostics GmbH, Mannheim, Germany)

using the triacylglycerol (GPO-PAP) and cholesterol kit

(CHOD-PAP) from Roche Diagnostics, and the phospho-

lipid kit from DiaSys (Diagnostic Systems HmbH, Holz-

heim, Germany). Coefficient of variation (CV) was

determined as 6.3 % for total cholesterol, 2.2 % for TAG,

3.2 % for LDL, 3.8 % for PL, and below 1.5 % for NEFAs,

HDL, and free cholesterol. Total liver fatty acid composi-

tion was analyzed in controls and 20 % CP-fed rats as

described previously [21]. The procedure of fatty acid

determination was regularly verified for specificity, inter-

day, and intra-day reproducibility (CV around 1 % for

major fatty acids and 2–4 % for minor fatty acids), preci-

sion (CV below 2 %), stability, and other validation

parameters.

Plasma carnitine composition

Free carnitine, short-, medium-, and long-chain acylcarni-

tines, and the precursor for carnitine, c-butyrobetaine, wereanalyzed in plasma using LC MS/MS as described previ-

ously [22], with some modification as described by Vige-

rust et al. [23].

Plasma and diet pattern of amino acids and amine

metabolites

The amino acids in the diets were determined after

hydrolysis in 6 M HCl at 110 �C for 22 h and pre-

derivatization with phenylisothiocyanate according to the

method of Cohen and Strydom [24]; thus, the sum of

aspartic acid and aspargine, and the sum of glutamic acid

and glutamine, is written as Asx and Glx, respectively.

EDTA plasma from controls and 20 % CP-fed rats

(200 lL) was deproteinized by adding 10 % sulfosalisylic

(1:1, v/v) with norleucine as internal standard (final

0.5 mM) and stored for 1 h at 4 �C before centrifugation

at 8,000 rpm for 30 min as described by Liaset et al. [25].

The supernatant was filtered, and amino acids were

characterized by a Biochrom 20 plus amino acid analyser

as previously described [25].

Quantitative determination of total bile acids

Measurement of bile acids was performed enzymatically

on frozen plasma samples on a Molecular P Roche Diag-

nostics instrument using kit from Diazyme Laboratories

(Cat. No. 05471605001). Total bile acids were measured as

the rate of formation of Thio-NADH determined by mea-

suring specific change of absorbance at 405 nm.

Table 2 Amino acid composition in the control diet and 20 % CP

diet fed to male Wistar rats for 4 weeks

Control 20 % CP

Amino acid (mg/g)

Hyp 0 11.86

His 5.39 3.20

Tau 0.01 0.64

Ser 11.07 5.68

Arg 5.77 9.98

Gly 3.65 22.67

Asx 15.87 11.58

Glx 49.44 22.11

Thr 9.15 4.71

Ala 6.37 11.79

Pro 23.46 15.04

Lys 17.15 7.69

Tyr 8.24 2.34

Met 5.73 2.60

Val 13.47 5.41

Ile 11.20 4.11

Leu 19.65 8.49

Phe 10.32 5.01

Amino acid ratio

Met/Gly 1.57 0.11

Lys/Arg 2.97 0.77

Ala alanine, Arg arginine, Asx apsartic acid ? aspargine, Glc glu-

tamic acid ? glutamine, Gly glycine, His histidine, Hyp hydroxy-

proline, Ile isoleucine, Leu leucine, Lys lysine, Met methionine, Phe

phenylalanine, Pro proline, Ser serine, Tau taurine, Thr threonine, Tyr

tyrosine, Val valine

Eur J Nutr (2015) 54:193–204 195

123

Page 116: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Hepatic enzyme activities

After killing, livers were removed, chilled on ice,

weighed, and snap-frozen in liquid nitrogen. About

100 mg liver from each rat was homogenized in 1 mL

ice-cold sucrose medium (0.25 M sucrose, 10 mM

HEPES, and 1 mM Na4EDTA, adjusted to a pH of 7.4

with KOH) giving 10 % (w/v). The homogenates were

centrifuged at 600g force for 10 min at 4 �C, and the

post-nuclear fraction was removed and used for further

analysis. The assay for carnitine palmitoyltransferase 1

(CPT1) was performed according to Bremer et al. [26],

but with some modifications: The reaction mix contained

17.5 mM HEPES pH 7.5, 52.5 mM KCl, 5 mM KCN,

100 mM palmitoyl-CoA, and 6.67 mg BSA/mL. The

reaction was initiated with 100 lM [methyl-14C]-L-car-

nitine (1,100 cpm/gmol), and 30 lg total protein was

used. Palmitoyl-CoA oxidation was measured in the post-

nuclear fraction from liver as acid-soluble products [27].

Assay conditions for CPT2 were identical except that

BSA and KCN were omitted, 0.01 % Triton X-100 was

included, and a total of 35 lg protein was used. The

activity of fatty acyl-CoA oxidase 1 (ACOX1) was

measured in post-nuclear fractions using 20 lg protein, as

described by Madsen et al. [27]. Fatty acid synthase

(FAS) and glycerol-3-phosphate acyltransferase (GPAT)

were measured in post-nuclear fractions as described by

Skorve et al. [28], but with some modifications: FAS

activity was measured in post-nuclear fractions using

60 lg protein. GPAT was assayed for 5 min at 35 �C, andthe reaction was initiated by the addition of post-nuclear

fractions (150 lg protein) and terminated by the addition

of 400 ll water-saturated butanol.

Gene expression analysis

Total cellular RNA was purified from frozen liver samples

from the controls and 20 % CP-fed rats only, and cDNA

was produced as previously described [29]. Real-time PCR

was performed with Sarstedt 384 well multiply-PCR Plates

(Sarstedt Inc., Newton, NC, USA) on the following genes,

using probes and primers from Applied Biosystems:

ATP-binding cassette, subfamily B, member 4 (Abcb4,

Rn01529224_m1), acetyl-CoA carboxylase alpha (Acaca,

Rn00573474_m1), acyl-CoA oxidase 1 (Acox1, Rn005

69216), apolipoprotein B (Apob, Rn01499050_g1), bile

acid CoA:amino acid N-acyl transferase (Baat, Rn0056

8867_m1), CD36 antigen (Cd36 (Fat), Rn00580728_m1),

carnitine palmitoyltransferase 1a (Cpt1a Rn00580702_m1),

Cpt2 (Rn00563995_m1), cytochrome P450, family 7, sub-

family A, polypeptide 1 (Cyp7a1, Rn00564065_m1), fatty

acid synthase (Fasn, Rn00569112_m1), 3-hydroxy-3-

methylglutaryl-CoA synthase 1 (Hmgcs1, Rn00568579_

m1), Hmgcs2, (Rn00597339_m1), 3-hydroxy-3-methyl-

glutaryl-CoA reductase (Hmgcr, Rn00565598_m1), low-

density lipoprotein receptor (Ldlr, Rn00598438_m1),

peroxisome proliferator-activated receptor alpha (Ppara,

Rn00566193_m1), stearoyl-CoA desaturase 1 (Scd1,

Rn00594894_g1), and sterol regulatory element-binding

transcription factor 1 (Srebf1, Rn01495769_m1) and

Srebf2 (Rn01502638_m1). Three different reference genes

were included: 18 s [Kit-FAM-TAMRA (Reference RT-

CKFT-18 s)] from Eurogentec, Belgium, glyceraldehyde-

3-phosphate dehydrogenase (Gapdh, Rodent 4308313)

from Applied Biosystems, and ribosomal protein, large, P0

(Rplp0, Rn00821065_g1). GeNorm was used to evaluate

the reference genes [30], and data normalized to Gapdh are

presented.

Statistical analysis

Data sets were analyzed using Prism Software (Graph-Pad

Software, San Diego, CA) to determine statistical signifi-

cance. The results are shown as means of six rats per group

with their standard errors of the mean (SEM). Normal

distribution was determined by the Kolmogorov–Smirnov

test (with Dallal–Wilkinson–Lillie for p value). One-way

ANOVA with Tukey’s post hoc test was used to assess

statistical differences between groups. Unpaired t test was

performed to evaluate statistical differences between two

groups. p values\0.05 were considered significantly dif-

ferent from controls.

Results

The effect of CP on body weight, liver weight, and feed

intake

No change in weight gain was observed in rats fed 6 or

14 % CP compared to control (Fig. 1a, b). However, rats

fed 20 % CP gained less weight than rats fed a 20 %

casein diet (Fig. 1b). Already after 1 week, the 20 % CP-

fed rats reduced their growth rates (Fig. 1a) and continued

at this rate for the following 3 weeks. The average total

weight gain was 105 ± 12 g versus 152 ± 14 g (mean-

s ± SEM) for the 20 % CP group and control rats,

respectively (Fig. 1b). The overall mean feed consump-

tion was higher in the 14 and 20 % CP-fed rats as

compared to the control rats (Fig. 1c), and the overall

feed efficiency (g weight gain/g feed intake) was signif-

icantly lower for the 20 % CP-fed rats (Fig. 1d). Liver

weight was significantly reduced in the 20 % CP-fed rats

compared to casein-fed rats without affecting the liver/

body index (Fig. 1e, f).

196 Eur J Nutr (2015) 54:193–204

123

Page 117: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

CP-reduced plasma lipids and altered hepatic lipid

profile

The plasma concentrations of cholesterol, cholesterol esters,

TAG, and phospholipids were reduced in the CP-treated rats

compared to the casein-fed rats, measured as the difference

between the end and start of the experiment (D-values)(Fig. 2a–d). 14 and 20 % CP-fed rats had a similar choles-

terol- and phospholipid-reducing effect, while 6 % CP did

not affect these parameters. Moreover, plasma TAG

(Fig. 2c) and bile acids (Fig. 2e) levelswere reduced at 20 %

CP. The plasma values ofHDL cholesterol, LDL cholesterol,

nonesterified fatty acids (NEFAs), and glucose were not

significantly affected (data not shown). Hepatic lipids dem-

onstrated significantly lower total liver TAG (p = 0.017),

cholesterol (p = 0.002), and phospholipids (p = 0.0003) in

the 20 %CP group compared to control rats (Fig. 2f–h). The

plasma lipid levels of the Wistar rats were comparable to a

previous study [31].

Dietary supplementation with 20 % CP also changed the

fatty acid composition in the liver compared to the control

diet (Table 3). The n3 polyunsaturated fatty acids (PUFAs)

alpha-linolic acid (C18:3n3) and docosapentaenoic acid

(DPA) (C22:5n3) were significantly increased in liver after

20 % CP feeding, but eicosapentaenoic acid (EPA)

(C20:5n3) was decreased. The total n6 PUFAs tended to

increase, but did not reach significant (p = 0.064)

(Table 3). Altogether, the 20 % CP diet resulted in a

decrease in the n3/n6 PUFAs ratio. It was of interest to note

that the weight % of SFAs was significantly increased in

liver of 20 % CP-treated rats, and long-chain fatty acids

were increased more than short-chain fatty acids as C14:0

and C16:0 were unchanged, whereas C20:0–C24:0 were

increased. The amounts of monounsaturated fatty acids

(MUFAs) displayed a trend toward decrease (p = 0.061) in

liver of rats fed 20 % CP compared to controls (Table 3),

although probably contributing to significant decrease in

the D9 desaturase index (C16:1n7/C16:0) (Table 3).

Fig. 1 a Body weight, b body

weight gain, c feed intake,

d feed efficiency, e liver weight,and f liver index in male Wistar

rats fed a control diet with 20 %

casein or a diet with 6, 14, or

20 % CP for 4 weeks. Values

are means with their standard

errors represented by vertical

bars (n 6). a,bMean values with

unlike letters were significantly

different (p\ 0.05; one-

ANOVA with Tukey’s post hoc

test). (*p\ 0.05, **p\ 0.01;

unpaired t test)

Eur J Nutr (2015) 54:193–204 197

123

Page 118: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

The CP diets increased hepatic fatty acid oxidation

and decreased lipogenesis

Hepatic mitochondrial b-oxidation of long-chain fatty

acids with palmitoyl-CoA as substrate was significantly

increased in rats fed 20 % CP compared to casein-fed rats,

but no increase was observed with 6 % or 14 % CP

(Fig. 3a). In the presence of malonyl-CoA, the results were

unchanged in all groups (Fig. 3a). Gene expression of

Cpt1a was unchanged (Table 4), but the CPT2 activity was

increased by the dietary administration of 20 % CP

(Fig. 3b). CP feeding did not seem to increase the perox-

isomal b-oxidation capacity, as the ACOX1 activity and

gene expression were not changed (Fig. 3c; Table 4,

respectively). HMG-CoA synthase 2, which is involved in

ketone body production, was not affected at the levels of

activity (data not shown) or mRNA expression after the CP

treatment (Table 4), neither was the level of ketone bodies

(data not shown). Interestingly, the mRNA level of the

nuclear transcription factor Ppara, involved in the regula-

tion of lipid catabolic pathways, was significantly increased

in the rats fed 20 % CP (Table 4).

We then looked at enzymes involved in the fatty acid

synthesis pathway. The activity of ACC was significantly

reduced by the 20 % CP diet and tended to be reduced by

14 % CP (p = 0.103, Fig. 3d). A tendency toward reduc-

tion in the expression of the ACC-gene (Acaca) by the

20 % CP diet was observed at the mRNA level (Table 4).

The fatty acid synthase (FAS) activity showed a tendency

to decrease in all the CP-treated groups, but only signifi-

cantly by 20 % CP administration (Fig. 3e), and this

reduction in activity was reflected at the mRNA level

(Table 4). The activity of GPAT, which is involved in

TAG biosynthesis, was significantly decreased in both the

14 and 20 % CP-fed rats (Fig. 3f). Rats fed 20 % CP also

showed a lower expression level of the important lipogenic

gene stearoyl-CoA 1 (Scd1), also called delta 9 desaturase

(Table 4). Accordingly, the lipogenic transcription factor,

Srebf1, was significantly reduced at the mRNA level by

20 % CP (Table 4). Apob, which is the main apolipopro-

tein of VLDL, was not changed after 20 % CP adminis-

tration compared to control (Table 4).

CP showed no effect on cholesterol synthesis

While the mitochondrial Hmgcs2 is involved in the pro-

duction of ketone bodies, the cytosolic HMG-CoA syn-

thase 1 (Hmgcs1) is involved in cholesterol synthesis. Gene

expression of this enzyme was not significantly reduced by

20 % CP treatment (p = 0.277, Table 4). The 20 % CP-fed

rats showed a trend for enhanced Srebf2 mRNA level, but

the gene expression of Hmgcr, the rate-limiting enzyme in

cholesterol synthesis, was not affected compared to control

rats (Table 4). Gene expression of Ldlr was not altered by

the 20 % CP diet compared to the control diet (Table 4).

Elevated plasma bile acids level after intervention

with CP

The plasma concentration of total bile acids was signifi-

cantly increased in the 20 % CP-treated rats compared to

the casein-fed rats (Fig. 2e). Bile acids are normally con-

jugated with taurine and/or glycine, but the gene expression

of Baat, responsible for this conjugation, was unchanged

Fig. 2 a D cholesterol, b D cholesterol esters, c D triacylglycerols,

d D phospholipids, e plasma bile acids, f hepatic cholesterol, g hepatictriacylglycerols, and h hepatic phospholipids in male Wistar rats fed a

control diet with 20 % casein or a diet with 6, 14, or 20 % CP after

4 weeks. Values are means with their standard errors represented by

vertical bars (n 6). a,bMean values with unlike letters were

significantly different (p\ 0.05; one-way ANOVA with Tukey’s

post hoc test). (*p\ 0.05, **p\ 0.01, ***p\ 0,001; unpaired t test)

198 Eur J Nutr (2015) 54:193–204

123

Page 119: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

by the 20 % CP diet (Table 4). Hepatic gene expression of

the rate-limiting enzyme in bile acid synthesis, Cyp7a1,

was increased in the 20 % CP-fed rats compared to the rats

fed casein (Table 4), but the difference did not reach sta-

tistical significance (p = 0.102). It was of interest to note

that the gene expression of the canalicular exporter Abcb4,

an ATP-dependent efflux pump that may be involved in

bile export from liver to bile [32] and bile excretion

through urine [33], was significantly increased (Table 4).

No changes in the plasma activity levels of alanine ami-

notransferase (ALT) and aspartate aminotransferase (AST)

were found after CP treatment (data not shown).

CP influenced plasma amino acids and amine

metabolites

The plasma concentrations of the following free amino

acids were increased in the 20 % CP-treated rats compared

to controls: aspartic acid, hydroxyproline, serine, glycine,

citrulline, ornithine, L-methylhistidine, and arginine

(Table 5). Moreover, threonine, asparagine, valine, methi-

onine, isoleucine, tyrosine, phenylalanine, lysine, and

tryptophan were present in significantly lower concentra-

tions in 20 % CP-treated rats. Interestingly, a lower

methionine/glycine and lysine/arginine ratios were found

in plasma of rats fed a 20 % CP diet (Table 5). Plasma

concentrations of taurine, urea, and ammonium were

unchanged (Table 5). The taurine level in the 20 % CP

diet, however, was higher than in the control diet (Table 2).

Plasma acyl-carnitines in male Wistar rats increased

after feeding with CP

The CP-fed rats showed a significantly increased plasma-

free carnitine level compared to the control rats at all three

doses of CP in the diet (Fig. 4a). The direct carnitine pre-

cursor c-butyrobetaine was significantly increased in CP-

treated rats at doses of 6 and 20 % (Fig. 4b). Plasma acet-

ylcarnitine (two-carbon chain) was significantly increased

with CP diets at 14 and 20 % compared to control (Fig. 4c),

as were octanoylcarnitine (eight-carbon chain) and palmi-

toylcarnitine (16-carbon chain) (Fig. 4e, f). Propionylcar-

nitine (three-carbon chain) was elevated at all three doses

(Fig. 4d). The product of branched-chain amino acid

catabolism or odd-chain fatty acid metabolism, isovaleryl-/

valerylcarnitine (five-carbon chain), was not affected by the

CP diets (data not shown).

Discussion

In the present study, we demonstrate that a water-soluble

fraction of chicken protein has marked hypolipidemic

properties in rats and that this is related to differential

effects on lipid metabolic pathways. Hypolipidemic prop-

erties have previously been shown with a chicken collagen

hydrolysate [34], but to our knowledge, no studies have

investigated the mechanism of action of lipid lowering by

dietary chicken proteins.

The results from this study suggest a TAG-lowering effect

in CP-fed rats mediated through increased catabolism and

decreased synthesis of fatty acids. Indeed, the mitochondrial

Table 3 Fatty acid composition in liver of male Wistar rats given an

20 % casein diet (control) or a 20 % CP diet for 4 weeks (mean

values with standard errors, n 6 per group)

Control 20 % CP

Mean SEM Mean SEM

Fatty acids(w/w)

R SFAs 35.18 0.22 36.39* 0.35

C14:0 0.56 0.03 0.42 0.07

C16:0 19.00 0.33 18.83 0.80

C17:0 0.22 0.01 0.33*** 0.01

C18:0 14.13 0.32 15.41 0.74

C20:0 0.05 0.00 0.06*** 0.00

C22:0 0.15 0.00 0.20** 0.01

C24:0 0.44 0.01 0.57* 0.04

R MUFAs 20.69 0.38 16.85 1.78

C16:1n7 3.15 0.27 1.33*** 0.30

C16:1n9 0.23 0.01 0.13*** 0.01

C18:1n7 4.58 0.21 2.22*** 0.11

C18:1n9 11.90 0.21 12.36 1.42

R n6 PUFAs 35.18 0.57 38.35 1.41

C18:2n6 11.41 0.57 14.46*** 0.27

C18:3n6 0.15 0.01 0.26*** 0.01

C20:3n6 1.21 0.11 1.04 0.04

C20:4n6 21.36 1.10 21.08 1.23

C22:4n6 0.33 0.45 0.54** 0.03

C22:5n6 0.44 0.05 0.65*** 0.07

R n3 PUFAs 8.50 0.24 8.02 0.42

C18:3n3 0.25 0.03 0.37* 0.04

C18:4n3 0.01 0.00 0.03*** 0.00

C20:5n3 0.53 0.05 0.39* 0.03

C22:5n3 0.64 0.02 1.06*** 0.07

C22:6n3 6.93 0.30 6.02 0.39

R n3/R n6 ratio 0.24 0.01 0.21* 0.00aD5 (n3) desat index 4.10 0.27 2.72* 0.18bD9 desat index 0.17 0.80 0.07*** 0.37cD9 desat index 0.84 0.27 0.80 0.78

* Mean values were significantly different from controls (* p\ 0.05,

** p\ 0.01, *** p\ 0.001)a D5 (n3) desaturase index = C20:5n3/C20:4n3b D9 desaturase index = C16:1n7/16:0c D9 desaturase index = C18:1n9/18:0

Eur J Nutr (2015) 54:193–204 199

123

Page 120: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

b-oxidation of fatty acids, mitochondrial CPT2 activity, and

the acetylcarnitine plasma levelwere increased in rats fedCP

compared to rats fed casein. In agreement with this, the 20 %

CP component significantly increased gene expression of

Ppara. The increase inmitochondrial b-oxidation seen in the20 %CP-fed rats compared to controlsmay have contributed

to the lower bodyweight in these rats. Ketone body synthesis

is directly related to the concentration of acetyl-CoA.

However, induced acetyl-CoA formation due to increased

mitochondrial b-oxidation in CP-fed rats may have been

used for Krebs cycle intermediates and not ketone body

production, as HMG-CoA synthase activity and gene

expression of Hmgcs2 were not increased. The peroxisomal

fatty acid oxidation system was unchanged by dietary CP, as

ACOX1 activity and gene expression were not affected.

About 20 % CP-treated rats showed increased hepatic and

plasma levels of long-chain fatty acids (C17:0 and C20:0–

C24:0) compared to control. This increase in long-chain fatty

acids is presumably a consequence of the lack of stimulated

chain-shortening by the peroxisomal fatty acid system, and

thus a preferential increase in the mitochondrial b-oxidationof short-chain fatty acids. The lower liver n3/n6 ratio relates

to the lower D5 desaturation of n3 PUFAs (Table 3).

Although the C18:1n9/18:0 ratio was unchanged, the sig-

nificantly lower C16:1n7/16:0 ratio in the 20 % CP-fed rats

corresponds to the significantly lower gene expression of

Scd1. This indicates a decrease in D9 desaturation [35] and

thus may have caused the trend to decrease in MUFAs

compared to control.

The overall changes in the mitochondrial b-oxidationprocess were further assessed by measuring plasma carni-

tine, carnitine precursor, and acyl-carnitines, as such pro-

files reflect the intramitochondrial accumulation of acyl-

CoA esters [36]. The increase in both plasma carnitine and

the carnitine precursor c-butyrobetaine at all three doses

shows that CP affected carnitine biosynthesis differently

from a casein diet. The increased level of acetylcarnitine in

the CP-fed rats is probably due to increased fatty acid

oxidation, or amino acid degradation, as acyl-CoA is the

end product of both processes. The increased propionyl-

carnitine level reflects increased oxidation of odd-chain

fatty acids or activated degradation of branched amino

acids [37]. Altogether, by increasing carnitine levels

through the CP treatment, a balanced acyl-CoA/CoA ratio

is maintained, potentially lipotoxic mitochondrial metabo-

lites are exported, and mitochondrial capacity could be

improved.

Compared to the control rats, the amount of hepatic

TAG in the 20 % CP-fed rats was reduced. CP treatment

resulted in an inhibition of the enzyme activities of FAS

and GPAT. Moreover, the gene expression of the lipogenic

genes Fasn, Acaca, and Scd1 was lower in rats treated with

20 % CP than in casein-fed rats. The reduction in lipo-

genesis and TAG biosynthesis together with stimulated

fatty acid catabolism can explain the decrease in liver and

plasma TAG content. The parallel decrease in GPAT and

FAS activities, as well as in hepatic and plasma TAG

concentrations, without any effect on gene expression of

Fig. 3 Mitochondrial b-oxidation with a palmitoyl-CoA as a sub-

strate, b enzyme activity of carnitine palmitoyltransferase 2 (CPT2),

c enzyme activity of acyl-CoA oxidase (ACOX1), d enzyme activity

of acetyl-CoA carboxylase (ACC), e enzyme activity of glycerol-3-

phosphate acyltransferase (GPAT), and f enzyme activity of fatty acid

synthase (FAS) in male Wistar rats fed a control diet with 20 %

casein or a diet with 6, 14 or 20 % CP for 4 weeks. Values are means

with their standard errors represented by vertical bars (n 6). a,bMean

values with unlike letters were significantly different (p\ 0.05; one-

way ANOVA with Tukey’s post hoc test)

200 Eur J Nutr (2015) 54:193–204

123

Page 121: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

Cd36 and Apob mRNA, suggests that the hypotriglyceri-

demic effect, combined with reduced liver fat of 20 % CP-

fed rats, is caused by effects on both synthesis and catab-

olism as transportation is unaffected.

The effects of CP on plasma cholesterol levels could be

due to reduced cholesterol synthesis and/or increased

degradation and secretion. The gene expressions of cyto-

solic Hmgcs1 and Hmgcr were not altered in 20 % CP-fed

rats. However, the 20 % CP-fed rats had an increased

plasma level of bile acids, with a tendency to increased

gene expression of Cyp7a1 (p\ 0.102) compared to

casein-fed rats. Therefore, we suggest that the decrease in

plasma and hepatic cholesterol was mediated largely by

increased cholesterol degradation, and not at the synthesis

level. Bile acids and phosphatidylcholine (PC) are the main

components of bile and are essential for uptake of dietary

fat. After synthesis, bile acids conjugate with glycine or

taurine making them more soluble. The bile salt export

pump, ABCB11, and the PC floppase, ABCB4, are both

Table 4 Hepatic gene expression in male Wistar rats fed an 20 %

casein diet (control) or a 20 % CP diet for 4 weeks (mean values with

their standard errors, n 6)

Gene Function Diet groups p value

Control 20 % CP

Mean SEM Mean SEM

Transcriptional factors

Srebf1 Fatty acid

synthesis

1.00 0.12 0.42 0.02 0.000

Srebf2 Cholesterol

homeostasis

1.00 0.24 1.22 0.14 0.138

Ppara Energy

metabolism

1.00 0.05 1.22 0.08 0.019

Lipid import, b-oxidation, and ketogenesis

Cd36 Fatty acid

import

1.00 0.10 1.56 0.40 0.217

Cpt1a b-Oxidation 1.00 0.96 1.55 0.40 0.851

Acox1 b-Oxidation 1.00 0.10 1.04 0.15 0.264

Hmgcs2 Ketogenesis 1.00 0.02 0.91 0.07 0.467

Lipogenesis and cholesterol synthesis and import

Fasn Fatty acid

synthesis

1.00 0.11 0.42 0.06 0.001

Acaca Fatty acid

synthesis

1.00 0.07 0.77 0.09 0.071

Scd1 D9 desaturase 1.00 0.11 0.28 0.05 0.000

Hmgcs1 Cholesterol

synthesis

1.00 0.08 1.68 0.59 0.277

Hmgcr Cholesterol

synthesis

1.00 0.12 1.18 0.27 0.562

Ldlr Cholesterol

import

1.00 0.05 0.96 0.09 0.684

Apob Cholesterol

import

1.00 0.09 0.90 0.04 0.349

Cyp7a1 Bile acid

synthesis

1.00 0.20 4.37 1.80 0.102

Baat Bile

conjugation

1.00 0.05 1.03 0.06 0.704

Abcb4 Bile transporter 1.00 0.06 1.66 0.15 0.002

Mean values were significantly different from controls (* p\0.05, **

p\ 0.01, *** p\ 0.001)

Table 5 Plasma pattern of selected amino acid and amino metabo-

lites in male Wistar rats given a 20 % casein diet (control) or an 20 %

CP diet for 4 weeks (mean values with their standard errors, n 6)

Amino acids and amine

metabolites (lmol/ll)Control 20 % CP

Mean SEM Mean SEM

Tau 22.55 1.20 23.11 1.30

Urea 745.62 34.30 695.69 44.73

Amm 12.59 1.49 13.01 1.01

Asp 1.50 0.20 2.29** 0.08

Hypro 2.86 0.18 16.07*** 0.79

Thr 47.11 3.35 27.46*** 1.99

Ser 26.55 1.47 70.91*** 3.38

Asn 11.11 0.69 4.81*** 0.17

Glu 13.90 2.91 17.53 0.86

Gln 72.40 1.48 71.18 3.45

Pro 45.23 4.13 27.94** 1.05

Gly 17.23 0.79 77.33*** 2.40

Ala 7.79 4.35 47.74*** 1.95

Citr 10.18 0.47 12.02* 0.61

Val 31.39 1.24 11.83*** 0.36

Cysteine 2.69 0.24 2.26 0.22

Met 7.82 0.52 5.9*** 0.11

Ile 13.58 0.66 5.23*** 0.18

Leu 21.25 1.21 8.52*** 0.27

Tyr 16.49 1.98 9.33** 0.23

Phe 7.36 0.27 5.50*** 0.26

Orn 5.03 0.21 7.06** 0.41

Lys 59.17 1.68 44.35*** 1.28

1-Mhis 0.89 0.10 10.74*** 0.27

His 7.22 0.34 6.45 0.17

Trp 11.16 0.43 4.94*** 0.24

Arg 12.77 0.41 19.23*** 0.73

Amino acid ratio

Met/Gly 0.46 0.05 0.08*** 0.00

Lys/Arg 4.65 0.19 2.32*** 0.11

Ala alanine, Amm ammonium, Arg arginine, Asn asparagine, Asp

aspartic acid, Citr citrulline, Cys cysteine, Gln glutamine, Glu glu-

tamate, Gly glycine, His histidine, Hypro hydroxyproline, Ile iso-

leucine, Leu leucine, Lys lysine, Met methionine, 1-Mhis 1-

methylhistidine, Orn ornithine, Phe phenylalanine, Pro proline, Ser

serine, Tau taurine, Thr threonine, Trp tryptophane, Tyr tyrosine, Val

valine

* Mean values significantly different from the control group (*

p\ 0.05, ** p\ 0.01, *** p\ 0.001)

Eur J Nutr (2015) 54:193–204 201

123

Page 122: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

members of the ATP-binding cassette (ABC) superfamily

of transport proteins, and responsible for the efflux of bile

acids [38]. A significantly increased Abcb4 mRNA level

was observed in parallel with an increased concentration in

plasma bile acids. We support the previous findings that

ABCB4 may be involved in bile acids excretion from liver

to blood [39]. The increase in bile acids in the 20 % CP

group concomitantly with decreased body weight and high

feed intake could indicate a poorer digestibility of the

protein. Also, some of the undigested peptides might bind

bile acids, increasing bile excretion through feces [40, 41].

Although the CP diets were not deficient in indispensible

amino acids, in further studies protein and bile acids in

feces should be investigated. However, the increase in

plasma bile acids is not caused by liver damage as no

increased plasma activities of ALT and AST were found.

Treatment with bile acids has been shown to decrease

plasma TAG concentrations in humans [42], as well as in

animals, and to prevent TAG accumulation in liver [43], as

observed in our study. Thus, the increase in plasma bile

acids in the 20 % CP group may have been partly involved

in the mechanism of lipid lowering.

The effect of CP on b-oxidation, bile formation, and

lipogenesis may be due to changes in the amino acid

composition compared to casein. Several studies have

suggested that dietary proteins with low ratios of methio-

nine/glycine and lysine/arginine have both cholesterol-

lowering [17, 44, 45] and TAG-lowering effects [46]. Data

suggest that dietary methionine and lysine are hypercho-

lesterolemic, whereas arginine has anti-atherogenic effects

and glycine has hypocholesterolemic effects [46, 47].

Compared to casein supplementation, it was of interest to

note that rats treated with 20 % CP had lower plasma

lysine levels and higher plasma arginine level, resulting in

a lower lysine/arginine ratio. The 20 % CP diet was

characterized by low ratio of lysine/arginine. In the present

study, 20 % CP-treated rats showed an almost sevenfold

lower ratio of plasma methionine/glycine than control rats.

It is also noteworthy that the 20 % CP diet was charac-

terized of a low methionine/glycine ratio. However, the

lower body weight of the 20 % CP rats is probably not

caused by methionine deficiency as studies of methionine-

deprived rats have shown hypercholesterolemia [48] and

fatty liver [49]. Although it is likely that the amino acid

composition of CP has indirect effects on energy metabo-

lism in rats, the mechanisms behind the hypolipidemic

effect of dietary amino acids are unknown and must be

further studied.

In summary, the present study demonstrates that the

treatment of normolipidemic rats with a water-soluble

fraction of CP decreases plasma and liver TAG and cho-

lesterol compared to casein as a protein source. The plasma

TAG-lowering effect seems to be primarily mediated at the

synthesis level by reducing lipogenesis thereby lowering

the flux of fatty acids for TAG biosynthesis, in addition to

an increase in the mitochondrial fatty acid oxidation. The

hypocholesterolemic effect of CP might be related to the

enterohepatic circulation and bile excretion. Whether the

hypolipidemic effect and the bioactivity of CP are related

to low lysine/arginine and methionine/glycine ratios should

Fig. 4 Plasma levels of a free carnitine, b c-butyrobetaine, c acetyl-

carnitine, d propionylcarnitine, e octanoylcarnitine, and f palmitoyl-

carnitine in male Wistar rats fed a control diet with 20 % casein or a

diet with either 6, 14, or 20 % CP for 4 weeks. Values are means with

their standard errors represented by vertical bars (n 6). a,b,cMean

values with unlike letters were significantly different (p\ 0.05; one-

way ANOVA with Tukey’s post hoc test)

202 Eur J Nutr (2015) 54:193–204

123

Page 123: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

be considered. The most striking finding in this study was

the low body weight despite the high feed intake in the

20 % CP-fed rats compared to control. This might be due

to a change in energy balance.

Acknowledgments We thank Kari Williams, Liv Kristine Øysæd,

Randi Sandvik, Kari Helland Mortensen, Svein Kruger and Torunn

Eide for technical assistance, and Eline Milde and the staff at the

Laboratory Animal Facility, University of Bergen, for care of the

animals. This project has been founded by the University of Bergen

through the Clinical Nutrition Program and the company Norilia AS.

Conflict of interest The authors declare that they have no com-

peting interests.

References

1. Ren J, Pulakat L, Whaley-Connell A, Sowers JR (2010) Mito-

chondrial biogenesis in the metabolic syndrome and cardiovas-

cular disease. J Mol Med (Berl) 88:993–1001

2. Subramanian S, Chait A (2012) Hypertriglyceridemia secondary

to obesity and diabetes. Biochim Biophys Acta 1821:819–825

3. Burri L, Thoresen GH, Berge RK (2010) The role of PPARalpha

activation in liver and muscle. PPAR Res. doi:10.1155/2010/

542359

4. Rakhshandehroo M, Knoch B, Muller M, Kersten S (2010) Per-

oxisome Proliferator-activated receptor alpha target genes. PPAR

Research, Vol 2010, Article ID 612089. doi:10.1155/2010/

612089

5. Eberle D, Hegarty B, Bossard P, Ferre P, Foufelle F (2004)

SREBP transcription factors: master regulators of lipid homeo-

stasis. Biochimie 86:839–848

6. Osborne TF (2000) Sterol regulatory element-binding proteins

(SREBPs): key regulators of nutritional homeostasis and insulin

action. J Biol Chem 275:32379–32382

7. Ascencio C, Torres N, Isoard-Acosta F, Gomez-Perez FJ, Her-

nandez-Pando R, Tovar AR (2004) Soy protein affects serum

insulin and hepatic SREBP-1 mRNA and reduces fatty liver in

rats. J Nutr 134:522–529

8. Moriyama T, Kishimoto K, Nagai K, Urade R, Ogawa T, Utsumi

S, Maruyama N, Maebuchi M (2004) Soybean beta-conglycinin

diet suppresses serum triglyceride levels in normal and geneti-

cally obese mice by induction of beta-oxidation, downregulation

of fatty acid synthase, and inhibition of triglyceride absorption.

Biosci Biotechnol Biochem 68:352–359

9. Velasquez MT, Bhathena SJ (2007) Role of dietary soy protein in

obesity. Int J Med Sci 4:72–82

10. Choi JY, Jeon JE, Jang SY, Jeong YJ, Jeon SM, Park HJ, Choi

MS (2011) Differential effects of powdered whole soy milk and

its hydrolysate on antiobesity and antihyperlipidemic response to

high-fat treatment in C57BL/6 N mice. J Agric Food Chem

59:2584–2591

11. Lee YP, Mori TA, Puddey IB, Sipsas S, Ackland TR, Beilin LJ,

Hodgson JM (2009) Effects of lupin kernel flour-enriched bread

on blood pressure: a controlled intervention study. Am J Clin

Nutr 89:766–772

12. Rigamonti E, Parolini C, Marchesi M, Diani E, Brambilla S,

Sirtori CR, Chiesa G (2010) Hypolipidemic effect of dietary pea

proteins: impact on genes regulating hepatic lipid metabolism.

Mol Nutr Food Res 54(Suppl 1):S24–S30

13. Wergedahl H, Liaset B, Gudbrandsen OA, Lied E, Espe M, Muna

Z, Mork S, Berge RK (2004) Fish protein hydrolysate reduces

plasma total cholesterol, increases the proportion of HDL

cholesterol, and lowers acyl-CoA: cholesterol acyltransferase

activity in liver of Zucker rats. J Nutr 134:1320–1327

14. Gudbrandsen OA, Wergedahl H, Liaset B, Espe M, Mork S,

Berge RK (2008) Dietary single cell protein reduces fatty liver in

obese Zucker rats. Br J Nutr 100:776–785

15. Zhang Y, Kouguchi T, Shimizu M, Ohmori T, Takahata Y, Mor-

imatsu F (2010) Chicken collagen hydrolysate protects rats from

hypertension and cardiovascular damage. J Med Food 13:399–405

16. Saiga A, Iwai K, Hayakawa T, Takahata Y, Kitamura S, Ni-

shimura T, Morimatsu F (2008) Angiotensin I-converting

enzyme-inhibitory peptides obtained from chicken collagen

hydrolysate. J Agric Food Chem 56:9586–9591

17. Morita T, Oh-hashi A, Takei K, Ikai M, Kasaoka S, Kiriyama S

(1997) Cholesterol-lowering effects of soybean, potato and rice

proteins depend on their low methionine contents in rats fed a

cholesterol-free purified diet. J Nutr 127:470–477

18. Bjorndal B, Berge C, Ramsvik MS, Svardal A, Bohov P, Skorve

J, Berge RK (2013) A fish protein hydrolysate alters fatty acid

composition in liver and adipose tissue and increases plasma

carnitine levels in a mouse model of chronic inflammation. Lipids

Health Dis 12:143

19. Grimstad T, Bjørndal B, Cacabelos D, Aasprong OG, Omdal R,

Svardal A, Bohov P, Pamplona R, Portero-Otin M, Berge RK,

Hausken T (2013) A salmon peptide diet alleviates experimental

colitis as compared with fish oil. J Nutr Sci 2:1–8

20. Bligh EG, Dyer WJ (1959) A rapid method of total lipid

extraction and purification. Can J Biochem Physiol 37:911–917

21. Bjorndal B, Vik R, Brattelid T, Vigerust NF, Burri L, Bohov P,

Nygard O, Skorve J, Berge RK (2012) Krill powder increases

liver lipid catabolism and reduces glucose mobilization in tumor

necrosis factor-alpha transgenic mice fed a high-fat diet.

Metabolism 61:1461–1472

22. Vernez L, Wenk M, Krahenbuhl S (2004) Determination of

carnitine and acylcarnitines in plasma by high-performance liquid

chromatography/electrospray ionization ion trap tandem mass

spectrometry. Rapid Commun Mass Spectrom 18:1233–1238

23. Vigerust NF, Bohov P, Bjorndal B, Seifert R, Nygard O, Svardal

A, Glintborg D, Berge RK, Gaster M (2012) Free carnitine and

acylcarnitines in obese patients with polycystic ovary syndrome

and effects of pioglitazone treatment. Fertil Steril 98:1620–1626

24. Cohen SA, Strydom DJ (1988) Amino acid analysis utilizing

phenylisothiocyanate derivatives. Anal Biochem 174:1–16

25. Liaset B, Julshamn K, Espe M (2003) Chemical composition and

theoretical nutritional evaluation of the produced fractions from

enzymic hydrolysis of salmon frames with Protamex (TM). Proc

Biochem 38:1747–1759

26. Bremer J (1981) The effect of fasting on the activity of liver

carnitine palmitoyltransferase and its inhibition by malonyl-coa.

Biochim Biophys Acta 665:628–631

27. Madsen L, Rustan AC, Vaagenes H, Berge K, Dyroy E, Berge

RK (1999) Eicosapentaenoic and docosahexaenoic acid affect

mitochondrial and peroxisomal fatty acid oxidation in relation to

substrate preference. Lipids 34:951–963

28. Skorve J, Al-Shurbaji A, Asiedu D, Bjorkhem I, Berglund L,

Berge RK (1993) On the mechanism of the hypolipidemic effect

of sulfur-substituted hexadecanedioic acid (3-thiadicarboxylic

acid) in normolipidemic rats. J Lipid Res 34:1177–1185

29. Vigerust NF, Cacabelos D, Burri L, Berge K, Wergedahl H,

Christensen B, Portero-Otin M, Viste A, Pamplona R, Berge RK,

Bjørndal B (2011) Fish oil and 3-thia fatty acid have additive

effects on lipid metabolism but antagonistic effects on oxidative

damage when fed to rats for 50 weeks. J Nutr Biochem

23:1384–1393

30. Rekawiecki R, Rutkowska J, Kotwica J (2012) Identification of

optimal housekeeping genes for examination of gene expression

in bovine corpus luteum. Reprod Biol 12:362–367

Eur J Nutr (2015) 54:193–204 203

123

Page 124: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

31. Onyeali EU, Onwuchekwa AC, Monaga CC, Monanu MO (2010)

Plasma lipid profile of Wistar albino rats fed palm oil-supple-

mented diets. Int J Biol Chem Sci 4:1163–1169

32. Liaset B, Madsen L, Qin Haob GC, Mellgrenc G, Marschall H-U,

Hallenborg P, Espea M, Frøylanda L, Kristiansen K (2009) Fish

protein hydrolysate elevates plasma bile acids and reduces vis-

ceral adipose tissue mass in rats. Biochim Biophys Acta Mol Cell

Biol Lipid 1791:254–262

33. Trauner M, Halilbasic E (2011) Nuclear receptors as new per-

spective for the management of liver diseases. Gastroenterology

140(1120–1125):e1112

34. Zhang Y, Kouguchi T, Shimizu K, Sato M, Takahata Y, Mor-

imatsu F (2010) Chicken collagen hydrolysate reduces proin-

flammatory cytokine production in C57BL/6.KOR-ApoEshl

mice. J Nutr Sci Vitaminol (Tokyo) 56:208–210

35. Flowers MT (2009) The delta9 fatty acid desaturation index as a

predictor of metabolic disease. Clin Chem 55:2071–2073

36. Hoppel C (2003) The role of carnitine in normal and altered fatty

acid metabolism. Am J Kidney Dis 41:S4–S12

37. Lysiak W, Toth PP, Suelter CH, Bieber LL (1986) Quantitation

of the efflux of acylcarnitines from rat heart, brain, and liver

mitochondria. J Biol Chem 261:13698–13703

38. Dikkers A, Tietge UJ (2010) Biliary cholesterol secretion: more

than a simple ABC. World J Gastroenterol 16:5936–5945

39. Belinsky MG, Bain LJ, Balsara BB, Testa JR, Kruh GD (1998)

Characterization of MOAT-C and MOAT-D, new members of the

MRP/cMOAT subfamily of transporter proteins. J Natl Cancer

Inst 90:1735–1741

40. Gatchalian-Yee M, Arimura Y, Ochiai E, Yamada K, Sugano M

(1997) Soybean protein lowers serum cholesterol levels in ham-

sters: effect of debittered undigested fraction. J Nutr 13:633–639

41. Sugano M, Goto S, Yamada Y, Yoshida K, Hashimoto Y, Matsuo

T, Kimoto M (1988) The hypocholesterolemic action of the

undigested fraction of soybean protein in rats. Atherosclerosis

72:115–122

42. Angelin B, Einarsson K, Leijd B (1978) Effect of chenodeoxy-

cholic acid on serum and biliary lipids in patients with hyper-

lipoproteinaemia. Clin Sci Mol Med 54:451–455

43. Watanabe M, Houten SM, Wang L, Moschetta A, Mangelsdorf

DJ, Heyman RA, Moore DD, Auwerx J (2004) Bile acids lower

triglyceride levels via a pathway involving FXR, SHP, and

SREBP-1c. J Clin Invest 113:1408–1418

44. Gudbrandsen OA, Wergedahl H, Liaset B, Espe M, Berge RK

(2005) Dietary proteins with high isoflavone content or low

methionine-glycine and lysine-arginine ratios are hypocholeste-

rolaemic and lower the plasma homocysteine level in male

Zucker fa/fa rats. Br J Nutr 94:321–330

45. Xu YJ, Arneja AS, Tappia PS, Dhalla NS (2008) The potential

health benefits of taurine in cardiovascular disease. Exp Clin

Cardiol 13:57–65

46. Kritchevsky D, Tepper SA, Czarnecki SK, Klurfeld DM (1982)

Atherogenicity of animal and vegetable protein: influence of the

lysine to arginine ratio. Atherosclerosis 41:429–431

47. Vega-Lopez S, Matthan NR, Ausman LM, Harding SV, Rideout

TC, Ai M, Otokozawa S, Freed A, Kuvin JT, Jones PJ, Schaefer

EJ, Lichtenstein AH (2010) Altering dietary lysine: arginine ratio

has little effect on cardiovascular risk factors and vascular reac-

tivity in moderately hypercholesterolemic adults. Atherosclerosis

210:555–562

48. Moundras C, Remesy C, Levrat MA, Demigne C (1995) Methi-

onine deficiency in rats fed soy protein induces hypercholester-

olemia and potentiates lipoprotein susceptibility to peroxidation.

Metabolism 44:1146–1152

49. Barakat HA, Hamza AH (2012) Glycine alleviates liver injury

induced by deficiency in methionine and or choline in rats. Eur

Rev Med Pharmacol Sci 16:728–736

204 Eur J Nutr (2015) 54:193–204

123

Page 125: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

IV

Page 126: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were
Page 127: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

1

A water-soluble extract of chicken reduced plasma triacylglycerols, but showed 1

no anti-atherosclerotic activity in apoE-/- mice 2

3

Rita Vika*, Trond Brattelidb, Jon Skorvea, Ottar Nygårda.c, Jan E. Nordrehauga,d, Rolf 4

K. Bergea,c and Bodil Bjørndala 5 6 aDepartment of Clinical Science, University of Bergen, 5020 Bergen, Norway 7 bNational Institute of Nutrition and Seafood Research, NIFES, 5817 Bergen, Norway 8 cDepartment of Heart Disease, Haukeland University Hospital, 5021 Bergen, Norway 9 dSection of Cardiology, Stavanger University Hospital, 4068 Stavanger, Norway 10

11

*Corresponding author at: Department of Clinical Science, University of Bergen, 12

5020 Bergen, Norway. Tel.: +4755975846. E-mail address: [email protected] (R. 13

Vik). 14

15

E-mail addresses: 16

TB: [email protected] 17

JS: [email protected] 18

ON: [email protected] 19

JEN: [email protected] 20

RKB: [email protected] 21

BB: [email protected] 22

23

24

25

Page 128: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

2

ABSTRACT 26

27

Background 28

A water-soluble protein extract of chicken (CP) has been reported to modulate plasma 29

lipids and hepatic lipid catabolism. Atherosclerosis is the primary contributing factor 30

for cardiovascular disease and there is evidence that both lipid abnormalities and 31

chronic inflammation have crucial involvement in the initiation and progression of 32

atherosclerosis. The aim of the study was to assess the impact of CP on 33

atherosclerotic development in mice predisposed for developing atherosclerotic 34

lesions. 35

36

Methods 37

24 apoE-/- mice were divided into two groups and fed a high-fat diet. The control 38

group received a 20% casein diet, whereas the intervention group was fed a diet with 39

15% water-soluble extract of chicken protein and 5% casein. Body weight and feed 40

intake was measured regularly, and indirect calorimetry was measured at week 1 and 41

10. After 12 weeks the mice were sacrificed, and blood, liver, heart and aorta were 42

harvested. Plasma and liver lipids and fatty acid composition analyses were carried 43

out, in addition to gene expression in liver and heart. Also, en-face analysis was 44

performed on aorta, and plasma inflammatory markers were determined. 45

46

Results 47

The dietary intervention with CP only resulted in minor reduction of plasma 48

triacylglycerol (TAG) and no change in the plasma cholesterol level compared to 49

control. The TAG lowering was associated with induction of hepatic carnitine 50

acyltransferase 2 activity and gene expression. Mice fed CP also displayed a lower 51

respiratory exchange ratio during the light cycle, indicating a higher degree of fatty 52

acid oxidation in the fasting state. Aorta images displayed no differences in plaque 53

development between mice fed CP or control diet, and gene expression of 54

inflammatory markers in heart was unchanged. Moreover, CP administration did not 55

influence plasma levels of several inflammatory cytokines in apoE-/- mice. 56

57

Conclusion 58

Page 129: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

3

CP displayed a slight potential to increase mitochondrial fatty acid oxidation and 59

reduce plasma TAG. However, CP did not affect plasma cholesterol levels, 60

inflammation status or atherosclerotic development in apoE-/- mice. Based on these 61

results, dietary intervention with CP does not have sufficient capacity to influence 62

atherosclerotic development in apoE-/- mice. 63

64

Key Words: Atherosclerosis, Chicken protein, Triacylglycerol, mitochondrial β-65

oxidation 66

67

68

Page 130: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

4

1. Introduction 69

70

Apolipoprotein E (apoE) is primarily synthesized in liver and most commonly found 71

in circulating chylomicrons and intermediate density lipoprotein (IDL) particles. 72

ApoE is involved in lipid metabolism through the catabolism of triacylglycerol 73

(TAG)-rich particles, in that it binds to receptors belonging to the low-density 74

lipoprotein (LDL) receptor family thus clearing chylomicrons and remnants from 75

plasma [1]. As a ligand for receptor-mediated hepatic clearance of remnant particles, 76

apoE is essential in removing excess TAG and cholesterol from the blood stream. 77

Elevated cholesterol, particularly LDL cholesterol, is strongly associated with 78

atherosclerosis as it contributes to a clustering of LDL and its oxidized form in the 79

intima. ApoE knockout mice (apoE-/-) lack this apolipoprotein leading to an 80

accumulation of VLDL remnants in plasma and the development of spontaneous 81

atherosclerotic lesions [2, 3]. This characteristic gives the opportunity to investigate 82

atherogenesis, without a diet rich in fats and cholesterol. A high-fat, cholesterol-83

containing diet will however, accelerate the process. 84

85

Atherosclerosis, together with hypertension, is the primary cause of coronary artery 86

disease (CAD), stroke peripheral arterial disease and cardiovascular disease (CVD), 87

which is responsible for the majority of morbidity and mortality in the Western world 88

(World Health Organization Global Health Observatory, http://www.who.int/gho/en/). 89

Intervention studies have suggested that consumption of certain protein sources lower 90

cholesterol levels and thus may lower the risk of cardiovascular events. Soy protein is 91

most studied in this regard [4-6], and suggested mechanisms for its cholesterol-92

lowering effect are linked to the presence of non-protein content, i.e isoflavones, the 93

potential role in up-regulation of LDL receptor activity [7], or the low 94

methionine/glycine- and lysine/arginine ratios [8, 9]. Proper folding of apoB, the main 95

protein in LDL, is essential for its function after synthesis. The amino acid 96

composition in optimal diets may influence protein synthesis, hence counteract 97

potential conformational changes in the apoB entity of LDL by facilitating proper 98

folding. This is important as changes in the surface charge of the protein can promote 99

atherosclerosis [10] because loss of apoB characteristic structure will contribute to 100

increased uptake by alternate receptors, like scavenger receptors, accumulation of 101

LDL due to resistance towards proteolysis, and also the promotion of oxidation [11]. 102

Page 131: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

5

Arginine is the principal substrate of nitric oxide (NO) production, and elevated levels 103

of this amino acid has been linked to improved endothelial cell function [12]. In 104

contrast, high levels of methionine is linked to elevated levels of homocysteine, which 105

might cause endothelial damage by decreasing NO-bioavailability [13]. More 106

recently, a lipid-lowering effect of protein from marine origin has been found. Plasma 107

cholesterol-lowering as a result of dietary fish protein is proposed to be due to 108

increased cholesterol excretion through bile. This has been associated with the 109

presence of the organic acid taurine, which conjugates with bile acids forming bile 110

salts, thus increasing their solubility [14, 15]. Recently, chicken protein has showed 111

promising results in lowering hypertension through an angiotensin I-converting 112

enzyme (ACE)-inhibitory effect [16]. 113

We previously found that water soluble chicken protein (CP) affected hepatic lipid 114

and bile acid metabolism in Wistar rats, displaying a decrease in both plasma and 115

hepatic TAG, increased mitochondrial β-oxidation, accompanied by a cholesterol-116

lowering effect linked to increased bile acid formation [17]. Based on these data, we 117

wished to investigate whether a CP-diet could attenuate the progression of 118

atherosclerosis in the apoE knockout mouse model. 119

120

2. Methods 121

122

2.1. Animals and diets 123

124

The animal study was conducted according to Guidelines for the care and use of 125

Experimental Animals, and the Norwegian state Board of Biological Experiments 126

with Living Animals approved the protocol. 24 female apoE knockout mice (apoE-/-) 127

generated in the strain B6, 129P2-Apoetm1Unc/J, four weeks old, were purchased from 128

Jackson Laboratory, Italy. They were randomised and housed in groups of three mice 129

per cage in a closed system under standard laboratory conditions with temperature 22 130

2 C, light/dark cycles of 12/12 h, relative humidity 55 5% and with unrestricted 131

access to chow feed and tap water. The animals were allowed to adjust to this 132

environment for 1 week before cages were distributed into two groups of twelve mice. 133

The control diet contained 20% (w/w) protein from bovine milk casein (Dyets Inc., 134

Bethlehem, PA, USA), and 21% (w/w) fat consisting of 19% lard (Ten Kate Vetten 135

Page 132: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

6

BV, Musselkanaal, Netherlands) and 2% soy oil (Dyets Inc.). The diet also contained 136

0.15% cholesterol (Sigma-Aldrich AS, St. Louis, MO, USA). In the intervention 137

group, 75% of the casein was replaced with chicken protein constituting 15% of the 138

diet (Norilia, Oslo, Norway). The chicken protein was obtained as previously 139

described [17]. The other constituents of the diets were cornstarch, dyetrose, sucrose, 140

fiber, AIN-93G-MX, AIN-93 vitamin mix, L-cysteine, choline bitartrate (all from 141

Dyets Inc.), and tert-Butyl-hydroquinone (Sigma-Aldrich). All mice were fed ad 142

libitum for 12 weeks. 143

144

2.2. Indirect calorimetry 145

After 1 week and 10 weeks of feeding, 5 to 6 animals from each group were 146

transferred to metabolic cages for 3 consecutive days under standard laboratory 147

conditions with free access to feed and water. The cages were monitored by a 148

Comprehensive Laboratory Animal Monitoring system (CLAMS) (Columbus 149

Instruments, Columbus, OH, USA) of individual live-in cages to measure energy 150

expenditure by indirect calorimetry continuously for 30 sec every 17 minutes. 151

CLAMS allow automated, non-invasive data collection monitoring oxygen 152

consumption (VO2) and carbon dioxide production (VCO2). The respiratory exchange 153

ratio (RER) was calculated as the ratio between VCO2 and VO2, while heat was 154

calculated by the formula: 3.815 + 1.232 × RER. The first 7 hours was excluded from 155

the results as acclimatization to the CLAMS cages. 156

157

2.3. Tissue harvesting 158

159

EDTA-blood was collected from the retro-orbital plexus at start and after 11 weeks of 160

the experiment. At sacrifice, mice were anesthetized by inhalation of 2-5% 161

sevoflurane (Abbolt Laboratories Ltd., Berkshire, UK) after four hours of fasting. 162

Blood was collected from the heart into a tube containing 7.5% EDTA, the samples 163

were chilled on ice for about 15 minutes, plasma was separated by centrifugation and 164

stored at -80°C until further analysis. After perfusion with phosphate-buffered saline 165

(PBS), aorta was dissected from the aortic root to the iliac bifurcation, adventitial 166

tissue present was carefully removed, the aorta was put in a tube with 10% formalin 167

overnight, and then transferred to PBS containing tubes. Heart and liver were freeze-168

clamped in liquid nitrogen and stored at -80°C until further analysis. 169

Page 133: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

7

170

2.4. En-face analysis of the aorta 171

172

Aorta dissection took place in a pan filled with PBS. The aorta was split open 173

circumferentially under a magnifier (Carl Zeiss OPMI pico), pinned flat on a pad of 6 174

layers of parafilm covered with black vulcanized insulation tape, placed in 70% 175

ethanol for 10 minutes, and then soaked in Sudan IV (Sigma) for 20 minutes, and 176

finally destained using 80% ethanol for 3 minutes. To avoid particle-interference 177

when photographed, the pinned aorta was rinsed under running water to remove all 178

ethanol. Images were then captured with a digital camera (Canon 5D Mark II). 179

Assessment of plaque area was performed using the software Image J 180

(http://rsb.info.nih.gov/ij/download.html). 181

182

2.5. Plasma lipids and fatty acid composition 183

184

Plasma lipids were measured enzymatically on a Hitachi 917 system (Roche 185

Diagnostics GmbH, Mannheim, Germany) using the triacylglycerol (GPO-PAP) and 186

cholesterol kit (CHOD-PAP) from Roche Diagnostics, and the phospholipids FS kit 187

from DiaSys (Diagnostic Systems GmbH, Holzheim, Germany). Total plasma fatty 188

acid composition was analyzed as previously described [18]. 189

190

2.6. Hepatic enzyme activities 191

192

Fresh liver samples were homogenized as described earlier [19]. Briefly; 100 mg liver 193

were homogenized in 1 mL ice-cold sucrose medium, centrifuged, and the post-194

nuclear fraction removed. Palmitoyl-CoA oxidation was measured in the post-nuclear 195

fraction from liver as acid-soluble products, as described by Bremer et al. [20], with 196

some modifications [21]. The activities of carnitine palmitoyltransferase (CPT)-2, 197

fatty acyl-CoA oxidase (ACOX)-1, glycerol-3-phosphate acyltransferase (GPAT) 198

were measured in the post-nuclear fraction after storage at 80°C as described by 199

Skorve et al. [22], with some modifications [17]. 200

201

2.7. Gene expression analysis 202

203

Page 134: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

8

Total cellular RNA was purified from frozen heart and liver samples, and cDNA was 204

produced as previously described [23]. Real-time PCR was performed with Sarstedt 205

384 well multiply-PCR Plates (Sarstedt Inc., Newton, NC, USA) on the following 206

genes, using probes and primers from Applied Biosystems (Foster City, CA, USA): 207

Acetyl-CoA carboxylase (Acaca, Mm01304277_m1), acyl-coenzyme A oxidase 208

(Acox1, Mm00443579), apolipoprotein B (Apob, Mm01545156), CD36 antigen 209

(Cd36/Fat, Mm00432403), carnitine palmitoyltransferase (Cpt1a, Mm00550438), 210

Cpt2 (Mm00487202), cytochrome P450, family 7, subfamily A, polypeptide 1 211

(Cyp7a1, Mm00484152), glycerol-3-phosphate acyltransferase, mitochondrial (Gpam, 212

Mm00833328), intracellular adhesion molecule (Icam, Mm00516023_m1), low-213

density lipoprotein receptor (Ldlr, Mm00440169), monocyte chemoattractant protein 214

(Mcp1, Mm00441242), nitric oxide synthase 2 (Nos2, Mm00440502_m1), solute 215

carrier family 25, also known as carnitine/acylcarnitine translocase (Slc25a20, 216

Mm00451571_m1), stearoyl-CoA desaturase (Scd1, Mm0077229_m1), vascular cell 217

adhesion molecule 1 (Vcam1, Mm00443281), very low-density lipoprotein receptor 218

(Vldlr, Mm00443281_m1), and Solaris qPCR gene Expression Assays (Thermo 219

Fisher Scientific Inc.,Waltham, MA, USA): fatty acid desaturase 1 (Fads1, AX-220

064722-00-0100), fatty acid/Δ6 desaturase 2 (Fads2, AX-049816-00-0100). 221

Reference genes used in liver: 18s (Kit-FAM-TAMRA (Reference RT-CKFT-18 s)) 222

from Eurogentech, Seraing, Belgium, ribosomal protein, large, P0 (Rplp0, AX-223

061958-00-0100) and TATA-box binding protein (Tbp, AX-041188-00) both from 224

Thermo Fisher Scientific Inc. (Waltham, MA, USA). Reference genes tested for heart: 225

Rplp0, polymerase (RNA) II (DNA directed) polypeptide A, (Polr2a, AX-046005-00) 226

and hypoxanthine guanine phosphoribosyltransferase 1 (Hprt1, AX-045271-00) also 227

from Thermo Fisher Scientific Inc. Normfinder (moma.dk/normfinder-software) was 228

used to assess the optimal reference genes, and data normalized to 18s, Rplp0 and Tbp 229

were used for liver, and Rplp0 for heart. 230

231

2.8. Plasma inflammatory markers 232

233

Plasma concentrations of interleukin (IL)-1 IL-1β, IL-2, IL-6, IL-10, IL-17, 234

granulocyte colony-stimulating factor (G-CFS), granulocyte macrophage colony-235

stimulating factor (GM-CFS), interferon gamma (INF- ), monocyte chemotactic 236

Page 135: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

9

protein 1 (MCP-1/CCL2), chemokine (C-C motif) ligand 5 (Rantes/ CCL5), and 237

tumour necrosis factor alpha (TNF- ) were determined using a custom-made 238

multiplex MILLIPLEX MAP kit (Millipore Corp., St. Charles, IL, USA). The 239

antibody-conjugated beads were allowed to react with the sample and a secondary 240

antibody in a 96-well plate to form a capture sandwich immunoassay. Finally, the 241

assay solution was read by the Bio-Plex array reader (Bio-Rad, Hercules, CA, USA) 242

and determined with the Bio-Plex Manager Software 4.1. 243

244

2.9. Statistical analysis 245

246

Data sets were analyzed using Prism Software (Graph-Pad Software, San Diego, CA) 247

to create figures and to evaluate statistical significance. The results are shown as 248

means of 8-12 animals pr. group with their standard deviations, unless otherwise 249

stated. Shapiro-Wilk test was used to estimate normal distribution and unpaired t-test 250

was used to determining statistical significance between the two groups. P-values < 251

0.05 were considered significant. 252

253

3. Results 254

255

3.1. Increased feed intake and energy expenditure in CP treated mice 256

257

After 12 weeks of CP-feeding we found no change in body weight, but a significant 258

increased feed intake compared to control (Table 1). The CP group displayed an 259

increase in liver weight, but the liver index was unchanged (Table 1). The metabolic 260

rate of the animals was measured by indirect calorimetry after 1 week and 10 weeks 261

on high-fat diets. No change was seen in metabolic cage-feed intake, activity or 262

calculated RER in this period (data not shown). Energy expenditure calculated as heat 263

production was reduced in week 10 compared to week 1 in both feeding groups (Fig. 264

1A). During the first stay in CLAMS the feeding groups had a similar weight gain 265

(data not shown). However, in week 10, the control group gained weight (mean ± SD; 266

0.89 ± 0.8 g), while the CP group lost weight (mean ± SD; -0.4 ± 0.6 g), despite a 267

similar feed intake (Fig. 1B). Activity was comparable in CP-fed and control-fed mice 268

both in the dark (night) and light (day) phase (Fig. 1C). However, during the light 269

Page 136: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

10

phase, the CP-fed group demonstrated lower RER than control after 10 weeks of 270

dietary intervention (Figure 1D). Figure 1E and F show a graphic representation of 271

heat production and RER during the day/night cycle, respectively. 272

273

3.2. Minor influence of CP on plasma lipids 274

275

Non-fasted plasma TAG levels decreased over time in both the intervention and 276

control group, although more so in CP-fed mice (Fig. 2A). Thus, the ΔTAG 277

(difference between week 11 and baseline) value was lower in the CP group than the 278

control group (Fig. 2B). The end-point measurements of TAG at week 12 showed a 279

minor lower level of plasma TAG in CP-treated mice compared to controls (Fig. 2C). 280

Plasma total cholesterol levels increased in both groups over time (Fig. 2D, E), with 281

no difference between the two groups at the end of the treatment period (Fig. 2F). The 282

HDL cholesterol, LDL cholesterol, phospholipids, NEFAs and cholesteryl esters all 283

increased in both groups during the treatment period, and there was no difference 284

between the two groups at termination (data not shown). The CP intervention did not 285

change the plasma fatty acid composition after 12 weeks of diet (data not shown). 286

287

3.3 Hepatic enzyme activity and gene expression after CP intervention 288 289 The CP-diet did not affect palmitoyl-CoA catabolism in hepatic homogenates, and the 290

activity with and without the inhibitory component malonyl-CoA was not influenced 291

(Fig. 3A). However, the CPT2 activity and mRNA level of Cpt2 was tended to 292

increased, which could contribute to increased mitochondrial fatty acid oxidation (Fig. 293

3B and Table 2) as CPT2 participate in fatty acid transport into the matrix, essential 294

for their oxidation. Peroxisomal fatty acid oxidation was not affected, as ACOX1 295

activity and gene expression was unchanged (Fig. 3C and Table 2). CP did not seem 296

to have an effect on fatty acid synthesis as FAS activity and the mRNA level of Fasn 297

as well as the rate-limiting enzyme in lipogenesis, Acaca, displayed comparable levels 298

between CP-treated mice and controls (Fig. 3D, Table 2, respectively). The gene 299

expression of Slc25a20, mediating the transport of acylcarnitines into the 300

mitochondrial matrix, was not modified in CP-treated mice (Table 2). In addition, no 301

change was observed in the expression Scd1, Fads1 and Gpam, involved in 302

desaturation and glycerolipid synthesis, respectively (Table 2). Fads2 was 303

Page 137: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

11

significantly down regulated, while Cd36, probably involved in fatty acid import, was 304

unchanged. No influence on cholesterol import was observed as Apob, Ldlr and Vldlr 305

were unchanged (Table 2). 306

307

3.4. Atherosclerotic lesion formations and gene expression in the heart 308

309

En-face analysis was used to reveal plaque development in the aortic arch (Fig. 4A). 310

As shown in Figure 4B, we detected a comparable amount of plaque in the aorta of 311

CP-fed mice compared to controls. Expression in the heart of genes related to 312

inflammation supported these findings as no change was found in mRNA levels of 313

Icam, Vcam1, Nos2 and Mcp1 (Fig. 4C-F, respectively). 314

315

3.5. Effect of CP on systemic inflammation 316

317

To assess systemic inflammation, plasma levels of several inflammatory components 318

were determined after 12 weeks on high-fat diets. CP administration did not influence 319

the cytokines and chemokines measured, except a small, but significant increase in 320

IFN- levels in CP-fed mice compared to controls. 321

322

4. Discussion 323

324

Atherosclerosis is a multifactorial complex CVD with a bidirectional interaction 325

between lipids and inflammation as a major feature. Risk factors such as dyslipidemia 326

are involved in the progression of atherosclerosis. Specific amino acid content from 327

certain protein sources have emerged as both cholesterol- and TAG-lowering 328

constituents of foods [8, 9, 24]. In the present study, we only found a small reduction 329

in plasma TAG levels of apoE-/- mice after CP-feeding and all classes of cholesterol 330

increased over time in both control and CP-treated apoE-/-- mice in response to high-331

fat feeding. The minimal TAG-lowering effect in the CP group seemed to be related 332

to a slight increase in mitochondrial fatty acid oxidation, as both CPT2 activity and 333

gene expression displayed a marginal increase. These results are in line with a 334

reduced RER during the fasting state observed in CP-fed mice. However, these 335

findings are not nearly as prominent as in a previous experiment with CP in Wistar 336

Page 138: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

12

rats, where a clear reduction in plasma and liver TAG were detected, linked to both 337

increased fatty acid oxidation and reduced lipogenesis [17]. In the present study in 338

apoE-/- mice, lipogenesis seemed unaffected, as FAS activity and gene expression of 339

lipogenic genes were unchanged. 340

341

The atherosclerotic development in aortic arch in apoE-/- mice was not influenced by 342

CP administration, further supporting that the vague reduction in plasma TAG was 343

insufficient to prevent plaque development in this model. As implied previously [21], 344

differences in atherosclerotic development in response to protein diets could be 345

independent of plasma lipids and fatty acids, and more related to anti-inflammatory 346

processes. Studies have demonstrated anti-inflammatory potential of marine bioactive 347

peptides in apoE-/- mice as well as cell culture [21, 25]. Here, plasma inflammatory 348

markers were unchanged, as were gene expression of Vcam1, Icam, Nos2 and Mcp1 in 349

the heart. Another study using chicken collagen hydrolysate fed C57BL/6.KOR-350

ApoEshl mice, revealed unaltered TAG levels between the intervention group and 351

control, accompanied by no differences in aortic plaque area [26]. The failure to 352

reduce plaque area, plasma cholesterol level and lipogenesis, combined with the 353

physiologically irrelevant reduction in plasma TAG levels in the present study, 354

indicate that CP is not able to counteract the negative effect of disrupted lipoprotein 355

metabolism in apoE-/- mice. 356

357

An interesting observation was the higher feed intake in CP-fed mice compared to 358

controls, without a corresponding increase in weight gain. This could be connected to 359

the observed reduction in RER, when monitoring the mice in metabolic cages 360

(CLAMS), implying that fat was the preferred energy source. We detected no 361

differences in fat deposit weights between the CP-fed group and controls (data not 362

shown), suggesting that the increased energy intake was compensated for with 363

increased fat combustion, thus avoiding weight gain in the CP-fed mice. Noteworthy, 364

the apoE-/- model is not suitable for studying body weight due to the lack of a gene 365

important for lipoprotein metabolism; therefore these allegations are strictly 366

speculations. Heat production as well as activity level displayed comparable levels 367

between the two groups both the first period (1 week) and second period (10 week) of 368

CLAMS, meaning that energy consumption was approximately similar. The lower 369

heat production observed in both groups in the second period of CLAMS is probably 370

Page 139: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

13

related to adaptation to the high-fat diet. As heat production constitutes energy 371

expenditure, and levels of activity and heat production were similar between groups, 372

the lower RER value in the CP-fed mice suggest a higher energy contribution from fat 373

in this group. 374

375

4. Conclusion 376

377

After feeding apoE-/- mice a high-fat casein control diet or a high-fat CP diet for 12 378

weeks, we detected no differences regarding plasma inflammatory markers or 379

atherosclerotic lesions area. Although slightly lower plasma TAG concentration was 380

detected, linked to a possible increase in hepatic mitochondrial fatty acid oxidation 381

and energy expenditure from fat, the reduction was insignificant to counteract 382

atherosclerotic development. Previously, CP displayed promising results in Wistar 383

rats in reducing body weight, plasma TAG and cholesterol levels. However, CP did 384

not seem to influence lipid metabolism in apoE-/- mice compared to normolipidemic 385

rats, and was not able to ameliorate atherosclerosis. 386

387

ACKNOWLEDGMENTS 388 389 The authors are grateful to Liv Kristine Øysæd, Randi Sandvik, Kari Williams, and 390

Svein Krüger for technical assistance, and Kari Helland Mortensen, Eline Milde 391

Nævdal and the staff at the animal facility Vivariet, UiB, for assisting with care of the 392

animals. 393

394

FUNDING 395

396

The research was supported by The Board of Nutrition Programmes – University of 397

Bergen, Norilia AS (Oslo, Norway) and NFR project 212984. 398

399

DISCLOSURE STATEMENT 400

Norilia AS provided the chicken protein and partly funded the animal experiment, but 401

had no involvement in the study design, interpretation of the data, writing of the 402

manuscript, or decision to publish. 403

Page 140: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

14

404

AUTHOR CONTRIBUTION 405

BB, RKB, ON, JEN, and RV designed the study. RV and BB conducted the animal 406

study and TB performed the aorta dissection. RV and BB performed the plaque 407

staining, and RV analyzed the images. RV, JS, and BB analyzed the CLAMS data. 408

RV performed the qPCR. RV and BB analyzed and interpreted the data. RV wrote the 409

first draft, and BB and RKB participated in the finalization of the manuscript. All 410

authors revised the manuscript critically and approved the final version of the 411

manuscript. 412

413

REFERENCES 414

415

[1] Eichner JE, Dunn ST, Perveen G, Thompson DM, Stewart KE, Stroehla BC. 416

Apolipoprotein E polymorphism and cardiovascular disease: a HuGE 417

review. Am J Epidemiol. 2002;155:487-95. 418

[2] Ishibashi S, Herz J, Maeda N, Goldstein JL, Brown MS. The two-receptor model 419

of lipoprotein clearance: tests of the hypothesis in "knockout" mice 420

lacking the low density lipoprotein receptor, apolipoprotein E, or both 421

proteins. Proc Natl Acad Sci U S A. 1994;91:4431-5. 422

[3] Zhang SH, Reddick RL, Piedrahita JA, Maeda N. Spontaneous 423

hypercholesterolemia and arterial lesions in mice lacking apolipoprotein 424

E. Science. 1992;258:468-71. 425

[4] Erdman JW, Jr. AHA Science Advisory: Soy protein and cardiovascular 426

disease: A statement for healthcare professionals from the Nutrition 427

Committee of the AHA. Circulation. 2000;102:2555-9. 428

Page 141: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

15

[5] Rebholz CM, Reynolds K, Wofford MR, Chen J, Kelly TN, Mei H, et al. Effect of 429

soybean protein on novel cardiovascular disease risk factors: a 430

randomized controlled trial. Eur J Clin Nutr. 2013;67:58-63. 431

[6] Sirtori CR, Lovati MR. Soy Proteins and Cardiovascular Disease. Current 432

Atherosceloris Reports 2001;3:47–53. 433

[7] Kirk EA, Sutherland P, Wang SA, Chait A, LeBoeuf RC. Dietary isoflavones 434

reduce plasma cholesterol and atherosclerosis in C57BL/6 mice but not 435

LDL receptor-deficient mice. J Nutr. 1998;128:954-9. 436

[8] Kritchevsky D, Tepper SA, Czarnecki SK, Klurfeld DM. Atherogenicity of 437

animal and vegetable protein. Influence of the lysine to arginine ratio. 438

Atherosclerosis. 1982;41:429-31. 439

[9] Morita T, Oh-hashi A, Takei K, Ikai M, Kasaoka S, Kiriyama S. Cholesterol-440

lowering effects of soybean, potato and rice proteins depend on their low 441

methionine contents in rats fed a cholesterol-free purified diet. J Nutr. 442

1997;127:470-7. 443

[10] Ursini F, Davies KJA, Maiorino M, Parasassi T, Sevanian A. Atherosclerosis: 444

another protein misfolding disease? TRENDS in Molecular Medicine 445

2002;8:370-374. 446

[11] Davies KJ, Delsignore ME. Protein damage and degradation by oxygen 447

radicals. III. Modification of secondary and tertiary structure. J Biol Chem. 448

1987;262:9908-13. 449

[12] Siasos G, Tousoulis D, Antoniades C, Stefanadi E, Stefanadis C. L-Arginine, 450

the substrate for NO synthesis: an alternative treatment for premature 451

atherosclerosis? Int J Cardiol. 2007;116:300-8. 452

Page 142: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

16

[13] Zulli A, Hare DL, Buxton BF, Black MJ. High dietary methionine plus 453

cholesterol exacerbates atherosclerosis formation in the left main 454

coronary artery of rabbits. Atherosclerosis. 2004;176:83-9. 455

[14] Balkan J, Oztezcan S, Hatipoglu A, Cevikbas U, Aykac-Toker G, Uysal M. Effect 456

of a taurine treatment on the regression of existing atherosclerotic lesions 457

in rabbits fed on a high-cholesterol diet. Biosci Biotechnol Biochem. 458

2004;68:1035-9. 459

[15] Elvevoll EO, Eilertsen KE, Brox J, Dragnes BT, Falkenberg P, Olsen JO, et al. 460

Seafood diets: hypolipidemic and antiatherogenic effects of taurine and n-461

3 fatty acids. Atherosclerosis. 2008;200:396-402. 462

[16] Saiga A, Iwai K, Hayakawa T, Takahata Y, Kitamura S, Nishimura T, et al. 463

Angiotensin I-converting enzyme-inhibitory peptides obtained from 464

chicken collagen hydrolysate. J Agric Food Chem. 2008;56:9586-91. 465

[17] Vik R, Bjorndal B, Bohov P, Brattelid T, Svardal A, Nygard OK, et al. 466

Hypolipidemic effect of dietary water-soluble protein extract from 467

chicken: impact on genes regulating hepatic lipid and bile acid metabolism. 468

Eur J Nutr. 2014; DOI: 10.1007/s00394-014-0700-5. 469

[18] Bjorndal B, Vik R, Brattelid T, Vigerust NF, Burri L, Bohov P, et al. Krill 470

powder increases liver lipid catabolism and reduces glucose mobilization 471

in tumor necrosis factor-alpha transgenic mice fed a high-fat diet. 472

Metabolism. 2012;61:1461-72. 473

[19] Berge RK, Flatmark T, Osmundsen H. Enhancement of long-chain acyl-CoA 474

hydrolase activity in peroxisomes and mitochondria of rat liver by 475

peroxisomal proliferators. Eur J Biochem. 1984;141:637-44. 476

Page 143: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

17

[20] Bremer J. The effect of fasting on the activity of liver carnitine 477

palmitoyltransferase and its inhibition by malonyl-coa. Biochim Biophys 478

Acta 665. 1981628-631. 479

[21] Parolini C, Vik R, Busnelli M, Bjorndal B, Holm S, Brattelid T, et al. A salmon 480

protein hydrolysate exerts lipid-independent anti-atherosclerotic activity 481

in ApoE-deficient mice. PLoS One. 2014;9:e97598. 482

[22] Skorve J, al-Shurbaji A, Asiedu D, Bjorkhem I, Berglund L, Berge RK. On the 483

mechanism of the hypolipidemic effect of sulfur-substituted 484

hexadecanedioic acid (3-thiadicarboxylic acid) in normolipidemic rats. J 485

Lipid Res. 1993;34:1177-85. 486

[23] Vigerust NF, Cacabelos D, Burri L, Berge K, Wergedahl H, Christensen B, et 487

al. Fish oil and 3-thia fatty acid have additive effects on lipid metabolism 488

but antagonistic effects on oxidative damage when fed to rats for 50 489

weeks. J Nutr Biochem. 2012;23:1384-93. 490

[24] Xu YJ, Arneja AS, Tappia PS, Dhalla NS. The potential health benefits of 491

taurine in cardiovascular disease. Exp Clin Cardiol. 2008;13:57-65. 492

[25] Ahn CB, Cho YS, Je JY. Purification and anti-inflammatory action of tripeptide 493

from salmon pectoral fin byproduct protein hydrolysate. Food Chem. 494

2015;168:151-6. 495

[26] Zhang Y, Kouguchi T, Shimizu K, Sato M, Takahata Y, Morimatsu F. Chicken 496

collagen hydrolysate reduces proinflammatory cytokine production in 497

C57BL/6.KOR-ApoEshl mice. J Nutr Sci Vitaminol (Tokyo). 2010;56:208-498

10. 499

500

Page 144: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

18

501

502

503

Page 145: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

19

504

505

506

Table 1 Body weight and feed intake in apoE-/- mice on a high-fat diet casein diet

(Control), or a high-fat diet supplemented with 15% water-soluble extract of chicken

protein (CP) for 12 weeks

Control CP

Start weight (g) 16.69 3.20 17.33 1.85

End Weight (g) 24.40 2.11 25.24 0.78

Weight gain (g) 6.22 1.70 8.11 1.34

Liver Weight (g) 1.08 0.12 1.20 0.14*

Liver index % of body weight 4.42 0.46 4.77 0.46

Total feed intake (g/mouse) 188 5.22 225 22.9*

Feed efficiency (weight gain/feed intake) 0.03 0.033 0.04 0.058

Data are shown as mean values SD (n = 12). Statistical differences from control wer

determined by unpaired t-test. *P < 0.05.

Page 146: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

20

Table 2 Hepatic gene expression in female apoE-/- mice fed a high-fat casein diet (Control), or a

high-fat diet with 15% water-soluble extract of chicken protein (CP) diet for 12 weeks

Diet groups

Lipid import and β-oxidation

Control CP

Gene Function Mean SD Mean SD P-value

Acox1 β-oxidation 1.00 0.160 1.10 0.186 0.235

Cpt1a β-oxidation 1.00 0.441 1.25 0.142 0.100

Cpt2 β-oxidation 1.00 0.134 1.28 0.243 0.011*

Cd36 Fatty acid import 1.00 0.269 1.12 0.380 0.478

Slc25a20 Fatty acid import 1.00 0.206 1.05 0.172 0.599

Lipogenesis and cholesterol synthesis and

import

Control CP

Gene Function Mean SD Mean SD P-value

Acaca Fatty acid synthesis 1.00 0.208 1.20 0.323 0.151

Apob Cholesterol import 1.00 0.763 0.69 0.105 0.224

Fads1 Δ5 desaturase 1.00 0.204 0.97 0.212 0.767

Fads2 Δ6 desaturase 1.00 0.177 0.75 0.234 0.023*

Gpam Glycerolipid synthesis 1.00 0.136 1.00 0.234 0.876

Ldlr Cholesterol import 1.00 0.208 0.92 0.184 0.379

Scd1 Δ9 desaturase 1.00 0.380 0.95 0.220 0.750

Vldlr Cholesterol import 1.00 0.390 0.92 0.167 0.709

Page 147: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

21

507

508

Bile synthesis

Control CP

Gene Function Mean SD Mean SD P-value

Cyp7a1 Bile acid synthesis 1.00 0.581 0.71 0.487 0.260

Data are shown as mean values SD (n = 8-10). Statistical differences from control were

determined by unpaired t-test, *P < 0.05.

Abbr.: Acaca, Acetyl-CoA carboxylase; Acox1, acyl-CoA oxidase; Apob, apolipoprotein B;

Cd36/Fat, CD36 antigen; Cpt1a, carnitine palmitoyltransferase 1a; Cpt2, carnitine

palmitoyltransferase; Cyp7a1, cytochrome P450, family 7, subfamily A, polypeptide 1; Fads1,

fatty acid desaturase 1; Fads2, fatty acid desaturase 2; Gpam, glycerol-3-phosphate

acyltransferase, mitochondrial; Icam, intracellular adhesion molecule; Ldlr, low-density

lipoprotein receptor; Mcp1, monocyte chemoattractant protein; Nos2, nitric oxide synthase 2;

Slc25a20, solute carrier family 25; Scd1, stearoyl-CoA desaturase; Vcam1, vascular cell

adhesion molecule 1, Vldlr, very low-density lipoprotein receptor.

Page 148: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

22

509

Table 3 Plasma cytokine levels in apoE-/- mice fed a high-fat casein diet (Control), or

a high-fat diet containing 15% water-soluble extract of chicken protein (CP) for 12

weeksa

Cytokine Control-diet CP-diet P-value

IL-1 43.6 ± 14.1 53.8 ± 13.0 0.182

IL-1 707.1 ± 152.1 589.0 ± 82.2 0.077

IL-2 56.5 ± 16.3 64.9 ± 30.3 0.524

IL-6 26.0 ± 9.0 28.2 ± 6.6 0.597

IL-10 316.2 ± 67.5 363.4 ± 62.2 0.201

IL-17 3732.7 ± 1742.6 5178.7 ± 885.1 0.059

G-CFS 267.0 ± 78.8 469.8 ± 547.0 0.351

GM-CFS 3589.6 ± 866.4 3472.8 ± 251.3 0.720

INF- 108.1 ± 28.0 138.3 ± 23.8 0.042*

MCP-1 780.2 ± 93.6 686.2 ± 177.5 0.866

Rantes 55.1 ± 18.3 57.7 ± 26.8 0.829

TNF- 1901.8 ± 149.5 2106.9 ± 374.7 0.200

Page 149: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

23

apmol/mL

Abbreviations: G-CFS, granulocyte colony-stimulating factor; GM-CFS, granulocyte

macrophage colony-stimulating factor; IL, interleukin; INF- interferon gamma,

MCP-1, monocyte chemotactic protein 1; Rantes, chemokine (C-C motif) ligand 5;

TNF- , tumour necrosis factor alpha.

Data are shown as mean ± SD (n = 6-9). Student´s t-test was used to determine

significantly different values (P < 0.05).

510

Page 150: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

24

FIGURE LEGENDS 511

512

Figure 1: Metabolic parameters in apoE-/- mice fed a high-fat casein diet (control), or 513

a high-fat diet with 15% water-soluble extract of CP during a 40 h stay in CLAMS. 514

A) Energy expenditure (heat) after 1 and 10 weeks of dietary intervention. B) Feed 515

intake, C) Activity, and D), Respiratory exchange ratio (RER) during the day/night 516

cycle after 10 weeks of dietary intervention. E) Plot of continuous energy expenditure 517

(heat) and F) RER at week 10. Data are shown as mean ± SD (n = 5-6). Unpaired t-518

test was used to detect statistical significance (***P < 0.001). 519

520

Figure 2: Plasma lipid levels in apoE-/- mice fed a high-fat casein diet (control), or a 521

high-fat diet with 15% water-soluble extract of chicken protein (CP). A) 522

Triacylglycerols (TAG; n = 2, pooled samples) at week 1, 5 and 11, B) ΔTAG (n = 2), 523

week 1-11, C) TAG end-point, week 12 (n = 7-9), D) Cholesterol (n = 2) at week 1, 5 524

and 11, E) ΔCholesterol (n = 2), week 1-11, and F) Cholesterol end-point, week 12 (n 525

= 7-9). Data are shown as mean ± SD. Unpaired t-test was used to detect statistical 526

significance (**P < 0.01). 527

528

Figure 3: Hepatic mitochondrial β-oxidation and enzyme activity in apoE-/- mice fed 529

a high-fat casein diet (control), or a high-fat diet with 15% water-soluble extract of 530

chicken protein (CP). A) Palmitoyl-CoA catabolism in the presence and absence of 531

malonyl-CoA, B) Carnitine palmitoyltransferase 2 (CPT2) activity, C) Acyl-CoA 532

oxidase activity (ACOX1) and D) Fatty acid synthase (FAS) activity. Data are shown 533

as mean ± SD (n = 6-12). Unpaired t-test was used to detect statistical significance 534

(**P < 0.01). 535

Page 151: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

25

536

Figure 4: Atherosclerotic development and mRNA levels in the heart of apoE-/- mice 537

fed a high-fat casein diet (Control), or a high-fat diet with 15% chicken protein (CP). 538

A) Representative images of aortic arch from a control and a CP mouse, and B) 539

calculated plaque area. mRNA levels of: C) intracellular adhesion molecule (Icam), 540

D) vascular cell adhesion molecule 1 (Vcam1), E) inducible nitric oxide synthase 541

(Nos2), and F) monocyte chemotactic protein 1 (Mcp1). Data are shown as mean ± 542

SD (B: n = 12, C – F: n = 8). Unpaired t-test was used to detect statistical 543

significance. 544

545

Page 152: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

26

546

Figure 1 547

548 549

550

551

552

553

554

RER week 10

Time

VCO

2/VO

2

19:0

0

00:0

0

07:0

0

12:0

0

19:0

0

00:0

0

07:0

0

19:0

0

00:0

0

07:0

0

0.7

0.8

0.9

1.0 ControlCP

Night Day

0.6

0.7

0.8

0.9

1.0

VCO

2/VO

2

ControlCP

***

RER week 10

0.0

Heat production

Week 1 Week 100

5

10

15

20

25

Kcal

/hr/k

g

Control CP

*** ***

Activity week 10

Night Day0

1×104

2×104

3×104

4×104

Cou

nts

ControlCP

Feed intake week 10

g/m

ouse

/12

hour

s

Night Day0.0

0.5

1.0

1.5

2.0

2.5

ControlCP

A B

C

F

E

D

Heat week 10

19:0

0

00:0

0

07:0

0

12:0

0

19:0

0

00:0

0

07:0

0

12:0

0

19:0

0

00:0

0

07:0

0

0.25

0.30

0.35

0.40

0.45

0.50

0.55

Time

Kcal

/hr

ControlCP

Page 153: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

27

Figure 2 555

556

557 558 Figure 3 559 560

561 562 563

TAG

0 5 110

1

2

3

mm

ol/L

Week

ControlCP

Cholesterol

0 5 110

10

20

30

40

mm

ol/L

Week

ControlCP

Control CP0.0

0.2

0.4

0.6

0.8

1.0

mm

ol/L

TAG at sacrifice

**

Control CP0

10

20

30

mm

ol/L

Cholesterol at sacrifice

Control CP-1.5

-1.0

-0.5

0.0

mm

ol/L

TAG

Cholesterol

Control CP0

5

10

15

20

mm

ol/L

A B C

D E F

Page 154: %LRDFWLYH SURWHLQV DQG SHSWLGHV LQIOXHQFH OLSLG …chainweb.synology.me/nutrition/RitaVik_Dissertation_PhD.pdf · markers were reduced, whereas plasma TAG and cholesterol levels were

28

Figure 4564

565 566


Recommended